Neal Halfon · Christopher B. Forrest
Richard M. Lerner · Elaine M. Faustman Editors
Handbook of
Life Course
Health
Development
Handbook of Life Course
Health Development
Neal Halfon • Christopher B. Forrest
Richard M. Lerner • Elaine M. Faustman
Editors
Handbook of
Life Course Health
Development
Editors
Neal Halfon
Department of Pediatrics
David Geffen School of Medicine
UCLA, Los Angeles, CA, USA
Department of Health Policy
and Management
Fielding School of Public Health
UCLA, Los Angeles, CA, USA
Department of Public Policy
Luskin School of Public Affairs
UCLA, Los Angeles, CA, USA
Center for Healthier Children
Families, and Communities
UCLA, Los Angeles, CA, USA
Christopher B. Forrest
Applied Clinical Research Center
Children’s Hospital of Philadelphia
Philadelphia, PA, USA
Elaine M. Faustman
Institute for Risk Analysis and Risk
Communication
Department of Environmental and
Occupational Health Sciences
School of Public Health
University of Washington
Seattle, Washington, USA
Richard M. Lerner
Tufts University
Medford, MA, USA
ISBN 978-3-319-47141-9
ISBN 978-3-319-47143-3
DOI 10.1007/978-3-319-47143-3
(eBook)
Library of Congress Control Number: 2017950672
© The Editor(s) (if applicable) and The Author(s) 2018, corrected publication 2018
Open Access This book is licensed under the terms of the Creative Commons Attribution 4.0
International License (http://creativecommons.org/licenses/by/4.0/), which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as long as you give
appropriate credit to the original author(s) and the source, provide a link to the Creative Commons
license and indicate if changes were made.
The images or other third party material in this book are included in the book’s Creative
Commons license, unless indicated otherwise in a credit line to the material. If material is not
included in the book’s Creative Commons license and your intended use is not permitted by
statutory regulation or exceeds the permitted use, you will need to obtain permission directly
from the copyright holder.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this
publication does not imply, even in the absence of a specific statement, that such names are
exempt from the relevant protective laws and regulations and therefore free for general use.
The publisher, the authors and the editors are safe to assume that the advice and information in
this book are believed to be true and accurate at the date of publication. Neither the publisher nor
the authors or the editors give a warranty, express or implied, with respect to the material
contained herein or for any errors or omissions that may have been made. The publisher remains
neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Printed on acid-free paper
This Springer imprint is published by Springer Nature
The registered company is Springer International Publishing AG
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland
Preface
Maternal and Child Health, Life Course Health
Development, and the Life Course Research Network
Prior to 1900, the health of mothers and children was considered a domestic
concern. Childbirth was often supervised by untrained birth attendants such
as family members; basic care of sick children was rudimentary and undeveloped, with the unfortunate but all-too-real expectation that some children
would not survive into adulthood (Rosenfeld and Min 2009). With the advent
of scientific medicine in the nineteenth century, discoveries in bacteriology,
and other sanitary reforms, childbirth came under greater medical scrutiny,
and pediatric hospitals were established to care for ailing children. A greater
focus on maternal nutrition, the spread of scientifically supported birthing
practices, and other newly minted public health practices – along with
improved social and living conditions – led to dramatic decreases in infant
mortality rates and to improved child survival. In 1912, the Children’s Bureau
was established in the United States as a federal agency with responsibility
for assuring the health of mothers and children. In 1935, Title V of the Social
Security Act established the Maternal and Child Health Bureau (MCHB),
which today administers a broad range of programs to address the health
needs of the nation’s maternal and child health (MCH) population.
For most of the twentieth century, MCH programs and policies continued
to focus on two basic areas: (1) promoting healthy births by preventing maternal and infant mortality and, more recently, (2) preventing premature births
and providing medical care for children with long-term medical and developmental disorders. Success was marked by decreasing rates of maternal and
infant mortality but was challenged by persistent disparities in outcomes,
especially differences in infant mortality between White and AfricanAmerican children. Similarly, while great strides were made in reducing child
deaths due to infectious disease and improving the effectiveness, availability,
and quality of medical interventions for a range of childhood conditions from
hemophilia to complex congenital heart diseases, the number of children
reported as being disabled due to a chronic health problem rose dramatically
from 2% in 1960 to over 8% in 2011 (Halfon et al. 2012).
In the late 1980s, a new and rapidly converging set of research findings from
the life course health sciences began to recast the importance of early life on
v
vi
lifelong health (Ben-Shlomo and Kuh 2002; Halfon and Hochstein 2002).
Research that was particularly relevant to the MCH field revealed how:
• Preconception health and perinatal risk can impact birth outcomes and
have a sustained and long-term impact on child and adult health several
decades later.
• Susceptibility and sensitivity of the developing brain to adversity, as well
as to supportive and caring relationships, can be measured not only in
brain morphology but also using functional measures of cognitive and
emotional performance, including school readiness, academic performance, and long-term mental health.
• Risky and chaotic family environments, and toxic and unpredictable social
environments, are transduced into a child’s biology, manifesting as disease
and causing changes in immune, inflammatory, and metabolic function
that can be linked with childhood health conditions like obesity and
ADHD and adult conditions like diabetes, hypertension, and heart disease,
to name a few.
These and other research findings also suggested new explanatory mechanisms for seemingly intractable problems such as the persistent racial and
ethnic gaps in infant mortality. The dominant biomedical approach to treating
infant mortality focused on prenatal care and the prevention of pathological
signs and symptoms (e.g., eclampsia), but what the findings from the life
course health sciences began to suggest is that women’s preconception reproductive capacity – including neuroendocrine response patterns, vascular
health, and stress reactivity – could condition their response to pregnancy, the
timing of parturition, and the likelihood of prematurity (Lu and Halfon 2003).
This work suggested that in addition to improving technical interventions to
pathophysiological responses that emerge during pregnancy by providing
access to high-quality prenatal care, more attention should be focused on
improving (if not optimizing) health during the preconception and interconception periods. This idea led to a set of new initiatives focused on girls’ and
women’s reproductive health trajectories, including public health strategies to
improve preconception health and research strategies to better understand
how adversity impacts reproductive health across the life course.
For the past two decades, there has been a growing recognition across the
MCH community that life course health science is building an important evidence base about the central and vital role of health during the prenatal period
and the early years on subsequent lifelong health (Halfon and Hochstein 2002;
Galobardes et al. 2004 and 2008; Power and Kuh 2013). Research on the
changing epidemiology of childhood chronic illness and the growing number
of longitudinal studies documenting the legacy of chronic illness in childhood
on patterns of adult health, morbidity, and mortality are also connecting the
dots between child health and the potential for healthy aging (Halfon 2012;
Wise 2004; Wise 2016). As the United States experiences rapidly rising healthcare costs due to rapidly increasing rates of chronic disease and multi-morbidity,
life course health science is shining a light on the early part of the life-span
when preventable risks are setting in motion the inflammatory, neuroendocrine,
Preface
Preface
vii
and metabolic processes that predispose an individual to degenerative chronic
disorders manifested decades later. The recent IOM report Shorter Lives,
Poorer Health that explores why the United States is the sickest of rich nations
also highlights that the health of children in the United States falls far behind
the health of children in other nations and that these life course determinants
cannot be ignored (Woolf and Aron 2013).
Perhaps the most salient and obvious reason for MCH to adopt a life
course perspective has come from the epidemic of childhood obesity, which
has demonstrated how childhood growth can influence rates of the most common and costly adult health conditions, including diabetes and cardiovascular
disease (Gillman 2004). It has also shown how a mother’s prenatal health,
along with her preconception weight, influences pregnancy outcomes, the
likelihood that an infant will be obese, and the potential for lifelong obesity
and resultant comorbidities (Oken and Gillman 2003; Gillman et al. 2008).
For at least the past two decades, life course health science research has
been reframing our approach to many persistent health and health-care issues,
from infant mortality to obesity, and from school readiness to lifelong cognitive potential and reserves. This research has influenced thought leaders,
researchers, policymakers, and service providers to consider the importance
and essential role of MCH as a vehicle for improving health outcomes for
mothers and children and, ultimately, for the population as a whole. In 2010,
as MCHB celebrated its 75th birthday, Peter Van Dyck, the Associate
Administrator of the Health Resources and Services Administration and
Director of MCHB, announced that the Bureau intended to launch a national
dialogue about the importance of life course health science in reaching MCH
goals. He also highlighted how MCH could use this science to help research,
programs, policies, and partnerships coalesce around moving life course theory into life course practice. The transformation would be accomplished by
an integrative approach to understanding how health and disease develop.
However, although this transformation is aimed at creating a rigorous
approach to the study of the development of heath across the life-span, there
is no doubt that there are still many outstanding questions about the relationship between early experiences and lifelong health and well-being, and about
how existing and emerging knowledge can be applied to the development of
evidence-based practice and policy.
Unfortunately, the lack of a strong research and data infrastructure, coupled with limits on funding currently available in the United States to support
the development of new methodologies and collaborative approaches, has
hampered the production of the transformative, transdisciplinary, and translational research that is needed to advance the emerging field we have termed
“life course health development” (LCHD). Moreover, the fact that researchers who are interested and engaged in LCHD research continue to work in
discipline-specific silos has been a significant impediment to rapid progress.
In recognition of and response to these challenges, in 2010, MCHB issued a
Request for Proposals to develop a Maternal and Child Health Life Course
Research Network (LCRN) that would be charged with providing a virtual
platform and undertaking a set of activities that would together serve as a new
infrastructure for catalyzing progress and enhancing funding to support basic,
viii
theoretical, and applied and translational LCHD research of relevance to
MCH practice and policy.
The UCLA Center for Healthier Children, Families, and Communities – with
the support and participation of a diverse array of colleagues from around the
United States – submitted a successful application to establish an LCRN with
the following goals:
1. Engage a diverse, active, and sustainable community of LCHD
stakeholders.
2. Increase capacity for, engagement in, and production of LCHD research.
3. Catalyze the translation and application of LCHD research to practice and
policy.
To launch the LCRN, the UCLA team initiated a strategic network design
process that engaged individuals with substantial expertise in health development, as well as those with deep knowledge of the science of network development and facilitation. This strategic design process included a series of
interviews with key informants (see http://www.lcrn.net/tag/expert-interviews), as well as an in-person meeting of the network’s 30-member design
team that resulted in the approval of the LCRN charter (see http://www.lcrn.
net/wp-content/uploads/2012/07/LCRN-charter-2.pdf), the development of a
scope of work comprised of specific activities intended to achieve the network’s aims, a concept for the network’s online presence including a website
and social networking platform, and the constitution of an advisory committee that would provide UCLA project staff with guidance for the duration of
the project (see http://www.lcrn.net/about).
Following the design meeting, project staff undertook a process to develop
a series of background papers that would serve as the basis for the MCH Life
Course Research Agenda-Setting Meeting that took place in February of
2013 in order to achieve the following aims:
1. Catalyze a paradigm shift in how researchers, practitioners, and policymakers think about, understand, and promote LCHD.
2. Evaluate, refine, and determine the utility of the seven proposed principles
of LCHD.
3. Identify the ways in which the topics discussed at the meeting are converging and/or diverging across disciplines.
4. Identify knowledge that is ready for application in order to assist MCH
and other practitioners in taking advantage of what we know now and
speeding the progression from research to translation.
5. Provide recommendations that will enable the LCRN to develop an MCH
Life Course Research Agenda (LCRA) that includes priorities in the areas
of basic research, translational research, and methods and data
development.
6. Provide background paper authors with input that will advance their
papers toward completion and publication.
7. Identify next steps for both the LCRN and the LCHD field as a whole.
Preface
Preface
ix
Background paper topics were selected by project staff with the input of
the LCRN advisory committee and MCHB staff, and included topics that
were selected strategically due to their potential to enhance our understanding of health development and advance the LCHD field, as well as topics that
were selected more opportunistically when researchers learned of the project
and wanted to ensure that the issues of importance to them had a chance of
making it into the preliminary version of the LCRA, version 1.0 (see concluding chapter of this volume).
The 2013 agenda-setting meeting brought together 90+ invited stakeholders
including researchers, practitioners, policymakers, funders, and other thought
leaders from the United States, Canada, and the United Kingdom. Over the
2-day meeting, participants engaged in a highly facilitated process of reviewing
the evidence base and providing the background paper authors with the feedback they would need to complete their research and develop a set of recommended research priorities. A highlight of the meeting was to critically examine
the seven proposed principles of LCHD (see Halfon and Forrest in this volume)
that were intended to provide a more unified theoretical foundation and a more
consistent set of terminology for this emerging field.
Following the agenda-setting meeting and in response to the enormous
amount of momentum and enthusiasm generated among the participants,
UCLA staff, again with the guidance of the LCRN advisory committee and
representatives from MCHB, began to pursue development and publication of
a volume that would contain revised versions of the background papers, as
well as several chapters to be commissioned based on gaps identified at the
agenda-setting meeting, plus a preliminary version of the LCRA. To this end,
a four-member LCRN editorial team was constituted and charged with working closely with the background paper authors to ready their drafts – with a
particular focus on trying to align the chapters with regard to the terminology
and, more importantly, the conceptual frameworks underlying the writings –
for inclusion in the Handbook of Life Course Health Development, and
develop additional chapters and material as needed.
Concurrent with the preparation of this volume, the LCRN has produced
three unique webinar series, organized research nodes focused on particular
topic areas, developed strategic partnerships aimed at enabling the translation
of LCHD research to practice and policy, and produced several peer-reviewed
publications, among other activities. We invite readers to learn more about
the LCRN – including how to join – at lcrn.net.
References
Ben-Shlomo, Y., & Kuh, D. (2002). A life course approach to chronic disease epidemiology: conceptual models, empirical challenges and interdisciplinary perspectives.
International journal of epidemiology, 31(2), 285293.
Galobardes, B., Lynch, J. W., & Davey Smith, G. (2004). Childhood socioeconomic circumstances and cause-specific mortality in adulthood: systematic review and interpretation.
Epidemiology Reviews, 26, 7–21.
Galobardes, B., Lynch, J. W., & Smith, G. D. (2008). Is the association between childhood
socioeconomic circumstances and cause-specific mortality established? Update of a
systematic review. Epidemiology and Community Health, 6(2), 387–90.
x
Gillman, M. W. (2004). A life course approach to obesity. A life course approach to chronic
disease epidemiology. 1, 473.
Gillman, M. W., Rifas-Shiman, S. L., Kleinman, K., Oken, E., Rich-Edwards, J. W., &
Taveras, E. M. (2008). Developmental origins of childhood overweight: potential
public health impact. Obesity, 16(7), 1651–6.
Halfon, N., & Hochstein, M. (2002). Life course health development: an integrated framework for developing health, policy, and research. Milbank Quarterly, 80(3), 433–79. iii.
Halfon, N., Houtrow, A., Larson, K., & Newacheck, P. W. (2012). The changing landscape
of disability in childhood. The Future of Children, 22(1), 13–42.
Lu, M. C., & Halfon, N. Racial and ethnic disparities in birth outcomes: a life-course perspective. Maternal and Child Health, 7(1), 13–30.
Oken, E., & Gillman, M. W. (2003). “Fetal origins of obesity.” Obesity research, 11.4,
496–506.
Power, C., Kuh, D., & Morton, S. (2013). From developmental origins of adult disease
to life course research on adult disease and aging: insights from birth cohort studies.
Annual Review of Public Health, 34, 7–28.
Rosenfeld, A., & Min, C. J. (2009). A history of international cooperation in maternal and
child health. Maternal and child health (pp. 3–17). US: Springer.
Wise, P. H. (2004). The transformation of child health in the United States. Health Affairs,
23(5), 9–25.
Wise, P. H. (2016). Child poverty and the promise of human capacity: childhood as a foundation for healthy aging. Academic Pediatrics, 16(3), S37–45.
Woolf, S. H., & Aron, L, (Eds). (2013) US health in international perspective: Shorter lives,
poorer health. National Academies Press.
Preface
Acknowledgments
This project is supported by the Health Resources and Services Administration
(HRSA) of the US Department of Health and Human Services (HHS) under
grant number UA6MC19803. This information or content and conclusions
are those of the author(s) and should not be construed as the official position
or policy of, nor should any endorsements be inferred by, HRSA, HHS, or the
US Government.
xi
Contents
Introduction to the Handbook of Life Course
Health Development.............................................................................
Neal Halfon, Christopher B. Forrest, Richard M. Lerner,
Elaine M. Faustman, Ericka Tullis, and John Son
Part I
Emerging Frameworks
The Emerging Theoretical Framework of Life Course
Health Development.............................................................................
Neal Halfon and Christopher B. Forrest
Part II
1
19
Life Stages
Preconception and Prenatal Factors and Metabolic Risk ................
Guoying Wang, Tami R. Bartell, and Xiaobin Wang
47
Early Childhood Health and the Life Course:
The State of the Science and Proposed Research Priorities .............
W. Thomas Boyce and Clyde Hertzman
61
Middle Childhood: An Evolutionary-Developmental
Synthesis................................................................................................
Marco DelGiudice
95
Adolescent Health Development: A Relational
Developmental Systems Perspective ...................................................
Richard M. Lerner, Claire D. Brindis, Milena Batanova,
and Robert Wm. Blum
Emerging Adulthood as a Critical Stage
in the Life Course .................................................................................
David Wood, Tara Crapnell, Lynette Lau, Ashley Bennett,
Debra Lotstein, Maria Ferris, and Alice Kuo
Pregnancy Characteristics and Women’s
Cardiovascular Health.........................................................................
Abigail Fraser, Janet M. Catov, Deborah A. Lawlor,
and Janet W. Rich-Edwards
109
123
145
xiii
xiv
Contents
Part III The Life Course Origins and Consequences
of Select Major Health Conditions and Issues
Early in the Life Course: Time for Obesity Prevention....................
Summer Sherburne Hawkins, Emily Oken, and Matthew W. Gillman
Pediatric Type 2 Diabetes: Prevention and Treatment
Through a Life Course Health Development Framework ...............
Pamela Salsberry, Rika Tanda, Sarah E. Anderson,
and Manmohan K. Kamboj
Life Course Health Development in Autism
Spectrum Disorders .............................................................................
Irene E. Drmic, Peter Szatmari, and Fred Volkmar
Self-Regulation .....................................................................................
Megan McClelland, John Geldhof, Fred Morrison,
Steinunn Gestsdóttir, Claire Cameron, Ed Bowers,
Angela Duckworth, Todd Little, and Jennie Grammer
A Life Course Health Development Perspective
on Oral Health ......................................................................................
James J. Crall and Christopher B. Forrest
Life Course Health Development Outcomes After
Prematurity: Developing a Community, Clinical,
and Translational Research Agenda to Optimize
Health, Behavior, and Functioning .....................................................
Michael E. Msall, Sarah A. Sobotka, Amelia Dmowska,
Dennis Hogan, and Mary Sullivan
A Life Course Approach to Hearing Health ......................................
Shirley A. Russ, Kelly Tremblay, Neal Halfon, and Adrian Davis
169
197
237
275
299
321
349
Chronic Kidney Disease: A Life Course Health Development
Perspective ............................................................................................ 375
Patrick D. Brophy, Jennifer R. Charlton, J. Bryan Carmody,
Kimberly J. Reidy, Lyndsay Harshman, Jeffrey Segar, David Askenazi,
David Shoham, and Susan P. Bagby
Part IV
Crosscutting Topics in Life Course Health Development
Growth and Life Course Health Development ..................................
Amanda Mummert, Meriah Schoen, and Michelle Lampl
From Epidemiology to Epigenetics: Evidence
for the Importance of Nutrition to Optimal Health
Development Across the Life Course..................................................
Marion Taylor-Baer and Dena Herman
405
431
Contents
xv
How Socioeconomic Disadvantages Get Under
the Skin and into the Brain to Influence Health Development
Across the Lifespan ..............................................................................
Pilyoung Kim, Gary W. Evans, Edith Chen, Gregory Miller,
and Teresa Seeman
463
Health Disparities: A Life Course Health Development Perspective
and Future Research Directions ......................................................... 499
Kandyce Larson, Shirley A. Russ, Robert S. Kahn, Glenn Flores,
Elizabeth Goodman, Tina L. Cheng, and Neal Halfon
Part V
Methodological Approaches
Core Principles of Life Course Health
Development Methodology and Analytics..........................................
Todd D. Little
Epidemiological Study Designs: Traditional
and Novel Approaches to Advance Life Course
Health Development Research ............................................................
Stephen L. Buka, Samantha R. Rosenthal, and Mary E. Lacy
523
541
Using the National Longitudinal Surveys of Youth (NLSY)
to Conduct Life Course Analyses........................................................
Elizabeth C. Cooksey
561
Using the Panel Study of Income Dynamics (PSID)
to Conduct Life Course Health Development Analysis ....................
Narayan Sastry, Paula Fomby, and Katherine McGonagle
579
Using the Fragile Families and Child Wellbeing Study (FFCWS)
in Life Course Health Development Research ..................................
Amanda Geller, Kate Jaeger, and Garrett Pace
601
Part VI
Future Directions
Life Course Research Agenda (LCRA),
Version 1.0.............................................................................................
Neal Halfon, Christopher B. Forrest, Richard M. Lerner,
Elaine M. Faustman, Ericka Tullis, and John Son
623
Errata to: Handbook of Life Course Health Development ..............
E1
Index ......................................................................................................
647
The original version of this book was revised. An erratum to this book can be found at
https://doi.org/10.1007/978-3-319-47143-3_27
About the Editors
Neal Halfon, MD, MPH is the Director of the UCLA Center for Healthier
Children, Families, and Communities. He is also a Professor of pediatrics in
the David Geffen School of Medicine at UCLA, of health policy and management in the UCLA Fielding School of Public Health, and of public policy in
the UCLA Luskin School of Public Affairs. Dr. Halfon’s research has spanned
clinical, health services, epidemiologic, and health policy domains. For more
than a decade, he has worked with national, state, and local initiatives aimed
at improving early childhood systems. Dr. Halfon has also played a significant role in developing new conceptual frameworks for the study of health
and health care, including the Life Course Health Development (LCHD)
framework. In 2006, Halfon received the Academic Pediatric Associations
Annual Research Award for his lifetime contributions to child health research.
He received his MD at the University of California, Davis, and MPH at the
University of California, Berkeley. He completed his pediatric residency at
the University of California, San Diego, and the University of California, San
Francisco. Dr. Halfon was also Robert Wood Johnson Clinical Scholar at the
University of California, San Francisco.
Christopher B. Forrest, MD, PhD is Professor of Pediatrics and Health
Care Management at the Children’s Hospital of Philadelphia (CHOP) and the
University of Pennsylvania. He is the Director of the CHOP Center for Applied
Clinical Research, which uses life course health development science to
advance clinical and health services research in pediatrics. Dr. Forrest serves as
the Principal Investigator of the PEDSnet (pedsnet.org), a national consortium
of children’s hospitals (>5 million children) that conducts patient-centered outcomes research among children and youth. He is the Chair of the Research
Committee for PCORnet, the national clinical research network funded by
PCORI. He also chairs the Steering Committee for the NIH program called
PEPR, which is conducting longitudinal studies on person-reported outcome
measures in children with chronic conditions. Dr. Forrest received his BA and
MD degrees from Boston University and completed his PhD in Health Policy
and Management at Johns Hopkins School of Public Health.
Richard M. Lerner, PhD is the Bergstrom Chair in Applied Developmental
Science and the Director of the Institute for Applied Research in Youth
Development at Tufts University. He went from kindergarten through PhD
within the New York City public schools, completing his doctorate at the City
xvii
xviii
University of New York in 1971 in developmental psychology. Dr. Lerner has
more than 700 scholarly publications, including 80 authored or edited books.
He was the Founding Editor of the Journal of Research on Adolescence and
Applied Developmental Science, which he continues to edit. He was a 1980–
1981 fellow at the Center for Advanced Study in the Behavioral Sciences and
is a fellow of the American Association for the Advancement of Science, the
American Psychological Association, and the Association for Psychological
Science. Dr. Lerner is known for his theory of relations between life-span
human development and social change and for his research about the relations
between adolescents and their peers, families, schools, and communities. His
work integrates the study of public policies and community-based programs
with the promotion of positive youth development and youth contributions to
civil society. He is married to Dr. Jacqueline V. Lerner, Professor in the
Department of Applied Developmental and Educational Psychology in the
Lynch School of Education at Boston College. They live in Wayland,
Massachusetts. They have three children: Justin, a Director and Screenwriter
living in Los Angeles; Blair, an Advertising Executive at Media Contacts in
Boston; and Jarrett, a Novelist and Editor living in Boston.
Elaine M. Faustman, PhD, DABT is Professor in the Department of
Environmental and Occupational Health and Director of the Institute for Risk
Analysis and Risk Communication at the School of Public Health and
Community Medicine at the University of Washington. She directs the
NIEHS- and EPA-funded Center for Child Environmental Health Risks
Research and led the Pacific Northwest Center for the National Children’s
Study. She is an elected fellow of both the American Association for the
Advancement of Science and the Society for Risk Analysis. Dr. Faustman is
an Affiliate Professor in the School of Public Affairs at the University of
Washington and has been an Affiliate Professor in the Department of
Engineering and Public Policy at Carnegie-Mellon University. She has served
on the National Toxicology Program Board of Scientific Counselors, the
National Academy of Sciences Committee on Toxicology, the Institute of
Medicine Upper Reference Levels Subcommittee of the Food and Nutrition
Board, and numerous editorial boards. Dr. Faustman chaired the National
Academy of Sciences Committee on Developmental Toxicology. She has or
is currently serving on the executive boards of the Society of Toxicology, the
Society for Risk Analysis, and NIEHS Council. She is past President of the
Teratology Society. Her research interests include understanding mechanisms
that put children and the public at risk of environmental agents. Currently she
is serving on the Committee on NAS Gulf War and Health, Volume 11:
Generational Health Effects of Serving in the Gulf War. In particular, Dr.
Faustman is interested in the molecular and cellular mechanisms of developmental and reproductive toxicants, characterizing in vitro techniques for
developmental toxicology assessment, and development of biologically
based dose-response models for non-cancer risk assessment. Dr. Faustman’s
research expertise includes development of decision-analytic tools for communicating and translating new scientific findings into risk assessment and
risk management decisions.
About the Editors
Contributors
Sarah E. Anderson, PhD The Ohio State University, College of Public
Health, Columbus, OH, USA
David Askenazi, MD University of Alabama Children’s Hospital, Pediatric
Nephrology, Birmingham, AL, USA
Susan P. Bagby, MD Division of Nephrology & Hypertension, Department
of Medicine, Moore Institute for Nutrition and Wellness, Oregon Health &
Science University, Portland, OR, USA
Tami R. Bartell, BA Stanley Manne Children’s Research Institute, Ann &
Robert H Lurie Children’s Hospital of Chicago, Chicago, IL, USA
Milena Batanova, PhD Institute for Applied Research in Youth
Development, Tufts University, Medford, MA, USA
Ashley Bennett, MD Department of Pediatrics, USC, Los Angeles,
CA, USA
Robert Wm. Blum, MD, MPH, PhD Johns Hopkins Bloomberg School of
Public Health, Baltimore, MD, USA
Ed Bowers Clemson University, Youth Development Leadership, Clemson,
SC, USA
Claire D. Brindis, DrPH University of California-San Francisco (UCSF),
Philip R. Lee Institute for Health Policy Studies and the Adolescent and
Young Adult Health National Resource Center, San Francisco, CA, USA
Patrick D. Brophy, MD, MHCDS University of Iowa Stead Family
Children’s Hospital, Pediatric Nephrology, Iowa City, IA, USA
J. Bryan Carmody, MD University of Virginia, Department of Pediatrics,
Division of Nephrology, Charlottesville, VA, USA
Stephen L. Buka Department of Epidemiology, Brown University,
Providence, RI, USA
Claire Cameron University at Buffalo, SUNY, Learning and Instruction,
Buffalo, NY, USA
xix
xx
Janet M. Catov Department of Obestetrics and Gynecology, University of
Pittsburgh, Pittsburgh, PA, USA
University of Pittsburgh, Department of Epidemiology, Pittsburgh, PA, USA
Magee-Womens Research Institute, Pittsburgh, PA, USA
Jennifer R. Charlton, MD, MSc University of Virginia, Department of
Pediatrics, Division of Nephrology, Charlottesville, VA, USA
Edith Chen Department of Psychology and Institute for Policy Research,
Northwestern University, Evanston, IL, USA
Tina L. Cheng, MD, MPH Department of Pediatrics, Johns Hopkins
University School of Medicine, Baltimore, MD, USA
Elizabeth C. Cooksey Center for Human Resource Research, The Ohio
State University, Columbus, OH, USA
James J. Crall Division of Public Health and Community Dentistry,
University of California Los Angeles (UCLA) School of Dentistry, Los
Angeles, CA, USA
Tara Crapnell, OTD, OTR/L UCLA, Department of Pediatrics, Los
Angeles, CA, USA
Adrian Davis University College London, NHS Newborn Hearing
Screening Program, London, UK
Amelia Dmowska Section of Developmental and Behavioral Pediatrics,
University of Chicago Comer Children’s Hospitals, Chicago, IL, USA
Irene E. Drmic Hospital for Sick Children, Toronto, ON, Canada
Angela Duckworth University of Pennsylvania, Department of Psychology,
Philadelphia, PA, USA
Gary W. Evans Department of Design and Environmental Analysis,
Department of Human Development, Cornell University, Ithaca, NY, USA
Elaine M. Faustman, PhD Institute for Risk Analysis and Risk
Communication, Department of Environmental and Occupational Health
Sciences, School of Public Health, University of Washington, Seattle, WA,
USA
Maria Ferris, MD, PhD, MPH UCLA, Department of Pediatrics, Los
Angeles, CA, USA
Glenn Flores, MD, FAAP Medica Research Institute, Division of Health
Policy and Management, University of Minnesota School of Public Health,
Minneapolis, MN, USA
Paula Fomby Survey Research Center, Institute for Social Research,
University of Michigan, Ann Arbor, MI, USA
Christopher B. Forrest, MD, PhD Applied Clinical Research Center,
Children’s Hospital of Philadelphia, Philadelphia, PA, USA
Contributors
Contributors
xxi
Abigail Fraser Medical Research Council Integrative Epidemiology Unit at
the University of Bristol, University of Bristol, Bristol, UK
John Geldhof Oregon State University, Human Development and Family
Sciences, Corvallis, OR, USA
Amanda Geller New York University, New York, NY, USA
Steinunn Gestsdóttir University of Iceland, Department of Psychology,
Reykjavik, Iceland
Matthew W. Gillman Harvard Medical School and Harvard Pilgrim Health
Care Institute, Boston, MA, USA
Marco DelGiudice Department of Psychology, University of New Mexico,
Albuquerque, NM, USA
Elizabeth Goodman, MD Division of General Academic Pediatrics, Mass
General Hospital for Children, Department of Pediatrics, Harvard Medical
School, Boston, MA, USA
Jennie Grammer University of California, Los Angeles, Graduate School
of Education and Information Studies, Los Angeles, CA, USA
Neal Halfon, MD, MPH Department of Pediatrics, David Geffen School of
Medicine, UCLA, Los Angeles, CA, USA
Department of Health Policy and Management, Fielding School of Public
Health, UCLA, Los Angeles, CA, USA
Department of Public Policy, Luskin School of Public Affairs, UCLA,
Los Angeles, CA, USA
Center for Healthier Children, Families, and Communities, UCLA, Los Angeles,
CA, USA
Lyndsay Harshman, MD University of Iowa Children’s Hospital, Pediatrics,
Iowa City, IA, USA
Summer Sherburne Hawkins Boston College, Chestnut Hill, MA, USA
Dena Herman Department of Family and Consumer Sciences, California
State University Northridge, Northridge, CA, USA
Clyde Hertzman Human Early Learning Partnership, School of Population
and Public Health, University of British Columbia, Vancouver, BC, Canada
Dennis Hogan, PhD Sociology and Demography, Population Research and
Training Center, Brown University, Providence, RI, USA
Kate Jaeger Princeton University, Princeton, NJ, USA
Robert S. Kahn, MD, MPH Division of General and Community Pediatrics,
Cincinnati Children’s Hospital Medical Center, University of Cincinnati
College of Medicine, Cincinnati, OH, USA
Manmohan K. Kamboj, MD The Ohio State University, College of
Medicine, Endocrinology, Metabolism and Diabetes, Nationwide Children’s
Hospital, Columbus, OH, USA
xxii
Pilyoung Kim Department of Psychology, University of Denver, Denver,
CO, USA
Alice Kuo, MD, PhD UCLA, Department of Pediatrics, Los Angeles, CA,
USA
Mary E. Lacy Department of Epidemiology, Brown University, Providence,
RI, USA
Michelle Lampl, MD, PhD Department of Anthropology, Emory University,
Atlanta, GA, USA
Center for the Study of Human Health, Emory University, Atlanta, GA, USA
Kandyce Larson, PhD Department of Research, American Academy of
Pediatrics, Elk Grove Village, IL, USA
Lynette Lau, PhD UCLA, Department of Pediatrics, Los Angeles, CA,
USA
Deborah A. Lawlor Medical Research Council Integrative Epidemiology
Unit at the University of Bristol, University of Bristol, Bristol, UK
Richard M. Lerner, PhD Tufts University, Medford, MA, USA
Todd Little Texas Tech University, Department of Educational Psychology
and Leadership, Lubbock, TX, USA
Debra Lotstein, MD, MPH UCLA, Department of Pediatrics, Los Angeles,
CA, USA
Megan McClelland Human Development and Family Sciences, 245 Hallie
E. Ford Center for Healthy Children and Families, Oregon State University,
Corvallis, OR, USA
Katherine McGonagle Survey Research Center, Institute for Social
Research, University of Michigan, Ann Arbor, MI, USA
Gregory Miller Department of Psychology and Institute for Policy Research,
Northwestern University, Evanston, IL, USA
Fred Morrison University of Michigan, Department of Psychology, Ann
Arbor, MI, USA
Michael E. Msall, MD Developmental and Behavioral Pediatrics University
of Chicago, Comer and LaRabida Children’s Hospitals, Chicago, IL, USA
JP Kennedy Research Center on Intellectual and Developmental Disabilities,
University of Chicago Comer Children’s Hospital, Section of Developmental
and Behavioral Pediatrics, Chicago, IL, USA
Amanda Mummert, PhD Department of Anthropology, Emory University,
Atlanta, GA, USA
Center for the Study of Human Health, Emory University, Atlanta, GA, USA
Emily Oken Harvard Medical School and Harvard Pilgrim Health Care
Institute, Boston, MA, USA
Contributors
Contributors
xxiii
Garrett Pace Doctoral Student, School of Social Work, Department of
Sociology, University of Michigan, Ann Arbor, MI, USA
Kimberly J. Reidy, MD Albert Einstein College of Medicine, Montefiore
Medical Center, Pediatric Nephrology, Bronx, NY, USA
Janet W. Rich-Edwards Connors Center for Women’s Health and Gender
Biology, Brigham and Women’s Hospital, Boston, MA, USA
Harvard Medical School, Boston, MA, USA
Harvard School of Public Health, Boston, MA, USA
Samantha R. Rosenthal Department of Epidemiology, Brown University,
Providence, RI, USA
Shirley A. Russ, MD, MPH UCLA Center for Healthier Children, Families
and Communities, Department of Pediatrics, David Geffen School of
Medicine, UCLA, Los Angeles, CA, USA
Pamela Salsberry, PhD, RN, FAAN College of Public Health, Division of
Health Behavior, Health Promotion, Institute for Population Health, The
Ohio State University, Columbus, OH, USA
Narayan Sastry Survey Research Center, Institute for Social Research,
University of Michigan, Ann Arbor, MI, USA
Meriah Schoen Center for the Study of Human Health, Emory University,
Atlanta, GA, USA
Department of Nutrition, Georgia State University, Atlanta, GA, USA
Teresa Seeman David Geffen School of Medicine, University of California –
Los Angeles, Los Angeles, CA, USA
Jeffrey Segar, MD University of Iowa Children’s Hospital, Neonatology,
Iowa City, IA, USA
David Shoham, PhD, MSPH Department of Public Health Sciences,
Loyola University Chicago, Maywood, IL, USA
Sarah A. Sobotka, MD, MS Section of Developmental and Behavioral
Pediatrics, University of Chicago Comer Children’s Hospitals, Chicago,
IL, USA
John Son, MPH Center for Healthier Children, Families and Communities,
UCLA, Los Angeles, CA, USA
Mary Sullivan, PhD, RN University of Rhode Island, College of Nursing,
Women and Infants Hospital, Providence, RI, USA
Peter Szatmari Centre for Addiction and Mental Health, Hospital for Sick
Children, University of Toronto, Toronto, ON, Canada
Rika Tanda, PhD, RN College of Health Science and Professions, Ohio
University, Athens, OH, USA
xxiv
Marion Taylor-Baer Department of Community Health Sciences, Fielding
School of Public Health, University of California Los Angeles, Los Angeles,
CA, USA
W. Thomas Boyce Departments of Pediatrics and Psychiatry, University of
California San Francisco, San Francisco, CA, USA
Kelly Tremblay Speech & Hearing Sciences College of Arts & Sciences,
University of Washington, Seattle, WA, USA
Ericka Tullis, MPP Center for Healthier Children, Families and
Communities, UCLA, Los Angeles, CA, USA
Fred Volkmar Child Study Center, Yale University School of Medicine,
New Haven, CT, USA
Guoying Wang, MD, PhD Department of Population, Family and
Reproductive Health, Center on the Early Life Origins of Disease, Johns
Hopkins University Bloomberg School of Public Health, Baltimore, MD,
USA
Xiaobin Wang, MD, MPH, ScD Center on the Early Life Origins of
Disease, Department of Population, Family and Reproductive Health, Johns
Hopkins University Bloomberg School of Public Health, Baltimore, MD,
USA
David Wood, MD, MPH Department of Pediatrics, ETSU College of
Medicine, Johnson City, TN, USA
Contributors
Introduction to the Handbook
of Life Course Health Development
Neal Halfon, Christopher B. Forrest,
Richard M. Lerner, Elaine M. Faustman,
Ericka Tullis, and John Son
1
Introduction
Over the past several decades, countless studies
have linked early life events and experiences with
adult health conditions, delineating the developmental origins of common chronic health conditions and specifying the processes by which both
adversity and opportunity are integrated into
developing biobehavioral systems (Baltes et al.
2006; Bronfenbrenner 2005; Elder et al. 2015). As
a result, there is a greater understanding of how
health and disease develop, which is leading to
new kinds of individual- and population-level
strategies that have the potential to prevent disease
and optimize health by minimizing the impact of
adversity, increasing protective factors, and targeting health-promoting interventions to coincide
with sensitive periods of health development.
Insights and evidence from life course chronic
disease epidemiology have converged with
research from the fields of developmental biology, neuroscience, and developmental science,
with studies of typical and atypical development
and with new findings from research examining
the developmental origins of chronic disease.
This wide-ranging research, all focused on understanding how health and disease develop, has
involved researchers from a wide variety of disciplines. Life-span developmental psychologists,
N. Halfon, MD, MPH (*)
Department of Pediatrics, David Geffen School of
Medicine, UCLA, Los Angeles, CA, USA
Department of Health Policy and Management,
Fielding School of Public Health, UCLA,
Los Angeles, CA, USA
Department of Public Policy, Luskin School of Public
Affairs, UCLA, Los Angeles, CA, USA
Center for Healthier Children, Families, and
Communities, UCLA, Los Angeles, CA, USA
e-mail: nhalfon@ucla.edu
C.B. Forrest, MD, PhD
Applied Clinical Research Center,
Children’s Hospital of Philadelphia,
Philadelphia, PA, USA
R.M. Lerner, PhD
Tufts University, Medford, MA, USA
E.M. Faustman, PhD
Institute for Risk Analysis and Risk Communication,
Department of Environmental and Occupational
Health Sciences, School of Public Health, University
of Washington, Seattle, WA 98105, USA
E. Tullis, MPP • J. Son, MPH
Center for Healthier Children, Families and
Communities, UCLA, Los Angeles, CA, USA
© The Author(s) 2018
N. Halfon et al. (eds.), Handbook of Life Course Health Development,
DOI 10.1007/978-3-319-47143-3_1
1
N. Halfon et al.
2
life course-focused sociologists, human capitalfocused economists, and political scientists
studying the structure of social institutions are
not only studying the same developmental processes; they are also working alongside epidemiologists, physicians, and basic scientists to better
understand how health develops over the life
course and how these health development processes promote human flourishing.
In response to this burgeoning knowledge,
there is growing momentum among practitioners
and policymakers to “connect the dots” between
what we know and what we do, that is, between
the rapidly expanding evidence base from the
emerging field of life course health development
(LCHD) and the practices and policies that are
implemented within the fields of medicine, public
health, nursing, mental health, education, urban
planning, community development, social welfare, and others (Halfon et al. 2014; Kuh et al.
2013; Braveman 2014; Gee et al. 2012; Lappé
and Landecker 2015). At the same time, there is a
strong impetus among researchers to continue to
fill the substantial gaps in our knowledge and to
ensure that research findings are appropriately
synthesized and translated before being applied in
clinical, public health, or public policy contexts.
Comprised of 26 chapters that grew out of the
2013 Maternal and Child Health (MCH) Life
Course Research Agenda-Setting Meeting that
was organized by the MCH Life Course Research
Network (LCRN) and funded by Health Resources
and Services Administration-Maternal and Child
Health Bureau (HRSA-MCHB), this volume represents a groundbreaking effort to explore the history of the LCHD field, to take stock of what we
know and do not know about how health and disease develop, to provide practitioners and policymakers with guidance regarding the kinds of
interventions and efforts that can be beneficial, and
to lay the foundation for a research agenda that
identifies high-priority areas for basic, clinical,
population, and translational investigations in
order to strategically target resources and efforts
and advance the life course health sciences.
Each chapter is written by a team of leading
experts that often spans several different disciplines
and therefore reflects a wide range of perspectives
on how innovative research, practice, and policy
can begin to address our most pressing health challenges. Similarly, the volume’s four editors represent different disciplines and perspectives that were
brought to bear on the process of selecting topics
and authors and on ensuring that each chapter
makes a substantial contribution to the field.
In this introductory chapter, we begin by providing a rationale for the publication of this volume, including an historical overview that traces
the emergence of the LCHD field and provides
evidence of a significant, but as yet incomplete,
transformation in how we think about and promote health. We go on to describe the purpose,
structure, and content of the volume and to examine some of the challenges for further field building in this area. Finally, we provide readers with
information about each section and chapter in
this volume, including the impressive backgrounds of the various experts who contributed
substantial amounts of both time and original
thinking in their roles as authors.
2
Rationale
2.1
The Emergence of a New Field
The science of health has been guided for well
over 150 years by a mechanical model that views
the body and its component cells as machines
and views disease as a breakdown in organ structure and function. Person-environment relationships as causes of disease are either ignored or
relegated to secondary concerns. Even though the
oversimplified perspective of the body as a
machine has been largely abandoned, reducing
physiologic and behavioral phenomena to their
smallest observable constituent parts remains a
mainstay of the biomedical model that dominates
contemporary health sciences. This reductionist
approach may tell us how parts of a neuron work,
but it does not provide an appropriate model for
understanding how the nervous system works,
how we think, or what produces consciousness
(Capra 1982). Even fields like human genomics
are moving away from the simplistic notion of
single-gene causation, which has failed to yield
substantial insights into disease causation, to
research on genetic networks and epigenetics
Introduction to the Handbook of Life Course Health Development
(Huang 2012; Lappé and Landecker 2015).
Complex disorders that manifest as a spectrum of
phenotypic variability – including cardiovascular
disease, obesity, diabetes, and autism – are
increasingly understood as manifestations of
relations among networks of genes and complex
gene-environment coactions that are mediated by
equally complex time signatures and temporal
coupling. Moreover, the mechanical model of
health cannot account for placebo effects, the
mind’s effects on the body, or psychosomatic illness. It presupposes a mind-body dualism and
reifies the distinction between physical and mental health, a vestige of the Cartesian mind-body
split (Overton 2015). A reductionist approach to
understanding health is inadequate for addressing
how different molecular, physiologic, social, and
environmental networks work together to produce dynamic stability and change, which are the
cornerstones of health outcome trajectories.
Many fields of science – including physics,
biology, and the social sciences (especially developmental science) – have shifted from a CartesianNewtonian mechanistic ontology to a more
complex system-oriented ontology (Lerner 2012).
The mechanistic view divides the world into separate or split categories (e.g., nature versus nurture)
and reduces it into discrete elements (genes, behaviors, molecules) that are combined, added, and
assembled to form what we perceive as biological
phenotypes, patterns of behavior, and personalities. As Overton (2012) and others have described,
this revolutionary shift in the epistemological and
ontological foundations of science took place during the twentieth century as Newtonian physics
gave way to general relativity theory and as contemporary formulations of knowing the world
were shown to lack explanatory power and utility
(Aldwin 2014). Thus, the need for new models that
explain the complex phenomena of human health
development became apparent.
The synthesis of human health development
as explained by theories associated with relational developmental systems (RDS) metatheory
is replacing the now anachronistic mechanical
model of health (Lerner and Overton 2008;
Lerner 2012; Overton 2012). Overton (2015)
explains that compared to earlier formulations
of understanding human development, RDS
3
metatheory focuses on process (systematic
changes in the developmental system), becoming
(moving from potential to actuality; a developmental process as having a past, present, and
future; Whitehead 1929/1978), holism (the meanings of entities and events derive from the context in which they are embedded), relational
analysis (assessment of the mutually influential
relations within the developmental system), and
the use of multiple perspectives and explanatory
forms. Within RDS metatheory, the organism is
seen as inherently active, self-creating (autopoietic), self-organizing, self-regulating (agentic),
nonlinear and complex, and adaptive (Overton
2015).
The RDS metatheory emphasizes the study
and integration of different levels of organization
ranging from biology and physiology to culture
and history as a means to understand life-span
human development (Lerner 2006; Overton
2015). Accordingly, the conceptual emphasis in
RDS theories is placed on mutually influential
relations between individuals and contexts, represented as individual ⇔ context relations. In a
bidirectional relational system, the embeddedness within history (temporality) is of fundamental significance (Elder et al. 2015). The presence
of such temporality in the developmental system
means that there always exists some potential for
systematic change and, thus, for (relative) plasticity in human development. In short, potential
plasticity in individual ⇔ context relations
derives from the “arrow of time” (Lerner 1984;
Lerner and Callina 2014; Overton 2015) running
through the integrated (relational) developmental
system. Such plasticity also suggests that there
are multiple developmental pathways, across the
life-span.
Similar conceptual advances have also been generated by systems biology, which focuses on the
complex interactions of biological systems using a
holistic framework and integrative relational strategies rather than traditional reductionist approaches
(Kitano 2002; Antony et al. 2012; Schadt and
Björkegren 2012, Kandel et al. 2014). This transformation has been catalyzed by a greater appreciation
of dynamical system theory and, more specifically,
complex adaptive system theory and its application
to molecular biology (Huang 2012). Moreover, as
4
our understanding of epigenetics and systems biology has matured, new insights into how complex
gene regulatory networks produce multilevel and
multidirectional relationships between genotype
and phenotype have been elucidated (Foster 2011;
Huang 2012; Piro and Di Cunto 2012; Schadt and
Björkegren 2012; Greenblum et al. 2012; DavilaVelderrain et al. 2015). This new knowledge would
not have been acquired using reductionist statistical
models that analyze data by reducing them to their
smallest components and estimating marginal
effects of linear models.
In summary, the study of human development
has evolved from a field dominated by split, reductionist (psychogenic or biogenic) approaches to a
multidisciplinary field that integrates observations,
evidence, and analysis that spans from biological
to cultural and historical levels of organization
across the life-span (e.g., Elder et al. 2015; Ford
and Lerner 1992; Gottlieb 1998; Lerner and
Callina 2014). Reductionist accounts have given
way to a more integrated framework associated
with RDS metatheory (Overton 2015; Lerner
2006). Across the past several decades, several
scholars have provided ideas contributing to the
evolution of this metatheory (e.g., Baltes et al.
2006; Bronfenbrenner 2005; Elder et al. 2015;
Lerner 2006; and, even earlier, see von Bertalanffy
1933).
For instance, in psychology, the transition away
from what some have characterized as the radical
empiricism and atomism of the early behaviorist to
ideas akin to those associated with RDS-based
theories has followed a similar ontological path
(Lerner 2006; Lerner and Overton 2008; Overton
2010, 2012). As Arnold Sameroff explains, psychologists were attempting to find and define the
laws that explain behavior and how the mind functions (Sameroff 2010). As it became clear that any
particular individual- or population-level signal
explained very little of the observed variance in
behaviors, developmental scientists began to create new techniques for analyzing intraindividual
patterns of change that focus on individuals’
unique person-environmental interactions and that
separate the behavioral signal from the noisy complexity of life, especially for long-term predictive
purposes (Molenaar and Newell 2010; Sameroff
N. Halfon et al.
2010). Rather than reducing cognitive, emotional,
or overall mental function to its mechanistic components, this more holistic approach views psychological functioning as the product of a relational
nexus that defines an individual in association with
multiple contexts that interact dynamically over
time.
In the same way that biology and psychology
have faced the limits of reductionist mechanical
models, medicine and health sciences are also
experiencing the constraints of the biomedical
approach that focuses more on the components of
the organism than on the totality of human health.
While the biomedical model has been remarkably successful in defining the components of
human anatomy, physiology, biochemistry, and
metabolism, and has provided useful frameworks
for understanding simple mechanics of more linear disease processes (such as infectious diseases), it is increasingly challenged by the
complexity of health development and by complex disease pathways that emerge out of multilevel and multiphasic processes that include
genetic, biological, behavioral, and wholeorganism processes (Halfon et al. 2014).
Similarly, at the clinical and population health
levels, simple mechanistic biomedical models, or
even more multifaceted biopsychosocial models
of health, have difficulty explaining a wide variety of health phenomena, such as how integration
of body systems and subsystems results in emergent properties of health at the level of the individual; how evolution constrains the timing and
plasticity of human health development; how epigenetic processes result in multiple intermediary
endophenotypes that may progress to pathological phenotypes, hover in subclinical states, or
resolve; how the adaptive capacities of individuals and populations interact with rapidly changing physical, natural, chemical, social, and
nutritional environments to reprogram developing physiology and other regulatory processes
through epigenetic modulations of previously
selected biological and behavioral scripts; and
how integration occurs between biological,
behavioral, and environmental systems, organized and driven by adaptive routines structured
around different developmentally entrained time
Introduction to the Handbook of Life Course Health Development
horizons. These conundrums have challenged
health researchers to develop new frameworks to
explain how each of these complex processes
contributes to the development of health over
time (i.e., contribute to health development).
2.2
The Maturation of the LCHD
Field
A vast amount of empirical literature investigating the developmental, genomic, and epigenetic
origins of health and disease – as well as on the
epidemiology of chronic disease across the life
course – has been generated in the past two
decades (Halfon and Hochstein 2002; Kuh and
Ben-Shlomo 2004; Gluckman and Hanson
2004; Gluckman et al. 2008; Kuh et al. 2013;
Berkman et al. 2014; Halfon et al. 2014; the
evolution of life course health science is
reviewed in Halfon and Forrest 2017). New academic research journals and international
research organizations have been spawned that
focus on the developmental origins of health
and disease. Established professional organizations now include life course and epigenetic and
developmental origins of health and disease
(DOHaD) tracts at their research meetings, and
many major scientific journals have published
special issues packaging articles that focus on
biological embedding, epigenetics, or other disease-causing mechanisms that are framed from
a life course perspective. The US National
Academy of Sciences and National Academy of
Medicine have both issued several reports on
the health, social, and behavioral determinants
of health, health measurement, health disparities, and health-care improvement that have
incorporated a life course perspective, and the
recent framework for the US Healthy People
2020 goals was upgraded to include life course
as an organizing principle of the overall framework (Committee on Future Directions for
Behavioral and Social Sciences Research 2001;
Committee on Evaluation of Children’s Health
2004; Committee on the Recommended Social
and Behavioral Domains and Measures for
Electronic Health Records 2015).
5
Perhaps the biggest stimulus for thinking differently about origins and development of chronic
disease came from a series of provocative studies
that were conducted by David Barker and his
team. Beginning in the 1980s, Barker’s studies
began to describe how the prevalence of heart disease in specific areas of England was related to the
distribution of birth weights in those same regions.
Barker and others went on to use longitudinal
datasets to solidify these observations that birth
weight, and the nutritional environment and exposures of the fetus and infant, had a direct influence
on the development of heart disease that was often
only clinically recognized many decades later
(Barker et al. 1989, 1993; Barker 1995). These
startling findings challenged conventional models
of direct or cumulative risk that posited that heart
disease was the result of either contemporaneous
or lifelong risks including poor nutrition, lack of
exercise, smoking, or other behaviors and suggested that there were important latent effects of
early nutrition that were somehow conditioning
later pathological response patterns. Barker’s studies brought attention to other research with similar
findings that were less dramatic but entirely consistent with the latent lifelong effects that the
Barker studies were revealing. As a result, developmental time frames started to become an important consideration, and the timing of exposures
and the recognition of sensitive periods of development all took on new salience.
As this new perspective on the developmental
origins of chronic disease began to unfold, there
was also another emerging set of new constructs
coming into play in what is now termed the field
of population health. Following on in the tradition of the 1974 Lalonde Report (produced under
the aegis of Canadian Minister of National Health
and Welfare) that challenged the dominance of
the biomedical model and proposed that
the health field needed to consider biology, environment, lifestyle, and health-care organization,
a broad multidisciplinary team of Canadian
researchers led by the economist Robert Evans
began to ask: why are some people healthy and
others not? (Hancock 1986; Evans et al. 1994).
This question led not only to a consideration of
the crucial influence of upstream social and
6
behavioral determinants on individual and population health but also to a concern about how
early social environments can mold lifelong
health trajectories.
Leading this exploration of the developmental
role that upstream social factors on health and
development for the Evans-led team was Clyde
Hertzman. Hertzman went on to solidify his analysis about the importance of what at the time he
termed “biological embedding” through a series
of studies, analyses of other studies, and reinterpretations of existing literature through this new
life course health development lens. In addition
to publishing several important articles of conceptual synthesis, Hertzman and Daniel Keating
edited the volume Developmental Health and the
Wealth of Nations in which they unpacked the
impact of social gradients on health development
and began to specify how different time-specific
and pathway effects were at play early in development (Hertzman 1999; Keating and Hertzman
1999; Hertzman and Boyce 2010). They synthesized a wealth of evidence on how early experience affects a child’s brain development, social
and emotional functioning, and overall health
capacities (Hertzman 1994; Keating and
Hertzman 1999). The Evans and Hertzman work
in Canada emerged about the same time that the
Independent Inquiry into Inequalities in Health
Report (1998) led by Sir Donald Acheson was
released in the UK (Acheson 1998). This review –
whose panel of experts included David Barker,
Michael Marmot, and Hilary Graham, among
others – very clearly identified how many health
inequalities have their roots in the conditions and
experience of mothers and children, with impacts
that feedforward across the life-span.
Diana Kuh and Yoav Ben-Shlomo edited a
volume entitled A Life Course Approach to
Chronic Disease Epidemiology, first published in
1997, which coined the term “life course epidemiology” and presented for the first time a series
of articles that integrated the empirical research
on several specific types of disease from a life
course perspective (Kuh and Ben-Shlomo 1997).
This work was followed by a second edition in
2004 that updated each of the chapters on the life
course approach to obesity or the life course
N. Halfon et al.
approach to cardiovascular disease, and that
began to provide an overarching framework
including chapters on life course pathways to
adult health (Kuh and Ben-Shlomo 2004). In that
volume, there were chapters on “Life course
approaches to differentials in health” (Davey
Smith and Lynch 2004), “A life course approach
to obesity” (Gillman 2004), “Socioeconomic
pathways between childhood and adult health”
(Kuh et al. 2004), and “Should we intervene to
improve childhood circumstances” (Boyce and
Keating 2004). Kuh and Ben-Shlomo have gone
on to edit a series of books on life course chronic
disease epidemiology that continue to analyze
and synthesize the literature on health development from a life course perspective (Lawlor and
Mishra 2009; Kuh et al. 2013).
In 2000 (Halfon et al. 2000) and 2002 (Halfon
and Hochstein 2002), Halfon and colleagues
reviewed and synthesized several different life
course-focused research streams and suggested
that beyond its increasingly well-documented
importance for understanding the mechanisms
involved with the origins and development of
health and disease, this new life course approach
was of profound importance for a consideration
of how health care should be organized, financed,
and delivered. They also suggested that some
general principles were emerging and beginning
to outline a new model or framework that they
termed “life course health development.” At the
same time, many other scientists provided their
own synthesis of this emerging literature and
what they considered to be its implications for
health, health-care delivery, and health policy
(Ben-Shlomo and Kuh 2002; Halfon and
Hochstein 2002; Lu and Halfon 2003; Forrest
and Riley 2004; Worthman and Kuzara 2005).
These various attempts at synthesizing the evidence from this new field constituted a tipping
point, and over the next decade, the number of
empirical studies accelerated at a much faster
pace as the early objections to the “Barker
hypothesis” melted away in the wake of many
confirmatory studies, and the explanatory power
of this new conceptualization began to take hold.
This early LCHD synthesis highlighted the
linked importance of biological conditioning; the
Introduction to the Handbook of Life Course Health Development
role of multiple, ecologically nested risk, protective, and promoting factors in influencing health
trajectories; the developmental significance of
different time frames; and the evolution of different health development pathways in relationship
to particular socially, culturally, and biologically
specified transitions and turning points in an individual’s life (Halfon and Hochstein 2002). Over
the intervening decade, research has continued to
accelerate, advancing in the use of more sophisticated methodologies; employing new and rapidly
advancing epigenetic, genomic, and other techniques from systems biology; and, at the same
time, supporting and providing more concrete
evidence on behalf of these early summative concepts. These threads of scientific inquiry have
coalesced to form a network of research that has
produced a much more robust and integrated conceptual framework.
3
The Purpose, Structure,
and Content of This Volume
The absence of an integrative conceptual framework through which scientists could organize
and extend the manifold insights about the individual and contextual processes involved in the
development of health across the life course was
a fundamental challenge constraining the acceptance and understanding of a LCHD perspective
(Halfon et al. 2014; Hanson and Gluckman
2014). However, as noted above, empirical and
conceptual advances over the last 30 years have
crystallized in the generation, and growing
acceptance, of just such an integrative perspective. As a result, the life course health sciences
are rapidly maturing and progressing, and the life
course health development framework is coming
into clearer focus. Nevertheless, there remain a
number of challenges and growing pains that are
evident as scientists, clinicians, and public health
professionals from different fields attempt to
incorporate LCHD notions into already established areas of scholarship, practice, and policy
development.
For example, the definitions of terms are not
always clear, some terms are being redefined to
7
be integrated into this new approach, and other
concepts and constructs are adopted before their
meaning and significance are fully vetted and
analyzed. Good examples of these challenges
include terms like “sensitive and critical periods,”
“developmental programming,” “biological
embedding,” “trajectories,” and “pathways.”
Terms like “programming” have been criticized
as being too deterministic given the implication
that a certain experience or set of risk factors can
program a disease pathway; such a term eschews
the nature of developmental plasticity and the
phenotypic range that can emerge as the organism or individual encounters other experiences.
Hanson and Gluckman have suggested that a
term like priming, induction, or conditioning be
used to describe the process by which an exposure or experience induces a phenotypic alteration that prepares the organism for a similar
environmental challenge later in the life course
(Hanson and Gluckman 2014). Throughout this
volume, we have sought and encouraged the
authors to avoid terms like programming and biological embedding in favor of conditioning or
priming. Similarly, notions of critical periods
have been part of embryology for over a century,
and many biologists will refer to critical periods
in biological development as a specific time that
usually starts and ends abruptly and during which
a given event or its absence has a specific impact
on development. The experiments by Hubel and
Wiesel to examine the development of the visual
cortex seemed to indicate that there were critical
periods for specific complex neurons to develop
(Hubel and Wiesel 1977). However, most developmental science suggests that because of the
inherent plasticity in human development and in
many specific regulatory systems, the term “sensitive period” is less deterministic and therefore
much more appropriate.
In addition to the confusion around terminology, Hanson and Gluckman suggest several other
reasons for why the related concept of developmental origins of health and disease has faced
challenges, including confusion between factors
correlated with disease and those involved in
causation, the assumption that the only pathway
connecting early exposures and later disease was
N. Halfon et al.
8
low birth weight, a lack of plausible biological
mechanisms, a failure to recognize developmental origins under normal rather than under
extreme conditions, and the lack of evidence of
its relative importance in relationship to other
risk factors (Hanson and Gluckman 2014).
Whereas all of these challenges are important
and have begun to be addressed, the relative risk
issues have been some of the most daunting since
these questions require longitudinal data over
very long time horizons to tease out.
This volume is designed to address these challenges and bridge the resultant gaps, including
the delay in broad acceptance and understanding
of how health develops across the life-span
(Hanson and Gluckman 2014), as well as in the
translation of that perspective to health practice
and policy.
Each of the six sections comprising this volume was conceptualized during the process of
planning the LCRN’s 2013 MCH Life Course
Research Agenda-Setting Meeting, with the ultimate goal of informing the development of an
MCH Life Course Research Agenda (LCRA) that
would provide MCHB, the National Institutes of
Health (NIH), and other funding institutions and
organizations with guidance regarding priority
research questions and topics worthy of investment. With input from the LCRN advisory committee, project staff determined that the LCRA
(Sect. 6), which is in many ways the culmination
of the LCRN's 6 years of work to date, would
have to address theoretical concerns (Sect. I), specific periods of the life course (Sect. II), specific
health conditions (Sect. III) – some common and
some rare but nevertheless important because of
their severity or their representativeness of a set of
conditions sharing similar life course origins and/
or implications – cross-cutting topics in LCHD
(Sect. IV), and methodological issues (Sect. V) to
support researchers in carrying out the kinds of
studies the research agenda would call for.
Similarly, the majority of the chapter topics
contained in this volume were selected early on
in the development of the agenda-setting process. However, as noted in the Preface, several
chapters were commissioned following the
2013 agenda-setting meeting in response to
identified gaps, and others were selected based
on author interest in submitting a chapter for
inclusion in the agenda-setting process and/or
published volume. The editors acknowledge
that several important topics have not been
included in this edition of the volume. In some
cases, this omission was due to the difficulty of
identifying a qualified author/author team willing to develop a chapter on a given topic; in
other instances, chapters were commissioned
but not completed in time for inclusion in this
edition. It is the editors’ hope that future editions of this volume will address these worthy
topics through new chapters on topics ranging
from asthma and ADHD to family experiences,
mental health conditions, and LCHD measures
and biomarkers, among others. For now, however, it is our hope that readers will appreciate
the range of topics included in this edition and
the potential for these 26 chapters – all of which
synthesize existing LCHD research, identify
knowledge gaps, and/or recommend priorities
related to future research and efforts to ensure
the appropriate and timely translation of that
research into practice and policy – to have a significant impact on how LCHD stakeholders
think about, study, and work to promote health.
Chapters
“Middle
Childhood:
An
Evolutionary-Developmental Synthesis” and
“Pregnancy Characteristics and Women’s
Cardiovascular Health” contain modified versions of previously published reviews and analyses of existing research. Given the relatively
recent emergence of the field of LCHD and our
goal to further coalesce that field through the
publication of this volume, we thought it important to include the content in the handbook so that
the important research both chapters contain
could be situated firmly within the growing body
of LCHD research and have a meaningful influence on the content of the LCRA.
3.1
Section I: Emerging
Frameworks
This section contains a single chapter by pediatricians and public health experts Neal Halfon, MD,
MPH, and Christopher B. Forrest, MD, PhD,
entitled The Emerging Theoretical Framework of
Introduction to the Handbook of Life Course Health Development
Life Course Health Development (Halfon and
Forrest 2017). The authors – who also served as
two of the editors of the volume – propose a set
of principles that together form a more unified
theoretical framework for the emerging LCHD
field. These principles have the potential to guide
future theory building, research, and policy pertaining to optimizing health development in the
USA and abroad. Each subsequent chapter refers
to the principles when appropriate to demonstrate
how they can help to explain empirical findings
or set the stage for future inquiry.
3.2
Section II: Life Stages
Section II is comprised of six chapters exploring
health development from the preconceptional/
prenatal period to early adulthood. Each describes
the kinds of experiences and exposures that result
in more (or less) optimal outcomes during a given
developmental period and the importance of that
period for outcomes over the remainder of the
life course. As such, this section of the volume
grounds the literature reviewed about each life
stage firmly within the LCHD framework.
Preconception and Prenatal Factors and
Metabolic Risk – by pediatrician, MCH
researcher, and molecular epidemiologist
Xiaobin Wang, MD, MPH, ScD, and colleagues
Guoying Wang, MD, PhD, and Tami R. Bartell –
examines health during the earliest part of the
life course, reviewing what is known about the
mechanisms underlying both its sensitivity to
alterations in the intrauterine environment. The
authors explain the importance of this life period
for lifelong and transgenerational health, including links to obesity, diabetes, cardiovascular disease, behavioral and psychiatric disorders, and
more (Wang et al. 2017).
Early Childhood Health and the Life
Course: The State of the Science and Proposed
Research Priorities was authored by social epidemiologist and developmental-behavioral
pediatrician W. Thomas Boyce, MD, and the
late Clyde Hertzman, MD, MSc, who, as noted
in this introduction, played a central role in
delineating early childhood development as a
determinant of lifelong health. The chapter
9
reviews the literature regarding the susceptibility of young children to social environmental conditions, explains how variation among
individuals in terms of both their susceptibility
and their exposures helps to explain variation
in health development outcomes, and examines
the process by which early adversity becomes
biologically embedded (Boyce and Hertzman
2017).
Marco Del Giudice’s chapter on Middle
Childhood: An Evolutionary-Developmental
Synthesis demonstrates the unique significance of
middle childhood by examining – from an evolutionary perspective – the cognitive, behavioral,
and hormonal processes that characterize this life
stage, as well as its function as a switch point in
the adaptive development of life history strategies and the implications for life course health
development (DelGiudice 2017).
Adolescent Health Development: A Relational
Developmental Systems Perspective is the result
of collaboration among experts in developmental
psychology, human development, public health,
and pediatrics. The authors are Richard
M. Lerner, PhD; Claire C. Brindis, DrPH; Milena
Batanova, PhD; and Robert Wm. Blum, MD,
MPH, PhD. This chapter relates the seven proposed principles of LCHD to the Relational
Developmental Systems (RDS) metatheoretical
perspective, illustrating their interrelationships
and differences. The authors discuss the implications of both conceptual frameworks for studying
the life course origins and impact of adolescent
health and for promoting thriving during adolescence (Lerner et al. 2017).
Early Adulthood as a Critical Stage in the
Life Course was produced by a group of authors
with expertise in pediatrics, occupational therapy, psychology, and public health. In this chapter, David Wood, MD, MPH; Tara Crapnell,
ORD, OTR/L; Lynette Lau, PhD; Ashley
Bennett, MD; Debra Lotstein, MD, MPH; Maria
Ferris, MD, PhD, MPH; and Alice Kuo, MD,
PhD, employ an ecological approach to examine
the period of life from 20 to 30 years of age. The
authors also discuss the impact of chronic disease and other factors that affect the transition to
adulthood in the educational, employment, and
social arenas (Wood et al. 2017).
N. Halfon et al.
10
Epidemiologists Abigail Fraser, MPH, PhD,
MRC; Janet Catov, PhD; Debbie Lawlor, MRC;
and Janet Rich-Edwards’, ScD, MPH, chapter on
Pregnancy Characteristics and Women’s
Cardiovascular Health is a unique contribution
exploring the link between women’s reproductive
outcomes and their risk for cardiovascular disease. The chapter presents the implications for
research and policy including the potential to
identify high-risk women earlier in the life course
(Fraser et al. 2017).
3.3
Section III: The Life Course
Origins and Consequences
of Specific Diseases
and Health Conditions
Section III of this volume examines the life
course origins and consequences of some of the
most common diseases and health conditions facing the US population today, as well as some less
common but nevertheless important ones. Each
of the eight chapters reviews and synthesizes
prior basic and intervention research, points out
the gaps in our knowledge, and speaks to the
importance of additional research and its application to the policy and practice arenas. Together,
these chapters demonstrate the value of the
LCHD perspective in understanding and improving outcomes across a variety of populations and
challenges.
Social epidemiologist Summer Sherburne
Hawkins, PhD, MS, collaborated with pediatrician and prenatal nutrition expert Emily Oken,
MD, MPH and pediatrician and preventive cardiology expert Matthew W. Gillman, MD, SM, to
produce Early in the Life Course: Time for Obesity
Prevention. This chapter adopts a multilevel
approach in identifying the periods and factors
that are the greatest contributors to obesity and
explores how innovative research methodologies
can be used to demonstrate causality (Hawkins
et al. 2017).
Pediatric Type 2 Diabetes: Prevention and
Treatment Through a Life Course Health
Development Framework was written by Pamela
Salsberry, PhD, RN, FAAN; Rika Tanda, PhD,
RN; Sarah E. Anderson, PhD; and Manmohan
K. Kamboj, MD, an author team representing a
range of fields and disciplines including nursing,
pediatrics, public health, endocrinology, and epidemiology. Their chapter uses an LCHD perspective to promote a better understanding of the
development of pediatric T2DM, as well as a
more effective approach to prevention and treatment (Salsberry et al. 2017).
Clinical psychologist Irene E. Drmic, PhD,
CPsych, collaborated with psychiatric and
genetic epidemiologist Peter Szatmari, MD,
MSc, and Journal of Autism and Developmental
Disabilities editor-in-chief Fred Volkmar, MD,
MA, to produce Life Course Health Development
in Autism Spectrum Disorders. This chapter
applies the LCHD framework to autism spectrum
disorders (ASD) in order to inform future
research and ultimately improve health development for individuals with ASD, as well as their
families and communities (Drmic et al. 2017).
Self-Regulation was written by a large and
diverse group of experts spanning developmental, quantitative, and educational psychology.
Together, Megan McClelland, PhD; John
Geldof, PhD; Frederick J. Morrison; Steinunn
Gestsdóttir, PhD; Claire Cameron, PhD; Ed
Bowers, MEd, PhD; Angela Duckworth, PhD;
Todd Little, PhD; and Jennie Grammer, PhD,
examine the development and importance of
self-regulation through an LCHD lens and from
the standpoint of the relational developmental
systems (RDS) metatheoretical framework
(McClelland et al. 2017).
A Life Course Health Development
Perspective on Oral Health was written by James
J. Crall, DDS, ScD, and Christopher Forrest,
MD, PhD. By applying the LCHD framework
and their expertise in pediatric oral health and
public health, the authors advance a more contemporary conceptualization and definition of
oral health as a more integral and integrated component of overall health and well-being (Crall
and Forrest 2017).
Life Course Health Development Outcomes
After Prematurity: Developing a Community,
Clinical, and Translational Research Agenda to
Optimize Health, Behavior and Functioning is
Introduction to the Handbook of Life Course Health Development
the result of a collaboration among pediatrician
Michael E. Msall, MD, and a group of colleagues
representing nursing, pediatrics, sociology, and
demography, including Sarah A. Sobotka, MD,
MS; Amelia Dmowska; Dennis Hoga, PhD; and
Mary Sullivan, PhD, RN. Together the authors
examine the underlying causes of observed disparities in LCHD outcomes among children born
prematurely, the value of life course-focused
population-level interventions for closing the
gap, and the need to improve the availability and
systematic provision of such services (Msall
et al. 2017).
A Life Course Approach to Hearing Health
was written by Shirley A. Russ, MB, ChB,
MRCP, FRACP, MCH; Kelly Tremblay, PhD,
CCC-A; Neal Halfon, MD, MPH; and Adrian
Davis, BSC, MSc, PhD, FFPH, FSS, OBE – an
author team representing both pediatrics and
speech and hearing. The chapter explores the origins of the full spectrum of hearing loss, including genetic, congenital, and environmental
causes, and the mechanisms by which they interact and act upon an individual’s hearing health
development over the life-span (Russ et al. 2017).
Nephrologists Patrick Brophy, MD; Jennifer
R. Charlton, MD, MSc; J. Bryan Carmody, MD;
Kimberly J. Reidy, MD; David Askenazi, MD;
and Susan P. Bagby, MD, teamed up with pediatrician Lindsay Harshman, MD; neonatologist
Jeffrey Segar, MD; and public health expert
David Shoham, PhD, MSPH, to produce Chronic
Kidney Disease: A Life Course Health
Development Perspective. In this chapter, the
authors find that the LCHD framework is of great
value in both elucidating the sequelae of CKD
risk and identifying the kinds of early life interventions that have the potential to mitigate it
(Brophy et al. 2017).
3.4
Section IV: Cross-Cutting
Topics in Life Course Health
Development
This section of the volume addresses four key
issues – growth, nutrition, adversity, and disparities – that are relevant to understanding and
11
addressing a wide range of diseases and health
conditions.
Growth and Life Course Health Development
was written by a team of authors spanning medicine, public health, and anthropology. In their
chapter, Amanda Mummert, MA, Meriah
Schoen, and Michelle Lampl, MD, PhD, employ
a systems biology approach to examine the
pathways affecting growth and explore auxology’s role in a variety of health trends (Mummert
et al. 2017).
From Epidemiology to Epigenetics: Evidence
for the Importance of Nutrition to Optimal
Health Development Across the Life Course was
written by nutrition experts Marion Taylor-Baer,
PhD, MSNS, RD, and Dena Herman, MSNS,
MPH, RD. Their chapter uses the LCHD framework to examine the crucial role that nutrition
plays in the development of health potential over
the life-span, including the role of evolutionarily
driven adaptive responses during the prenatal
and early childhood periods (Taylor-Baer and
Herman 2017).
In How Socioeconomic Disadvantages Get
Under the Skin and into the Brain to Influence
Health Development Across the Lifespan, developmental psychologists Pilyoung Kim, PhD;
Gary Evans, PhD; Edith Chen, PhD; and Gregory
Miller, PhD, collaborated with epidemiologist
Teresa Seeman, PhD, to explain the neurobiological mechanisms and processes by which
SES-related adversity, including chronic stress,
affect health trajectories from early life to old age
(Kim et al. 2017).
Health sciences researcher Kandyce Larson,
PhD, together with pediatricians Shirley A. Russ,
MD, MPH; Robert S. Kahn, MD, MPH; Glenn
Flores, MD, FAAP; Elizabeth Goodman, MD;
Tina L. Cheng, MD, MPH; and Neal Halfon,
MD, MPH, produced Health Disparities: A Life
Course Health Development Perspective and
Future Research Directions. This chapter
explores the factors and processes that contribute
to health disparities across lifetimes and generations from the perspective of life course health
development in order to illuminate potential
practice and policy solutions to this persistent
problem (Larson et al. 2017).
N. Halfon et al.
12
3.5
Section V: Methodological
Approaches
Section V is comprised of five chapters that will
be of practical use to researchers who are engaged
or who would like to engage in future research
aimed at enhancing our understanding of how
health develops over the life course and/or understanding how practice and policy can optimize
health development outcomes. The first two
chapters in this section review current research
methods and study designs that are of particular
value for LCHD research. The remaining three
chapters describe existing longitudinal datasets
that have the potential to be used to answer the
kinds of research questions that are described in
the Life Course Research Agenda, Version 1.0
(Halfon et al. 2017).
In Core Principles of Life Course Health
Development Methodology and Analytics, developmental and educational psychologist and
statistician-methodologist Todd Little, PhD,
acknowledges the inherent complexity of LCHD
studies. He suggests that underutilized techniques, such as structural equation modeling,
multilevel modeling, and mixture distribution
modeling, as well as new and collaborative teambased research practices, have the potential to
rapidly advance the field (Little 2017).
In Epidemiological Study Designs: Traditional
and Novel Approaches to Advance Life Course
Health Development Research, epidemiologists
Stephen L. Buka, PhD; Samantha R. Rosenthal,
PhD; and Mary E. Lacy explore the benefits and
limitations of the various approaches that can be
used to study the development of health and disease over the life course (Buka et al. 2017).
Using the National Longitudinal Surveys of
Youth (NLSY) to Conduct Life Course Analyses
by sociologist Elizabeth Cooksey, PhD, demonstrates how this long-running three-cohort longitudinal study – which provides a wealth of data
on health, education, employment, household
information, family background, marital history,
child care, income and assets, attitudes, substance use, and criminal activity – can be used to
explore various LCHD-related research questions (Cooksey 2017).
Sociologist and demographer Narayan Sastry,
PhD, worked with research scientists Paula
Fomby, PhD, and Katherine McGonagle, PhD,
to develop the chapter on Using the Panel Study
of Income Dynamics (PSID) to Conduct Life
Course Health Development Analysis. This
chapter explains how this nationally representative longitudinal study – which is the longestrunning household panel study in the world,
covering 47 years of data on a wide range of economic, demographic, social, and health topics –
can be used to examine health development over
the life course (Sastry et al. 2017).
In Using the Fragile Families and Child
Wellbeing Study (FFCWS) in Life Course Health
Development Research, sociologist Amanda
Geller, PhD, collaborates with FFCWS staff
members Kate Jaeger and Garrett Pace to
describe this nationally representative birth
cohort study that contains both biological and
social indicators. The authors provide examples
of its use for exploring questions about health
development in households with unmarried parents (Geller et al. 2017).
3.6
Section VI: Conclusions
Section VI of this volume contains a single chapter entitled Life Course Research Agenda (LCRA),
Version 1.0 in which editors Neal Halfon, MD,
MPH; Christopher B. Forrest, MD, PhD; Richard
M. Lerner, PhD; and Elaine Faustman, PhD,
together with LCRN staff members Ericka Tullis,
MPP, and John Son, MPH, synthesize the recommendations for future research contained in many
of the previous chapters and propose a set of priority research types, topics, and questions, as
well as a set of activities aimed at improving our
ability to carry out this critical research and
ensure its timely translation to practice and policy. The authors also recommend strategies that
can support the ongoing refinement of the LCHD
theoretical framework (Halfon et al. 2017). As
this volume goes to press, the LCRN is initiating
an inclusive process to review and refine this initial version of the LCRA so that it is sure to guide
both researchers and potential funders toward the
Introduction to the Handbook of Life Course Health Development
studies that will be of greatest benefit in terms of
enhancing our rapidly growing but as yet incomplete understanding of life course health
development.
References
Acheson, D. (1998). Independent inquiry into inequalities
in health report. Stationery Office.
Aldwin, C. M. (2014). Rethinking developmental science.
Research in Human Development, 11(4), 247–254.
Antony, P. M., Balling, R., & Vlassis, N. (2012). From
systems biology to systems biomedicine. Current
Opinion in Biotechnology, 23(4), 604–608.
Baltes, P. B., Lindenberger, U., & Staudinger, U. M.
(2006). Lifespan theory in developmental psychology.
In W. Damon & R. M. Lerner (Eds.), Handbook of
child psychology. Vol. 1: Theoretical models of human
development (6th ed., pp. 569–664). New York: Wiley.
Barker, D. J. (1995). Fetal origins of coronary heart disease. BMJ, 311(6998), 171.
Barker, D. J., Osmond, C., Winter, P. D., Margetts, B., &
Simmonds, S. J. (1989). Weight in infancy and death
from ischaemic heart disease. The Lancet, 334(8663),
577–580.
Barker, D. J., Godfrey, K. M., Gluckman, P. D., Harding,
J. E., Owens, J. A., & Robinson, J. S. (1993). Fetal
nutrition and cardiovascular disease in adult life. The
Lancet, 341(8850), 938–941.
Ben-Shlomo, B., & Kuh, D. (2002). A life course approach
to chronic disease epidemiology: Conceptual models,
empirical challenges and interdisciplinary perspectives. International Journal of Epidemiology, 31(2),
285–293.
Berkman, L. F., Kawachi, I., & Glymour, M. M. (Eds.).
(2014). Social epidemiology. Oxford: Oxford
University Press.
Braveman, P. (2014). What is health equity: And how
does a life-course approach take us further toward it?
Maternal and Child Health Journal, 18(2), 366–372.
Bronfenbrenner, U. (2005). Making human beings human.
Thousand Oaks: Sage.
Boyce, T., & Hertzman, C. (2017). Early childhood and
the life course: The state of the science and proposed
research priorities. In N. Halfon, C. B. Forrest, R. M.
Lerner, & E. Faustman (Eds.), Handbook of life course
health development. New York: Springer.
Boyce, T. W., & Keating, D. P. (2004). Should we intervene to improve childhood circumstances? In D. Kuh
& Y. Ben-Shlomo (Eds.), A life course approach to
chronic disease epidemiology (Vol. 2). New York:
Oxford University Press.
Brophy, P. D., Charlton, J. R., Carmody, J. B., Reidy, K. J.,
Harshman, L., Segar, J., Askenazi, D., & Shoham, D.
(2017). Chronic kidney disease: A life course healthdevelopment perspective. In N. Halfon, C. B. Forrest,
13
R. M. Lerner, & E. Faustman (Eds.), Handbook of life
course health development. New York: Springer.
Buka, S. L., Rosenthal, S. R., & Lacy, M. E. (2017).
Epidemiological study designs: Traditional and novel
approaches to advance life course health-development
research. In N. Halfon, C. B. Forrest, R. M. Lerner,
& E. Faustman (Eds.), Handbook of life course health
development. New York: Springer.
Capra, F. (1982). The turning point: Science, society, and
the rising culture. Toronto: Bantam Books.
Committee on Future Directions for Behavioral and
Social Sciences Research., Singer B. H., Ryff, C. D
(Eds.). (2001). New horizons in health: An integrative approach. Washington, DC: National Academies
Press.
Committee on Evaluation of Children’s Health; Board on
Children, Youth and Families; Division of Behavioral
and Social Sciences and Education; National Research
Council; Institute of Medicine. (2004). Children’s
health, the nation’s wealth: Assessing and improving
child health. Washington, DC: National Academies
Press.
Committee on the Recommended Social and Behavioral
Domains and Measures for Electronic Health Records,
Board on Population Health and Public Health
Practice, Institute of Medicine. (2015). Capturing
social and behavioral domains and measures in
electronic health records: Phase 2. Washington, DC:
National Academies Press (US).
Cooksey, E. (2017). Using the National Longitudinal
Surveys of Youth (NLSY) to conduct life course
analyses. In N. Halfon, C. B. Forrest, R. M. Lerner,
& E. Faustman (Eds.), Handbook of life course health
development. New York: Springer.
Crall, J. J., & Forrest, C. B. (2017). A life course healthdevelopment perspective on oral health. In N. Halfon,
C. B. Forrest, R. M. Lerner, & E. Faustman (Eds.),
Handbook of life course health development. New
York: Springer.
Davey Smith, G., & Lynch, J. (2004). Life course
approaches to socioeconomic differentials in health.
In D. Kuh & Y. Ben-Shlomo (Eds.), A life course
approach to chronic disease epidemiology (Vol. 2).
New York: Oxford University Press.
Davila-Velderrain, J., Martinez-Garcia, J. C., & AlvarezBuylla, E. R. (2015). Modeling the epigenetic attractors landscape: Toward a post-genomic mechanistic
understanding of development. Frontiers in Genetics,
6, 160.
DelGiudice, M. (2017). Middle childhood: An
evolutionary-developmental synthesis. In N. Halfon,
C. B. Forrest, R. M. Lerner, & E. Faustman (Eds.),
Handbook of life course health development. New
York: Springer.
Drmic, I. E., Szatmari, P., & Volkmar, F. (2017). Life
course health-development in autism spectrum disorders. In N. Halfon, C. B. Forrest, R. M. Lerner, &
E. Faustman (Eds.), Handbook of life course health
development. New York: Springer.
14
Elder, G. H., Shanahan, M. J., & Jennings, J. A. (2015).
Human development in time and place. In M. H.
Bornstein& T. Leventhal (Eds.), Handbook of
child psychology and developmental science. Vol.
4: Ecological settings and processes in developmental systems (7th ed., pp. 6–54). Editor-in-chief:
R.M. Lerner. Hoboken: Wiley.
Evans, R. G., Barer, M. L., & Marmor, T. R. (Eds.).
(1994). Why are some people healthy and others not?:
The determinants of health of populations. New York:
Aldine de Gruyter, 1994.
Ford, D. L., & Lerner, R. M. (Eds.). (1992). Developmental
systems theory: An integrative approach. Newbury
Park: Sage.
Forrest, C. B., & Riley, A. W. (2004). Childhood origins
of adult health: A basis for life-course health policy.
Health Affairs (Millwood), 23(5), 155–164.
Foster, K. R. (2011). The sociobiology of molecular systems. Nature Reviews Genetics, 12, 193–203.
Fraser, A., Catov, J. M., Lawlor, D. A., Rich-Edwards,
J. W. (2017). Pregnancy characteristics and women’s
cardiovascular health. In N. Halfon, C. B. Forrest,
R. M. Lerner, E. Faustman (Eds.), Handbook of life
course health development. New York: Springer.
Gee, G. C., Walsemann, K. M., & Brondolo, E. (2012). A
life course perspective on how racism may be related
to health inequities. American Journal of Public
Health, 102(5), 967–974.
Geller, A., Jaeger, K., & Pace, G. (2017). Using the fragile
families and child wellbeing study (FFCWS) in life course
health-development research. In N. Halfon, C. B. Forrest,
R. M. Lerner, & E. Faustman (Eds.), Handbook of life
course health development. New York: Springer.
Gillman, M. W. (2004). A life course approach to obesity. In D. Kuh & Y. Ben-Shlomo (Eds.), A life course
approach to chronic disease epidemiology (Vol. 2).
Oxford University Press.
Gluckman, P. D., & Hanson, M. A. (2004). Living with
the past: Evolution, development, and patterns of disease. Science, 305(5691), 1733–1736.
Gluckman, P. D., Hanson, M. A., Cooper, C., & Thornburg,
K. L. (2008). Effect of in utero and early-life conditions on adult health and disease. New England
Journal of Medicine, 359(1), 61–73.
Gottlieb, G. (1998). Normally occurring environmental
and behavioral influences on gene activity: From central dogma to probabilistic epigenesist. Psychological
Review, 105, 792–802.
Greenblum, S., Turnbaugh, P. J., & Borenstein, E.
(2012). Metagenomic systems biology of the human
gut microbiome reveals topological shifts associated with obesity and inflammatory bowel disease.
Proceedings of the National Academy of Sciences,
109(2), 594–599.
Halfon, N., & Forrest, C. B. (2017). The emerging theoretical framework of life course health-development. In
N. Halfon, C. B. Forrest, R. M. Lerner, & E. Faustman
(Eds.), Handbook of life course health development.
New York: Springer.
N. Halfon et al.
Halfon, N., Forrest, C. B., Lerner, R. M., Faustman, E.,
Tullis, E., & Son, J. (2017). Life course research agenda
(LCRA), version 1.0. In N. Halfon, C. B. Forrest, R. M.
Lerner, & E. Faustman (Eds.), Handbook of life course
health development. New York: Springer.
Halfon, N., & Hochstein, M. (2002). Life course health
development: An integrated framework for developing
health, policy and research. The Milbank Quarterly,
80(3), 433–479.
Halfon, N., Inkelas, M., & Hochstein, M. (2000). The
health development organization: An organizational
approach to achieving child health development. The
Milbank Quarterly, 78(3), 447–497.
Halfon, N., Larson, K., Lu, M., Tullis, E., & Russ, S.
(2014). Lifecourse health development: Past, present
and future. Maternal and Child Health Journal, 18(2),
344–365.
Hancock, T. (1986). Lalonde and beyond: Looking back
at “a new perspective on the health of Canadians”.
Health Promotion International, 1(1), 93–100.
Hanson, M. A., & Gluckman, P. D. (2014). Early developmental conditioning of later health and disease:
Physiology or pathophysiology? Physiological
Reviews, 94(4), 1027–1076.
Hawkins, S. S., Oken, E., Gillman, M. W. (2017). Early
in the life course: Time for obesity prevention. In
N. Halfon, C. B. Forrest, R. M. Lerner, E. Faustman
(Eds.), Handbook of life course health development.
New York: Springer.
Hertzman, C. (1994). The lifelong impact of childhood
experiences: A population health perspective. Daedalus,
123(4), 167–180.
Hertzman, C. (1999). The biological embedding of early
experience and its effects on health in adulthood.
Annals of the New York Academy of Sciences, 896(1),
85–95.
Hertzman, C., & Boyce, T. (2010). How experience gets
under the skin to create gradients in developmental
health. Annual Review of Public Health, 31, 329–347.
Huang, S. (2012). The molecular and mathematical basis
of Waddington’s epigenetic landscape: A framework for post-Darwinian biology? BioEssays, 34(2),
149–157.
Hubel, D. H., & Wiesel, T. N. (1977). Ferrier lecture:
Functional architecture of macaque monkey visual
cortex. Proceedings of the Royal Society of London B:
Biological Sciences, 198(1130), 1–59.
Kandel, E. R., Dudai, Y., & Mayford, M. R. (2014). The
molecular and systems biology of memory. Cell,
157(1), 163–186.
Keating, D. P., & Hertzman, C. (Eds.). (1999).
Developmental health and the wealth of nations:
Social, biological, and educational dynamics.
New York: Guilford Press.
Kim, P., Evans, G. W., Chen, E., Miller, G., & Seeman,
T. (2017). How socioeconomic disadvantages get
under the skin and into the brain to influence healthdevelopment across the lifespan. In N. Halfon,
C. B. Forrest, R. M. Lerner, & E. Faustman (Eds.),
Introduction to the Handbook of Life Course Health Development
Handbook of life course health development. New
York: Springer.
Kitano, H. (2002). Systems biology: A brief overview.
Science, 295(5560), 1662–1664.
Kuh, D., & Ben-Shlomo, Y. (Eds.). (1997). A life course
approach to chronic disease epidemiology (Vol. 1).
Oxford: Oxford University Press.
Kuh, D., & Ben-Shlomo, Y. (Eds.). (2004). A life course
approach to chronic disease epidemiology (Vol. 2).
New York: Oxford University Press.
Kuh, D., Cooper, R., Hardy, R., Richards, M., & BenShlomo, Y. (Eds.). (2013). A life course approach to
healthy ageing. Oxford: Oxford University Press.
Kuh, D., Power, C., Blane, D., & Bartley, M. (2004).
Socioeconomic pathways between childhood and
adult health. In D. Kuh & Y. Ben-Shlomo (Eds.), A
life course approach to chronic disease epidemiology
(Vol. 2). New York: Oxford University Press.
Lappé, M., & Landecker, H. (2015). How the genome
got a life span. New Genetics and Society., 34(2),
152–176.
Larson, K., Russ, S. A., Kahn, R. S., Flores, G., Goodman,
E., Cheng, T. L., & Halfon, N. (2017). Health disparities: A life course health-development perspective and
future research directions. In N. Halfon, C. B. Forrest,
R. M. Lerner, & E. Faustman (Eds.), Handbook of
life course health development. New York: Springer.
Lawlor, D. A., & Mishra, G. D. (Eds.). (2009). Family
matters: Designing, analysing and understanding
family based studies in life course epidemiology.
Oxford: Oxford University Press.
Lerner, R. M. (1984). On the nature of human plasticity.
New York: Cambridge University Press.
Lerner, R. M. (2006). Developmental science, developmental systems, and contemporary theories. In
R. M. Lerner (Ed.), Theoretical models of human
development. Volume 1 of Handbook of child psychology (6th ed.). Editors-in-chief: Damon W & Lerner
RM. Hoboken: Wiley.
Lerner, R. M. (2012). Developmental science: Past, present and future. International Journal of Developmental
Science., 6, 29–36.
Lerner, R. M., & Callina, K. S. (2014). The study of character development: Towards tests of a relational developmental systems model. Human Development, 57(6),
322–346.
Lerner, R. M., & Overton, W. F. (2008). Exemplifying the
integrations of the relational developmental system:
Synthesizing theory, research, and application to promote positive development and social justice. Journal
of Adolescent Research, 23, 245–255.
Lerner, R. M., Brindis, C. C., Batanova, M., & Blum, R. W.
(2017). Adolescent health: A relational developmental
systems perspective. In N. Halfon, C. B. Forrest, R. M.
Lerner, & E. Faustman (Eds.), Handbook of life course
health development. New York: Springer.
Little, T. D. (2017). Core principles of life course
health-development methodology and analytics. In
N. Halfon, C. B. Forrest, R. M. Lerner, & E. Faustman
15
(Eds.), Handbook of life course health development.
New York: Springer.
Lu, M., & Halfon, N. (2003). Racial and ethnic disparities
in birth outcomes: A life-course perspective. Maternal
and Child Health Journal, 7(1), 13–30.
McClelland, M., Morrison, F., Gestsdóttir, S., Cameron,
C., Bowers, E., Duckworth, A., Little, T., & Grammer,
J. (2017). Self-regulation. In N. Halfon, C. B. Forrest,
R. M. Lerner, & E. Faustman (Eds.), Handbook of life
course health development. New York: Springer.
Molenaar, P. C., & Newell, K. M. (Eds.). (2010).
Individual pathways of change: Statistical models
for analyzing learning and development. Washington,
DC: American Psychological Association.
Msall, M. E., Sobotka, S. A., Dmowska, A., Hoga, D., &
Sullivan, M. (2017). Life-course health-development
outcomes after prematurity: Developing a community,
clinical, and translational research agenda to optimize health, behavior and functioning. In N. Halfon,
C. B. Forrest, R. M. Lerner, & E. Faustman (Eds.),
Handbook of life course health development. New
York: Springer.
Mummert, A., Schoen, M., & Lampl, M. (2017). Growth
and life course health-development. In N. Halfon, C. B.
Forrest, R. M. Lerner, & E. Faustman (Eds.), Handbook
of life course health development. Springer.
Overton, W. F. (2010). Life-span development: Concepts
and issues. In W. F. Overton (Ed.)., Lerner RM (Editorin-Chief) Handbook of life-span development, Vol. 1:
Cognition, biology, and methods across the lifespan
(pp. 1–29). Hoboken: Wiley.
Overton, W. F. (2012). Evolving scientific paradigms:
Retrospective and prospective. In L. L’Abate (Ed.), The
role of paradigms in theory construction (pp. 31–65).
New York: Springer.
Overton, W. F. (2015). Process and relational developmental systems. In W. F. Overton, & P. C. Molenaar
(Eds.), Theory and method. Volume 1 of the Handbook
of child psychology and developmental science (7th
ed., pp. 9–62) Editor-in-chief: R. M. Lerner. Hoboken:
Wiley.
Piro, R. M., & Di Cunto, F. (2012). Computational
approaches to disease-gene prediction: Rationale,
classification and successes. The FEBS Journal,
279(5), 678–696.
Russ, S. A., Tremblay, K., Halfon, N., & Davis, A. (2017).
A lifecourse approach to hearing health. In N. Halfon,
C. B. Forrest, R. M. Lerner, & E. Faustman (Eds.),
Handbook of life course health development. New
York: Springer.
Salsberry, P., Tanda, R., Anderson, S. E., & Kamboj, M. K.
(2017). Pediatric type 2 diabetes: Prevention and treatment through a life course health-development framework. In N. Halfon, C. B. Forrest, R. M. Lerner, &
E. Faustman (Eds.), Handbook of life course health
development. New York: Springer.
Sameroff, A. (2010). A unified theory of development:
A dialectic integration of nature and nurture. Child
Development, 81(1), 6–22.
16
Sastry, N., Fomby, P., & McGonagle, K. (2017). Using
the panel study of income dynamics (PSID) to conduct
life course health-development analysis. In N. Halfon,
C. B. Forrest, R. M. Lerner, & E. Faustman (Eds.),
Handbook of life course health development. New
York: Springer.
Schadt, E. E., & Björkegren, J. L. (2012). NEW: Networkenabled wisdom in biology, medicine, and health care.
Science Translational Medicine, 4(115), 115rv1.
Taylor-Baer, M., & Herman, D. (2017). From epidemiology to epigenetics: Evidence for the importance of
nutrition to optimal health-development across the
lifecourse. In N. Halfon, C. B. Forrest, R. M. Lerner,
& E. Faustman (Eds.), Handbook of life course health
development. New York: Springer.
von Bertalanffy, L. (1933). Modern theories of development: An introduction to theoretical biology. London:
Oxford University Press.
N. Halfon et al.
Wang, G., Bartell, T. R., & Wang, X. (2017). Preconception
and prenatal factors and metabolic risk. In N. Halfon,
C. B. Forrest, R. M. Lerner, & E. Faustman (Eds.),
Handbook of life course health development. New
York: Springer.
Whitehead, A. N. (1929/1978). Process and reality: An
essay in cosmology. New York: The Free Press.
Wood, D., Crapnell, T., Lau, L., Bennett, A., Lotstein,
D., Ferris, M., & Kuo, A. (2017). Emerging adulthood as a critical state in the life course. In N. Halfon,
C. B. Forrest, R. M. Lerner, & E. Faustman (Eds.),
Handbook of life course health development. New
York: Springer.
Worthman, C. M., & Kuzara, J. (2005). Life history and
the early origins of health differentials. American
Journal of Human Biology, 17(1), 95–112.
Open Access This chapter is licensed under the terms of the Creative Commons Attribution 4.0 International License
(http://creativecommons.org/licenses/by/4.0/), which permits use, sharing, adaptation, distribution and reproduction in
any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license and indicate if changes were made.
The images or other third party material in this chapter are included in the chapter’s Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the chapter’s Creative Commons license
and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain
permission directly from the copyright holder.
Part I
Emerging Frameworks
The Emerging Theoretical
Framework of Life Course Health
Development
Neal Halfon and Christopher B. Forrest
1
Part 1: Context
and Background
Over the last century, we have witnessed the
power of the biomedical paradigm to treat the
sick and prevent diseases from occurring.
Conventional health science, as an applied field,
has tried to make sense of what constitutes health
by exploring the causes of disease in individual
patients (medicine) and populations (public
health). This approach has created a perspective
of health as absence of disease or its risk factors
and has been unsuccessful at explaining what it
means to be healthy, how health develops over
the lifespan, and the impact of health on the lives
of individuals.
N. Halfon, MD, MPH (*)
Department of Pediatrics, David Geffen School of
Medicine, UCLA, Los Angeles, CA, USA
Department of Health Policy and Management,
Fielding School of Public Health, UCLA,
Los Angeles, CA, USA
Department of Public Policy, Luskin School of Public
Affairs, UCLA, Los Angeles, CA, USA
Center for Healthier Children, Families, and
Communities, UCLA, Los Angeles, CA, USA
e-mail: nhalfon@ucla.edu
C.B. Forrest, MD, PhD
Applied Clinical Research Center, Children’s
Hospital of Philadelphia, Philadelphia, PA, USA
Concepts of what constitutes health, and theories about how health is produced and optimized,
are constantly evolving in response to myriad
social and cultural expectations shaped by our
contemporary worldview, scientific advances,
improvements in health interventions, and the
changing capacity of the health system. Stimulated
originally by a series of studies demonstrating
how growth during early life is related to chronic
health conditions that emerge many decades later,
an eruption of new research is identifying developmental processes that shape long-term health
trajectories (Ben-Shlomo and Kuh 2002; Kuh and
Ben-Shlomo 2004; Hanson and Gluckman 2014).
This research is demonstrating how complex
developmental processes integrate a range of
behavioral, social, and environmental influences
that modify gene expression, modulate physiologic and behavioral function, and dynamically
shape different pathways of health production
(Halfon and Hochstein 2002; Kuh and BenShlomo 2004; Halfon et al. 2014). These empirical findings are highlighting the limitations of the
more mechanistic biomedical and biopsychosocial models of health, which fail to offer comprehensive explanations about such phenomena as
the developmental origins of health, how stress
affects current and future health, and the consequences of dynamic interactions between individuals and their environments over time.
Informed by new theoretical perspectives emerging from such fields of study as developmental
© The Author(s) 2018
N. Halfon et al. (eds.), Handbook of Life Course Health Development,
DOI 10.1007/978-3-319-47143-3_2
19
20
psychology (Lerner 2012), systems biology (Kitano
2002; Antony et al. 2012; Schadt and Bjorkegren
2012), epigenetics (Egger et al. 2004), the developmental origins of chronic disease (Gluckman and
Hanson 2006a), and evolutionary developmental
biology (West-Eberhard 2003), a transdisciplinary
framework (Gatzweller and Baumuller 2013) is
emerging which we call Life Course Health
Development (LCHD) (Halfon and Hochstein 2002,
Halfon et al. 2014). As a framework, LCHD organizes several different theories and conceptual models in order to make sense of the enormously
challenging question of how health develops over
the lifespan.
The LCHD framework addresses the developmental origins of health, the role that biological
and behavioral plasticity play in facilitating different levels of adaptation, and how mismatches
between biological propensity and environmental
context interact to produce breakdowns in health,
known as disease. As a framework that organizes
numerous theories and concepts related to how
health develops, LCHD is bridging what have
sometimes been assumed to be polar opposites:
nature versus nurture, mind versus body, individual versus population, and short-term versus
long-term change. By unifying these dichotomies, LCHD offers a new perspective that will
guide future scientific inquiry on health development and facilitate a long overdue and needed
synthesis of medicine and public health—a synthesis that links treatment, prevention, and health
promotion and catalyzes more integrated and networked strategies for designing, organizing, and
implementing multilevel health interventions that
transcend individual and population dichotomies.
The LCHD framework will be increasingly useful as the human ecological footprint expands
and influences the health development of Earth
itself, creating new threats to human health via
rapid and disruptive changes in physical environments, geographic dispersion of populations, and
changes in social development.
This emergence of LCHD is reflective of larger
scientific trends that are transforming research in
the physical, natural, and social sciences. The
comfort and certainty of simple, linear, and deterministic causal pathways are giving way to the
N. Halfon and C.B. Forrest
uncomfortable uncertainty of nonlinear causal
clusters that are networked together into complex,
multilevel, interactive, and relational systems.
LCHD embraces this complexity as the salient
target of inquiry and requires research to be conducted with teams that are multidisciplinary, often
large, networked, and highly collaborative. These
shifts in scientific approach are helping us understand how our modern interdependent world is
organized, how it functions, and how it contributes to the production of human health.
Given the explosion of life course-focused
research in many scientific fields—including
chronic disease epidemiology, developmental
neuroscience, developmental psychology, evolutionary biology, genetics, epigenetics, environmental health sciences, economics, sociology,
and many more—there is a growing need to provide a systematic framework for understanding
and organizing this emerging knowledge base
(i.e., sense making) so that it can be more effectively applied to solving health problems and
guiding new and productive streams of exploration and discovery.
Our intent is not to provide a grand theory of
Life Course Health Development. Rather, we
seek to establish a set of principles that describe
the contours of the rapidly emerging health development knowledge base by organizing many
theories and conceptual models into a coherent
synthesis. We recognize that LCHD is a work in
progress. Our aim is to create conceptual coherence by contextualizing the meaning of disparate
research findings, identifying gaps and uncertainties—including how concepts are defined, operationalized, and interpreted—and moving inquiry,
application, and implementation forward. We
hope that the principles of LCHD presented here,
coupled with our explanatory narrative, will
encourage theory building and testing, inspire
innovative transdisciplinary research, and mature
the framework into a scientific model with
descriptive, explanatory, and predictive utility.
Furthermore, we hope that LCHD will shine a
light on the conundrum of how little attributable
risk is explained in many studies of chronic disease, how early experience conditions future biological response patterns, and how these early
The Emerging Theoretical Framework of Life Course Health Development
experiences play through complex, environmentally influenced, and developmentally plastic
health development pathways (Table 1).
In Part 2 of this chapter, we describe the ontological pathways—including theories, empirical
findings, and concepts—that led to the LCHD
framework, thereby orienting our view of health
development from simple, mechanistic, and
reductionist models to contemporary models that
are holistic, complex, dynamically relational, and
adaptive. In Part 3, we summarize the principles
of the LCHD framework, grounding them in a set
of transdisciplinary theories, models, and perspectives and addressing their implications for
future inquiry (Fig. 1).
Beginning with the simple, mechanical, and
mostly linear biomedical model, we chart how it
was transformed into a more hierarchical,
Table 1 Principles of
Development Framework
Principle
1. Health
Development
2. Unfolding
3. Complexity
4. Timing
5. Plasticity
6. Thriving
7. Harmony
the
Life
Course
Health
Brief description
Health development integrates the
concepts of health and developmental
processes into a unified whole
Health development unfolds
continuously over the lifespan, from
conception to death, and is shaped by
prior experiences and environmental
interactions
Health development results from
adaptive, multilevel, and reciprocal
interactions between individuals and
their physical, natural, and social
environments
Health development is sensitive to
the timing and social structuring of
environmental exposures and
experiences
Health development phenotypes are
systematically malleable and enabled
and constrained by evolution to
enhance adaptability to diverse
environments
Optimal health development
promotes survival, enhances
well-being, and protects against
disease
Health development results from the
balanced interactions of molecular,
physiological, behavioral, cultural,
and evolutionary processes
21
dynamic, and multiply determined biopsychosocial model, as the result of scientific breakthroughs in the understanding of the contribution
of behavioral, social factors and their influence
on individuals during specific life stages. This
biopsychosocial model has now evolved into a
more complex, relational, adaptive, dynamic, and
developmental model of Life Course Health
Development (LCHD) as result of the influence
of scientific breakthroughs in epigenetics, neurodevelopment, and life course chronic disease
epidemiology.
2
Part 2: Emergence of the Life
Course Health Development
Framework
The LCHD framework has emerged from a network of theories, conceptual models, and empirical findings and provides a more comprehensive
description of how health develops over the life
course than any single component part. In this
section, we describe the streams of scientific
inquiry, the key theories and models, and the
seminal scientific insights that are brought
together by the LCHD framework. Figure 1 provides an epistemological schematic, charting the
changing paradigms of health and how different
streams of research and their findings influenced
the flow of conceptual models.
A few decades ago, research linked fetal
development with degenerative diseases of old
age (Ben-Shlomo and Kuh 2002), stimulating
new ways of thinking about the mechanisms
underlying what was originally termed “fetal
programming” and other environmentally
induced modifications in gene expression that
presumably take place early in life. These insights
pointed out the need to better characterize interactions between genes and the environment, to
better understand gene regulation that occurs in
response to environmental signal transduction,
and to better integrate into explanatory models
the importance of the timing and phasing of these
developmental processes. The prevailing epidemiological framework, with its simple additive,
exposure-response models of risk accumulation
N. Halfon and C.B. Forrest
22
Simple,
Reductionistic,
Mechanistic,
Linear
Biomedical
Models
Framingham
Alameda
Social Epidemiology
Behavioral
Influences
Epidemiology
Smoking
Diet
Physical Activity
Stress
Hierarchical,
Dynamic,
Systems,
Multiple determinants
Life Span Human
Developmental Psychology
Biopsychosocial
Models
Life Course Sociology
Epigenetics
Developmental
Origins of
Adult Disease
(DoAD)
Complex,
Relational,
Adaptive,
Dynamic,
Developmental
Neurodevelopment
Life Course Chronic
Disease Epidemiology
Life Course Health
Development (LCHD)
Synthesis
National Birth
Cohort Studies
Pathway
Influence
Fig. 1 The evolution of conceptual models of health development
as the etiology for chronic disease in adult life,
could not satisfactorily explain these more complex time-dependent phenomena. Instead, a theoretical framework was needed to explain the
processes of translating an individual’s experiences and exposures into the development of
health over the life course.
Two converging streams of biological research
and conceptual constructs have contributed to the
LCHD framework. The first stream represents
the basic biology of human development,
informed by the neo-Darwinian synthesis that
resulted from the convergence of Darwin’s theory of evolution and Mendel’s notion of genes as
the building blocks of heredity. Although the central dogma of “genes/DNA → mRNA → proteins” has served as a foundational construct for
modern molecular biology, it led to overly deterministic genotype-to-phenotype models (Huang
2012). Recent advances in panomics (e.g.,
genomics, epigenomics, proteomics, metabolomics) and systems biology are redefining our
understanding of how gene networks are regulated and dynamically interact with each other
and the environment, resulting in a new synthesis
of biological systems development and function-
ing (Huang 2012; Forrest 2014; Davila-Velderrain
et al. 2015). Breakthroughs in understanding the
relationships between evolutionary processes and
biological development, and advances in the use
of life history theory to explain how mismatches
between biological propensities and modern
environments influence the onset of disease, have
also provided a new way of considering the relationship of an individual’s or a population’s
genetic endowment and the phenotypes that
emerge (Del Giudice et al. 2015; Green et al.
2015; Hanson and Gluckman 2014; Lieberman
2014; Gluckman and Hanson 2006a).
The second stream of inquiry, which interacted with genetic concepts and models, represents the evolution of models of disease causation,
informed by contributions from basic, clinical,
epidemiologic, social, and psychological research
disciplines. In the first era of health science, scientific methods applied to medicine resulted in
the development of a biomedical framework in
which anatomical-pathological disease models,
along with other mechanistic constructs, were
used to explain why disease develops. One prototypical theoretical construct was germ theory
(i.e., germs as the unique causes of infectious dis-
The Emerging Theoretical Framework of Life Course Health Development
eases) (Stewart 1968). Others included theories
of inheritance that were informed by simple and
mechanistic notions of genes as the unique causes
of inherited disorders and risk status. Several
decades of research on the “upstream” social and
behavioral determinants of health were stimulated by epidemiologic studies like the
Framingham Study and Alameda County Study
that highlighted how cardiovascular and other
chronic diseases were not caused by bad germs,
bad genes, or bad luck, but were related to behaviors like smoking, diet, exercise, and other social
factors as well as the metabolic changes that
these social and behavioral risk factors induced
(Dawber et al. 1974; Haynes 1980; Berkman and
Syme 1979). This led to a more dynamic ecological analysis of the multiple risk factors that lead
to disease causation, informing the creation of a
multi-causal, biopsychosocial framework of disease (Engel 1977). Over the last 20 years, this
biopsychosocial model of health has continued to
evolve as a result of the integration of concepts
from life course research in sociology (Elder
1995; Elder and Shanahan 2007), lifespan developmental research in psychology (Lerner 2012),
systems biology (Schadt and Bjorkegren 2012),
and longitudinal studies on the origins of chronic
disease (Gluckman and Hanson 2004b). The biopsychosocial model undergirds much of the current focus on the social determinants of health
and the important role that contextual factors
play in shaping health outcomes (McMichael
1999; Krieger 2001).
Midway through the twentieth century, social
scientists started examining how the rapidly
changing social circumstances of the second
industrial revolution were transforming the
developmental pathways of different generations.
Separate yet related streams of research emerged,
converging around notions of the life course, the
lifespan, and the human life cycle development.
Two lines of investigation in particular have
informed recent notions of health development:
life course sociology and lifespan human developmental psychology (Diewald and Mayer 2009).
Life course theories emerged in sociology
research in the 1960s, championed by Elder,
Clausen, and others. These theories distinguished
23
how social institutions and history shape the
roles, personal events, transitions, and trajectories of individuals who follow different developmental pathways (Clausen 1986; Elder et al.
2003). Macro-level social processes and social
relationships influence interweaving trajectories
at different ages, stages, and transitions of development (Elder 1995). Untangling age, period,
and cohort effects and understanding the cumulative impact of experience on socially and institutionally constructed life pathways form the basis
of life course sociology. For example, the experience of low socioeconomic status, discrimination, and racial segregation may have different
effects on health for different cohorts (i.e., groups
born at different times), based on prevailing
(period effects), compensatory, and mediating
factors such as the availability of healthcare or
the impact of different social policies (Chen et al.
2010; Masters et al. 2012).
Building on the work of Glen Elder, Duane
Alwin (2012) suggested five ways that the term
“life course” was used to describe etiologic processes in social and behavioral sciences: (1) lifespan development, humans develop over the life
course; (2) agency, individuals construct their
lives through choices and actions they take within
social structures that provide opportunities and
impose constraints; (3) cohort and geographic
variation, lives of individuals are embedded and
shaped by historical time and place where they
live; (4) timing, impact of events, experiences,
and transitions are conditional on their timing in
a person’s life; and, (5) linked lives, people’s
lives are lived interdependently (e.g., husband
and wife, siblings).
Lifespan human developmental psychologists
attempt to explain how individual differences
emerge at different ages and stages (Lerner 1984;
Lerner 2012). These differences are, in part,
determined by endogenous characteristics (i.e.,
each individual’s personal adaptability, plasticity,
resilience, and reactivity) interacting with exogenous factors (i.e., external physical, social, and
psychological environments that promote adaptation). These interactions cause human behavior
to continuously change from conception to death
(Lerner 1984, 2012). By focusing on the individ-
24
ual’s capacity to adapt to events and experiences
(Dannefer 1984; Alwin 2012), developmental
psychologists have suggested that lifespan human
development research concentrates on the plasticity associated with individual development
(ontogenesis), whereas life course social sciences
researchers emphasize “sociogenesis” or how life
pathways are informed and structured by socially
constructed developmental scaffolding and constraints. In short, psychologists have tended to
focus on how endogenous or constitutional ontogenetic processes influence lifelong developmental trajectories, while sociologists have been
more concerned with contextual or exogenous
factors.
Over the past 30 years, there has growing convergence between life course sociology and lifespan human developmental psychology. Research
on “linked lives,” where the common and differential impact of shared exposures is experienced
by individuals whose lives are linked geographically or socially (e.g., spouses, workers in a town,
friendship networks) and work on transitions and
turning points that are biologically (menarche,
menopause) or socially determined (e.g., transitions from preschool to kindergarten, school to
work, work to retirement), have each benefited
from consideration of endogenous and exogenous factors. As the sociological approaches to
life course and psychological approaches to lifespan research converge into a more integrated discipline of developmental science (Cairns et al.
1996; Bornstein and Lamb 2005; Diewald and
Mayer 2009), ongoing conceptual and empirical
integration is increasingly influenced by the
study of nonlinear dynamic systems, including
complex adaptive systems theory (Greenberg and
Partridge 2010).
Many researchers and thought leaders have
contributed to the conceptual evolution and
empirical evidence supporting a more integrated
developmental systems theory (Sameroff 1975;
Bronfenbrenner 1976; Baltes et al. 1980; Lerner
1984; Cicchetti and Cohen 1995; Magnusson
1995; Cairns et al. 1996; Bronfenbrenner and
Morris 2006; Sameroff 2010) which built upon
earlier behavioral and biological theories
(Greenberg and Partridge 2010; Marshall 2014).
N. Halfon and C.B. Forrest
Overton and Lerner have proposed a theoretical
construct that they call “relational developmental
systems theory (RDST)” (Lerner 2006; Lerner
and Overton 2008). Rejecting what they consider
a false dichotomy between individual and context, they suggest that a person’s development is
embedded in, organized by, and co-regulated by
his or her surrounding environments.
Developmental regulatory functions are best
understood as mutually influential, bidirectional,
person-context interactions. RDST sees individuals as active co-developers of their own developmental pathways, adaptively responding to
different biological, social, cultural, and physical
environmental contexts that they influence and
are also influenced by. RDST has been used as a
theoretical foundation for research on selfregulation and positive youth development and
has added a stronger relational dimension to life
course thinking.
Like the converging influences of life course
sociology and lifespan human developmental
psychology, many fields of the life sciences have
also informed this transition toward a life course
developmental view of health. Embryologists
and teratologists in the first part of the twentieth
century understood that environmental insults
could disrupt the normal processes of development leading to malformation and other “genetic”
abnormalities, and some scientists began to consider how childhood conditions might directly
influence adult mortality (Kuh and Davey Smith
2004). But it was not until the 1970s—when
Forsdahl suggested a relationship between childhood socioeconomic status and later cardiovascular disease, Barker studied the relationship
between birth weight and cardiovascular disease,
and Wadsworth observed that other early childhood factors influenced a range of adult health
outcomes—that a focus on what is called the
developmental origins of adult health and disease
(DOHaD) began to emerge (Forsdahl 1977;
Arnesen and Forsdahl 1985; Barker et al. 1989a,b;
Kuh and Wadsworth 1993).
The receptivity to this new perspective was
heightened by a growing number of challenges to
the biomedical model of causation. Echoing
George Engel and others, social epidemiologists
The Emerging Theoretical Framework of Life Course Health Development
like John Cassel, Leonard Syme, Lisa Berkman,
and Michael Marmot and health service researchers like Barbara Starfield began to adopt a more
complex, multidimensional “web of causation”
construct to explain the origins of disease (Cassel
1964; Starfield 1973; Starfield et al. 1984; Marmot
and Syme 1976; Syme and Berkman 1976;
Marmot et al. 1978a, b). At the same time, a growing body of new research in psychoneuroimmunology described the “embodiment of disease
risk” by demonstrating how different social, cultural, and psychological exposures quite literally
“get under the skin” and are encoded or embedded
into developing biobehavioral systems (Sapolsky
et al. 1985; Maier et al. 1994; Cohen and Herbert
1996; McEwan 1998; Repetti et al. 2002).
Over the past two decades, the Barker hypothesis, as it was commonly referred to, was further
elaborated by a series of studies examining the
impact of birth weight, fetal growth, placental
size, and weight gain in the first year of life on
metabolic regulation and cardiovascular disease
(Barker et al. 1989a,b, 1993; McMillen and
Robinson 2005; Barker et al. 2010). An entire
field of life course epidemiology was spawned
that has not only confirmed Barker’s findings in
several other cohorts and settings but vastly
expanded the empirical base linking fetal and
early childhood growth and nutrition to a growing array of adult health conditions. This work
has also gone beyond examining fetal and early
childhood origins to explore the developmental
origins of health and disease more broadly and
has generated various conceptual models to analyze and synthesize results (Schlotz and Phillips
2009; Gluckman et al. 2010; Entringer et al.
2012). A new generation of recent epigenetic
studies have begun to provide a stronger biological and theoretical basis for understanding how
developmental plasticity is manifested, how gene
expression may be modified in response to environmental cues, and how biological and behavioral traits can be perpetuated across multiple
generations (Hochberg et al. 2010; Gluckman
2014; Thayer and Kuzawa 2011; Davey Smith
2012; Lillycrop and Burdge 2012; Relton and
Davey Smith 2012; Gilbert et al. 2015; Cunliffe
2015).
25
Complementary studies of the developing
brain demonstrated how stress and social adversity influence the biology of human development
during sensitive periods (Hertzman 1999; Boyce
et al. 2012; Hertzman 2012). Building on earlier
studies of experience-dependent and experienceexpectant1 neuronal development, neurodevelopmental research demonstrated how development
is guided by the combined and interactive influences of genes and experience (Boyce et al.
2012). Animal models of experience-modified
neural development demonstrated how early
behavioral experiences of adversity or comfort
can lead to different DNA methylation patterns,
which are believed to affect gene regulation and
result in different functional levels of neurotransmission capacity (Meaney 2001; Szyf et al. 2005;
Meaney et al. 2007). Similar methylation alterations have been demonstrated in children who
have experienced adversity associated with
maternal stress in the early years (Essex et al.
2011). Research on the neurobiology of stress
and on the role that cumulative physiologic stress
can have on the function of neuroendocrine and
neuroimmunologic pathways has provided direct
evidence for how exposure to risk and/or highly
adverse environments is embedded in lifelong
biobehavioral function (Seeman 1997; McEwan
1998; Seeman et al. 2001; Repetti et al. 2011;
McEwen 2012). This research on neural development, stress, and biological priming provides an
important empirical and conceptual bridge
between observed social gradients in health and
the experience-dependent conditioning of biobe-
1
Experience-dependent neuronal development refers to
the role that experience plays in fortifying neuronal connections (e.g., a violin player who shows increased synaptic density in the area of the brain corresponding to the
motor cortex controlling the fingers, or the hypertrophy of
hippocampus in London cab drivers that is associated with
improved spatial navigation and spatial memory).
Experience-expectant neuronal development refers to
brain development that is contingent on experiences that
are expected to occur as part of normal development. For
example, typical development of the visual cortex occurs
in response to visual stimuli that are available in everyday
life. If vision is obstructed and the brain does not receive
these expected stimuli, the relevant synapses will either
not form or will atrophy.
N. Halfon and C.B. Forrest
26
havioral systems that occurs during the process
of human development (Hertzman 1999; Keating
and Hertzman 1999; Halfon and Hochstein
2002).
In several ways, the converging relationship
between life course chronic disease epidemiology, neurodevelopmental, and DOHaD research
is analogous to the converging relationship
between life course sociology and lifespan
human developmental psychology. DOHaD and
neurodevelopmental research have focused more
on the individual differences in developmental
plasticity from early development through old
age (ontogenesis), leading to a growing understanding that epigenetic factors influence nongermline heredity. For example, the exposures
grandparents experience can influence adaptive
responses two generations later. In contrast, life
course chronic disease epidemiology has focused
more on social class, social gradients, and the
social scaffolding of exposures (sociogenesis).
New longitudinal cohort studies integrate both
perspectives, including not only measures of phenotype but also genetic, epigenetic, and other
biobehavioral adaptations (Alfred et al. 2012;
Borghol et al. 2012; Elks et al. 2012).
3
Part 3: Principles of the Life
Course Health Development
Framework
In this section, we present the seven principles of
the Life Course Health Development framework
that emerged from our analysis of the network of
theories, concepts, models, and research findings
related to how health develops over the life
course.
Each principle is described, connected to relevant theories, models, and perspectives, and discussed in terms of its implications for future
research. Together, the principles constitute the
LCHD framework, which is proposed as a way of
systematically organizing the breadth of theories
and conceptual models that help to explain and
predict empirical findings on the production of
health and disease causation over the life course.
4
Principle 1: Health
Development
4.1
What We Mean by “Health
Development”
Health development integrates the concepts of
health and developmental processes into a unified whole.
We use the term “health development” to signify the framework’s central focus, which is the
linkage of health and development into a single
construct. Health is often understood as a set of
instrumental attributes that are employed when
individuals pursue goal-directed behavior
(Richman 2004; Forrest 2014). These attributes
can be thought of as “assets” that are desirable,
acquired, optimized, and maintained during the
life course, enabling growth of an individual, survival, and adaptation to manifold environments.
Examples of health assets that emerge at the level
of an individual include motor function (capacity
for movement), emotional regulation (capacity to
manage emotions during challenges or stressful
events), and cognitive function (capacity to perceive, process, and act on information leading to
the acquisition of knowledge).
Development, in this context, refers to the processes by which health attributes change (i.e.,
mature, weather, degrade) during the lifespan. If
health is a set of attributes that emerge at the level
of the whole individual, development refers to
the evolutionarily informed processes by which
these attributes enable adaptation to changing
social-environmental conditions. Health is the
“what” (i.e., what changes) and development is
the “how” (i.e., how health attributes change over
time) of health development.
As an expression of an organism’s livingness
and essential adaptive nature, health development
is an emergent property of a living system
(Forrest 2014). Importantly, because this principle combines both health and development, it
blends a temporal dimension into our conceptualization of human health. Health development has
time-dependent and transactional connotations
and is therefore dynamic.
The Emerging Theoretical Framework of Life Course Health Development
The health development of an individual at
the level of “self” cannot be understood by isolating the biological function (or dysfunction)
of an organ system or a particular behavior of an
individual—although of course these subsystems have their own unique health development
trajectories. The health development of an individual is comprised of an integrated set of
capacities that dynamically mature and are
involved in managing energy flows; processing
and acting on information; recovering from,
adapting to, and growing with environmental
challenges; learning and forming capabilities;
and producing offspring (Forrest 2014). Health
development is a life course-informed phenomenon that results from transactions between the
organism and its internal (i.e., gene, panomic,
organ system, and physiologic networks) and
external environments (i.e., family, social, cultural,
and
physical
networks
and
environments).
4.2
Theories and Frameworks
Relevant to Health
Development
This principle combines a rich set of theories and
conceptual frameworks related to health and
development. Conceptualizing health development as an emergent property of an organism differs from earlier linear and reductionistic
biomedical models of health and even from multilevel biopsychosocial models. It also sets the
stage for considering health development as complex adaptive processes that emerge from living
systems interacting with their environments. Its
relational ontology implies that other principles
contribute to the understanding of this complex
emergent process.
The optimization of health development is
codependent on several contributing developmental processes and resulting propensities that
are highlighted in the other LCHD principles and
drawn from relational developmental systems
theory (Lerner 2006; Overton 2007), developmental systems theory (Oyama 1985), dynamic
27
systems theory (Spencer et al. 2009), and the unified theory of development (Sameroff 2010).
4.3
Implications of the Health
Development Principle
The health development principle signals the
importance of context and our inability to reduce
health to its component parts divorced from the
contexts within which they develop. This type of
developmental systems thinking requires new
typologies to describe health development phenotypes. In effect, a new set of concepts is needed to
convey a language of health development as
observed and experienced at the level of whole
persons in dynamic interaction with their environments. As our understanding of the interrelationships between health development and a range of
influential environmental variables matures,
health development typologies can become fullfledged ontologies that help explain and predict
which relational influences are important and
have measurable consequences on health
development.
The creation of “whole person” health development metrics that operationalize health development concepts is necessary to capture
developmentally influenced continuity, consistency, and variability. To distinguish “health
development” from other fields in the developmental sciences, we will need to specify the
unique concepts that constitute it and the measures that assess health development’s multidimensional functionality (adaptation, energy
management, reproduction, information processing, capacity to execute tasks in response,
and restoration and their integration) as well as
its multilevel (from the molecular to the individual to the environmental) nature. Measures
of health development will also need to be
informed and reflective of the other LCHD principles outlined below. Such measures will be
particularly important in enabling and measuring the contribution of health-producing social
systems to the optimization of health
development.
N. Halfon and C.B. Forrest
28
5
Principle 2: Unfolding
5.1
What We Mean by “Unfolding”
Health development occurs continuously over the
lifespan, from conception to death, and is shaped
by prior experiences and environmental
interactions.
The unfolding principle describes the developmental processes by which expression of a few
thousand genes—none of which has a blueprint
or roadmap for constituting a viable, living
human body—can unfold in an ordered, coherent
pattern that has been shaped by the adaptive success of what has worked before. The nonlinear,
self-organizing process of development that is
made possible by molecule-to-molecule, cell-tocell, tissue-to-tissue, and human-to-human sensing and communication processes means that
health development is neither linear, passive, nor
static; rather, it is adaptive, self-organizing, and
autocatalytic (Davies 2014).
By “adaptive,” we mean those biological,
behavioral, and cultural differences that are privileged, prioritized, or selected for because of the
advantage they imbue on reproductive fitness and
success. Adaptive change occurs at multiple levels, from the biochemical and cellular to behavioral change at the level of individuals to
environmental change. For some biological systems, such as neural networks, adaptation occurs
quite rapidly, enabling real-time responses to
acute environmental challenges and acquisition
of novel information. However, some biological
and behavioral subsystems change slowly
responding to gradual shifts in the intensity and
quality of ecological exposures. Thus, the adaptation that characterizes health development transpires over multiple time scales enabling response
to both fast- and slow-changing variables. The
principles of complexity and timing will further
elaborate on these features of adaptation.
By “self-organizing,” we mean the dynamic
nonlinear process of self-assembly and selfperpetuation that emerges through multiple relational coactions between the components of a
system and its environments. In the case of
human health development, it describes how
internally determined structures emerge from a
genetic code that is regulated by layers of sensing, signaling, and feedback loops that organize
the expression of the code based on chemical
self-assembly into variable levels and forms of
differentiation. Simple differences in external
environments (at the cellular, tissue, organ system, organism, or cultural levels) transform the
pathways of development from dull uniformity to
autocatalytic diversity of forms and function
(Davies 2014).
By “autocatalytic,” we mean that health development produces the “fuel” that propels it forward (Henrich 2015). Health development
dynamically shapes and is shaped by environmental contexts. Today’s health development
serves as substrate for the emergence of future
health development states. The personenvironmental transactions that unfold during the
life course can influence gene regulation of
biobehavioral processes through epigenetic
changes. Better characterization of this set of
mechanisms is helping to explain how physical
and social exposures during childhood affect
health and disease during adulthood.
The adaptive, self-organizing, autocatalytic
processes of unfolding can help to explain how
genes and culture have coevolved. According to
Henrich (2015), as humans evolved, cultural
information and practices began to accumulate
and produce cultural adaptations. These new cultural adaptations feed forward and produce significant selection pressure on genes to improve
psychological capacities to further acquire, store,
process, and organize an array of fitness-enhancing
skills and practices. These new adaptive capacities in turn become increasingly available to others in the same cultural group. So as genetic
evolution improves the ability of our brains to
learn from others, cultural evolution can generate
adaptations (i.e., religions, markets, science) that
both enhance function and increase the selective
pressure on our brains to effectively navigate
these increasingly complex cultural forms.
Life history theory suggests that variation in
the process of unfolding result in part from the
optimization of fitness that occurs during func-
The Emerging Theoretical Framework of Life Course Health Development
tionally organized phases of the lifespan.
Borrowing from Paul Baltes’ lifespan theory
(Baltes, Lindenberger, Staudinger 2006), the
unfolding principle offers a conceptualization of
human health development as having four major
functional phases:
(1) Generativity—the preconception, prenatal,
and perinatal periods are dedicated to the formation of the organism.
(2) Acquisition of capacity—the early years
are dedicated to the acquisition, maturation, and
optimization of specific health development
capacities.
(3) Maintenance of capacity—the middle
years are dedicated to maintaining health development capacities in the face of accumulating
risks and ongoing weathering.
(4) Managing decline—the later years are
devoted to managing, adjusting, and adapting to
functional decline of various body and regulatory
systems, even as other aspects of health development such as stress management and positive
psychological functioning may improve with
age.
Each of these phases is conceptually distinct
but can contain overlapping elements, as is the
case when an older individual who is mostly in
the process of managing decline can also be
acquiring new capacities (e.g., learning to play
piano at 70). This becomes an adaptive process of
maintaining optimal function in the face of
declining capacities.
By reflecting evolutionarily defined developmental processes, levels of plasticity, and variation in expression within and across individuals
(and within and across biobehavioral systems in
the same individual), these four phases help us to
see and understand the patterns and coherence of
health development. For example, evolution has
ensured that the anatomic and metabolic process
of bone development in women produces strong
bones that enable additional weight carrying
associated with pregnancy, until the age of the
fourth and fifth decade when menopause emerges.
This is an anticipatory developmental process
whereby early anticipatory changes prepare the
individual to meet future developmental needs.
Optimizing bone metabolism and preventing
29
osteoporosis can take several forms, such as
physical activity (particularly on hard surfaces),
and include different strategies that can be
employed during the phases of acquisition and
maintenance of bone metabolism and strength
and during the management of decline after
menopause.
5.2
Theories and Frameworks
Relevant to Unfolding
The concept of health development as a continuously unfolding adaptive and self-organizing process comprised of distinct yet overlapping
functional phases provides a framework for considering how evolutionarily defined stages from
life history theory (Stearns 1992; Worthman and
Kuzara 2005; Del Giudice et al. 2015), psychological constructs from lifespan human development theory (Baltes 1983; Featherman 1983;
Lerner et al. 2010), and sociological constructs
from life course sociology (Elder 2000; Mayer
2009; Alwin 2012) can be aligned, compared,
and potentially integrated. It also provides a better way of articulating and assessing the alignment between biologically, psychologically,
socially, and culturally determined transitions
and turning points and understanding how they
impact health development over the lifespan
(Davies 2014; Henrich 2015) (see Principle
4--Timing). For example, the alignment among
the biological processes of menarche, the behavioral maturation of reproduction behaviors, and
the culturally created process of mating and marriage has dramatically changed as the age of
menarche has declined, the age of marriage has
increased, and the introduction of sexualized
behavioral stimuli has increased through a variety of different media and information
platforms.
5.3
Implications of the Unfolding
Principle
The adaptive, self-organizing, and autocatalytic
way that health development unfolds via com-
30
plex sensing, communication, and regulatory
processes implies that our basic, clinical, and
translational research needs to elucidate how
these processes influence the adaptive capacity of
individuals and populations. A clearer understanding of how similar self-organizing processes
unfold from the cellular level to the cultural level
could point to new ways of integrating preventive, health promoting and therapeutic interventions designed to optimize health development by
embracing a “cells to society” perspective.
We have proposed that there are four major
functional phases of LCHD: generativity, acquisition of capacity, maintenance of capacity, and
managing decline. Do these four phases provide
a logical staging for all aspects of health development? Are there subphases that need to be articulated to help us better understand the dynamics of
health development? To address these questions,
we need better measurement of the flow of health
development, both continuities and discontinuities, and its variation across and within
individuals.
Childhood obesity provides a useful example
to illustrate the research implications of the
unfolding principle. To evaluate obesity risk,
body mass index (BMI) is assessed at a fixed
point in time and contrasted with values obtained
with population age-sex-specific norms. Newer
statistical methods have been developed that
enable analysis of intraindividual trajectories to
more accurately characterize the pattern of childhood growth and uncover new associations
between the functional form of growth trajectories and future obesity and obesity-related comorbidities (Wen et al. 2012). To apply this sort of
methodology to health development more
broadly requires precise definitions and frequent
assessments of health development measures, as
well as an understanding of the expected trajectory of health development for the population.
These types of assessments are being made available by electronic health records and other digitized health data collected by healthcare
organizations, which are a new and ready data
source for health development research.
The interaction between various forms of
adversity and health development provides
N. Halfon and C.B. Forrest
another example. Understanding the effects of
social adversity and other environmental exposures on the unfolding of health development
entails not only connecting the specific types of
adversity to different outcomes but also developing a better understanding of the adaptive and
self-organizing neuronal and behavioral processes, pathways, and mechanisms by which
these outcomes are affected. These include
assessing the relationships between neurodevelopmental correlates of socioeconomic adversity
and differential structural and functional changes
in different regions of the brain (as measured
using functional MRIs) and understanding how
these changes feed forward and potentially compound or dissipate over time (Caspi et al. 2003;
Evans and Schamberg 2009; Hackman and Farah
2009; Noble et al. 2012; Power et al. 2005a, b;
Evans et al. 2012).
The short- and long-term effects of adverse in
utero exposures on health development are an
area of inquiry that is producing a wealth of
information and ripe for expansion (Gluckman
et al. 2008). This research includes studies regarding epigenetic programming associated with fetal
exposure to chemical compounds, environmental
toxicants, and smoking (Skinner et al. 2008;
Launay et al. 2009; Perera et al. 2009; Martino
and Prescott 2011), as well as studies examining
the impact of nutritional stresses on metabolic
function and future disease (Li et al. 2010).
Research on the epigenetic effects of adversity
on neurodevelopment has exploded in recent
years. Beginning with Meaney’s pathbreaking
work on the impact of maternal behavior on epigenetic mechanisms that influence gene expression and regulation of the endocrine response to
stress (including the glucocorticoid receptor and
the corticotropin-releasing factor (CRF) systems
that regulate the hypothalamic-pituitary-adrenal
axis), there have been a large number of studies
examining how different experiences, exposures,
and influences can lead to epigenetic alterations
affecting a wide range of biobehavioral functions
(Meaney 2001, 2010; Turecki and Meaney 2016;
Lester et al. 2016). One of the most interesting
and challenging areas of epigenetic research concerns the trans-generational transmission of
The Emerging Theoretical Framework of Life Course Health Development
exposures and risk through non-germline alterations of genetic information and the persistence
of these influences across subsequent generations
(Bale 2015).
6
Principle 3: Complexity
6.1
What We Mean
by “Complexity”
Health development results from adaptive, multilevel, and reciprocal relations between individuals and their physical, natural, and social
environments.
This principle indicates that health development occurs within living systems that are not
only adaptive, self-organizing, and autocatalytic
but also complex and hierarchically arranged.
The topologies of health development phenotypes cannot be fully understood using a traditional biomedical reductionist approach that
relies on an analysis and assembly of the parts of
subunits. Health development emanates from the
hierarchical and relational coactions of the biological and behavioral subsystems and their individual and collective relations with each other
and various interconnected external suprasystems (i.e., familial, social, cultural, ecological).
Health development phenotypes result from the
interplay between the individual and multiple
physical, biochemical, psychological, social, and
cultural networks that dynamically coact. As in
many complex adaptive systems, the directionality of these influences is often context dependent,
reciprocal, and influenced by feedback and feedforward influences. On the other hand, small
changes in particularly vulnerable parts of a
biobehavioral system—or at a specific timesensitive junction in a cascading process of
developmental change—can have profound nonlinear effects on the emergence of a capability or
health asset or on the overall robustness or fragility of the health development process.
Transactions between different environments
can influence gene expression, and gene expression and resultant phenotype can also influence
31
various environments, which will in turn influence additional gene expression. Processes at the
molecular level can dynamically coact with each
other, as well as with processes at the social and
ecological levels, and everywhere in between.
These are not simply hierarchical relationships of
dependent parts, but are holarchical in the sense
that each level is both a part and a whole, nested
and hierarchically aligned in the common purpose. In some cases, common purposes are optimizing health development, and in other
circumstances, they are aligned to ensure reproductive fitness at the expense of optimal health
development (Günther and Folke 1993).
6.2
Theories and Frameworks
Relevant to Complexity
The complexity principle adds the systemsoriented concepts of complexity, adaptation,
emergence, nonlinear change (i.e., small changes
can produce large effects and vice versa), and
multilevel person-environmental coactions. Key
theories, frameworks, and perspectives that support the conceptualization of this principle
include general systems theory (von Bertalanffy
1968), chaos theory (Gleick 1987; Lorenz 1993),
living systems theory (Miller 1978), humansystem framework (Brody 1973; Seeman 1989),
and complex adaptive systems theory (Holland
1998). The systems orientation to health development suggests a holistic, integrated view that
there is a need to understand the interdependence
of the parts that constitute the whole, which is
embedded in its natural and social environments.
6.3
Implications
of the Complexity Principle
Progress in genomics and network analysis is
enabling researchers to interrogate all known
gene-disease associations simultaneously and to
create a network view of patterns and principles
of human disease that would not be apparent by
examining genetic associations’ one disease at a
time (Goh et al. 2007). Extending this approach
N. Halfon and C.B. Forrest
32
to health development suggests the need to add
environmental exposures, or what has been called
the “exposome,” to analytic models to engender a
better understanding of how networks of genes
and networks of environments produce health
development phenotypes.
The time-honored scientific approach uses the
hypothetico-deductive method that derives its
cogency from the certainty of deductive inference and the plausibility of abductive inference.
Children exposed to the same interacting family,
school, and neighborhood environments experience patterns of risk, protective, and healthpromoting influences that emerge out of the
complex topography of those personenvironmental interactions. By statistically categorizing and analyzing children by their ethnicity,
family income, and family structure, the rich
interactions of different environmental factors
are often lost (Molenaar et al. 2003). Health
development is non-ergodic, meaning that each
individual’s experiences, environmental interactions, and health development phenotypes are
unique and that within a population there is
marked heterogeneity. Averaging effects across
groups tells us something about population
effects, but little about individual effects
(Molenaar and Campbell 2009). Furthermore,
even though individuals may have different life
course experiences, they can experience the same
health development phenotype, a phenomenon
called equifinality (Cicchetti and Rogosch 1996).
Systems biology and other systems-oriented
sciences offer a set of methods that can address
the non-ergodic attribute of health development
(Huang 2009). Rather than being hypothesis
driven, these methods are systems driven and
require a research strategy of interrogating the
system at the level of the whole (EA Roberts
2015, 2012). This more complex way of experimenting and generating scientifically valid information bears further discussion and explication.
New approaches to study design generation and
statistical analysis will be needed to understand
how patterns of health development are produced
by complex coactions of networks over time.
Identifying characteristic health development
phenotypes will require nonlinear models that
recognize and embrace the complexity of health
development. The focus should be on measuring
patterns of intraindividual health development,
which will require study designs that collect
detailed and large volumes of health and environmental information on individuals, forming big
health development data resources.
Environments coact with individual constitutional factors to produce health development phenotypes within a person. We have only a vague
understanding of the specific environmental variables responsible for these interactions. There is
an urgent need to create scientifically useful
typologies of environmental variables. This will
enable research to better understand how health
development signals are transduced from the
environment to the individual, altering biobehavioral system configuration and function. Better
characterization—and, ultimately, standardization—of environmental variables (the exposome)
will accelerate research on how an individual’s
contexts affect the epigenetic topography and
organize what complex systems science might
refer to as health development attractor2 states.
Standardization of concepts and measures
enables synthesis and meta-analyses across
studies.
2
An attractor is the end state of a dynamic system as it
moves over time. Once the object or data point goes into
the basin of attraction, it does not leave unless a strong
force is applied. The set of one or more attractors of a
dynamic system can be represented visually or graphically as trajectories in state space, where state space represents the multidimensional, abstract space of all possible
system behavior. There are four types of possible attractors: fixed points, limit cycles, toroidal attractors, and chaotic (or strange) attractors. Point attractors are regular,
terminating in a single point in state space. Cycle attractors are also regular, sometimes oscillating between two
or more fixed points or exhibiting a sinusoidal pattern
over time. Toroidal attractors are semi-regular, representing coupled rhythms whose ratio of periodicities terminates in an irrational rather than a rational number and
appearing in state space as a donut. Chaotic attractors are
fully irregular, represented by an aperiodic trajectory in
state space that never repeats or settles to a stable pattern,
whose basin of attraction is often fractal in shape; see
chaos. Regular point and cycle attractors are characteristics of relatively simple systems. Irregular toroidal and
chaotic attractors are more characteristics of complex
systems.
The Emerging Theoretical Framework of Life Course Health Development
We have almost no idea at present how to map
or represent multilevel emergence, because the
transactional nature of health development is not
well specified, measured, analyzed, or interpreted. For example, a child exposed to a stressful stimulus that is repeated in unpredictable and
unanticipated ways may experience epigenetic
changes in neurotransmitter metabolism that cannot be explained just by measuring the stressful
stimulus. The developmental timing of these
stressful events, their unpredictable repetition,
and the temporal rhythms of restorative processes
(e.g., sleep) are also at play, which brings us to
the next principle.
7
Principle 4: Timing
7.1
What We Mean by “Timing”
Health development is sensitive to the timing and
social structuring of environmental exposures
and experiences.
Health development is not a linear process in
which exposures to environmental stimuli or
internalized experiences have equal effects,
regardless of when in the life course they occur.
Instead, health development results from nonlinear interactions that are both time-specific and
time-dependent. There are sensitive periods of a
child’s life when the impact of certain exposures
can be greater than during other periods (Halfon
et al. 2014). Time-specific health development
pathways refer to biological conditioning that
occurs during these sensitive periods, when
developing systems are most adaptable and plastic and exogenous and endogenous influences
can result in different adaptive responses. In other
words, the same exposures can have very different effects depending on when during the life
course they occur. Because childhood is a phase
of life when biological and behavioral systems
are shaped by environmental exposures and
social experiences, the timing principle emphasizes the importance of nurturing children when
they are most sensitive to these influences (Conti
and Heckman 2013). For example, exposure to a
33
rich set of words during the early years of life can
greatly improve a child’s subsequent language
development, with cascading effects on subsequent school performance, health behaviors, and
future health status (Hart and Risley 2003).
Time-specific transitions and turning points in
health development also result from socially
structured pathways that link experiences and
exposures in time-influenced ways that create
recursive and mutually reinforcing patterns of
risk, protection, and promotion. Socially structured pathways have both period-specific and
time-dependent (cumulative) characteristics. By
arraying risk, protective, and promoting factors
into socially constructed and institutionally reinforced pathways that interact with sensitive periods of health development, societies can either
support the emergence of positive health development phenotypes or reinforce negative ones.
The role, relative dose, duration, and coaction of
risk, protective, and promoting factors during
formative, maintenance, and declining phases of
the life course all influence the slope, shape, and
contours of health development trajectories.
Thus, the timing principle summarizes a set of
models and constructs that elaborate the importance of the time dimension on health development. There are time-specific pathways that refer
to sensitive periods when environmental exposures and experiences can influence health development, and there are time-dependent pathways
that refer to the accumulation of repeated exposures to the same environmental stimuli that can
result in a weathering process that accelerates
aging (Geronimus 2013).
7.2
Theories and Frameworks
Relevant to Timing
The unfolding principle introduced the concept of
functional phases or epochs that organize the historical foundations of health development. The
timing principle adds the concepts of timedependent and time-sensitive health development
pathways that create periods of vulnerability and
robustness, as well as social structuring of environmental exposures and experiences. These con-
N. Halfon and C.B. Forrest
34
cepts are drawn from several theories, frameworks,
and perspectives including developmental origins
of health and disease (Gluckman and Hanson
2006a), life course perspective (Elder 2000;
Mayer 2009; Alwin 2012), biological embedding
(Hertzman and Boyce 2010), chronobiology
(Kreitzman and Foster 2004), developmental
time (Kuzawa and Thayer 2011), and adaptive
developmental plasticity (Gluckman et al. 2009).
7.3
Implications of the Timing
Principle
Scientists have accumulated tantalizing but as
yet limited evidence for time-sensitive health
development (Hanson and Gluckman 2014;
Hertzman 2012; Boyce and Kobor 2015).
Progress in this area has been slow in part
because of a lack of data systems that integrate
large volumes of biological (especially patterns
of gene response and epigenetic changes), clinical (such as electronic health records and biosensors), behavioral (self-report questionnaires),
and environmental data. Each of these data
sources exists in isolation. What is needed is a
new field of health development informatics that
is devoted to assembling large, integrated, longitudinal data resources and mining them for novel
associations between time, environment, and
health development outcomes.
Another challenge is the lack of research that
establishes the specific time-dependent pathways
by which human health development phenotypes
are altered or protected by various internal and
external factors. A variety of studies suggest that
physical and social environments can alter a person’s biology via epigenetic pathways that influence regulation of genetic pathways (Hertzman
and Boyce 2010). This “embedding” of experience seems to have its largest impact during specific sensitive periods of development. Why the
same experience engenders different outcomes
among individuals is one of the great mysteries
of health development. More work is needed to
elucidate these iterative and dynamic pathways
that connect environment to gene regulation to
physiological states to environmental impact.
8
Principle 5: Plasticity
8.1
What We Mean by “Plasticity”
Health development phenotypes are systematically malleable and are enabled and constrained
by evolution to enhance adaptability to diverse
environments.
The relative plasticity of these phenotypes is
responsive to transactions between evolutionarily
selected biological and behavioral conditioning
and supportive, challenging, and constraining
environments. These phenotypes have evolved to
provide adaptive capacity, plasticity (i.e., ability of
the organism to systematically alter its phenotype
in response to environmental challenges, opportunities, barriers, and constraints), and growth potential, which in aggregate refer to the robustness of
an individual’s health development. Heredity
transmits these evolutionary signals through
genetic, epigenetic, behavioral, and cultural
dimensions (Jablonka and Lamb 2006), which
establish the set of health development phenotypes
that, depending on environmental circumstances,
may or may not be selected and optimized to produce desirable outcomes. At the microlevel, there
are a range of strategies to introduce variable types
and levels of plasticity to optimize adaptability
from the molecular to the behavioral level. At a
macro-level, there are social and cultural strategies
that organize the phases and life stages of health
development into functionally productive entities.
Because developmental plasticity enables the
genome to produce a repertoire of possible phenotypes based on environmental cues, an individual begins their life with the capacity to
develop in different ways. Different exposures
and experiences select and instruct a developmental pathway to respond based on these evolutionary determined strategies. Underlying many
forms of plasticity are epigenetic process and
resulting cascades of secondary and tertiary
responses. Because plasticity can manifest at different levels, behavioral plasticity may be influenced by neural plasticity, and neural plasticity in
turn may be influenced by molecular plasticity
influenced by epigenetic mechanisms (Bateson
and Gluckman 2011).
The Emerging Theoretical Framework of Life Course Health Development
8.2
Theories and Frameworks
Relevant to Plasticity
Evolution both enables and constrains the portfolio of adaptive plastic responses that an individual may experience in response to environmental
interactions. Therefore, plasticity is relative, not
absolute. Although Darwin’s theory of evolution
(Darwin 1859; Huxley 1942) laid the foundation
for understanding the principle of plasticity,
more recent syntheses have expanded our understanding of heredity as including not just genetic
change but also epigenetic, behavioral, and cultural phenomena that are transmitted across generations (Waddington 1942; West-Eberhard
2003; Richardson and Boyd 2005; Jablonka and
Lamb 2006; Konner 2011; Henrich 2015). These
evolutionary forces act at the individual and
group level, a perspective known as multilevel
selection theory (Okasha 2006).
In some cases, health development outcomes
result from the developing individual “predicting” likely future environmental stimuli based on
the cues received during sensitive periods of
health development. This has been called “predictive adaptive responses” (Gluckman and
Hanson 2004). If the developing organism predicts incorrectly—that is, if the environment
experienced in the future is not compatible with
the cues received during periods of developmental plasticity—health development “mismatches”
will occur. This phenomenon can be observed
among individuals exposed in utero to maternal
malnutrition who later become obese and glucose
intolerant, a result of being born into an energyrich environment (Hales and Barker 1992).
8.3
Implications of the Plasticity
Principle
Evolution has acted on body systems in different
ways to encode various types and levels of health
development plasticity. The formation of some
biological subsystems is tightly controlled by
time and gene regulation (e.g., cardiovascular),
whereas others seem to have a range of phenotypes that can emerge as a result of interactions
35
with the environment (e.g., stress response, executive function). New models are needed to
explain the deep archeology of evolution as it
relates to the emergence of health development.
Fields like comparative biology can test some of
these hypotheses by examining the degree to
which specific processes and pathways of health
development vary or are preserved across species. For example, patterns of sleep have been
selected for and preserved across species in ways
that affect how sleep is regulated (Tamaki et al.
2016). Moreover, the success of human civilization has removed much of the selection pressure
exerted by mortality, so optimization of specific
pathways may be more strongly influenced by
culture, behavioral, and epigenetic heredity
rather than genetic forces (Enriquez and Gullans
2015). This hypothesis should be tested.
The predictive adaptive response hypothesis
has accumulated a substantial amount of animal
and human evidence for energy regulation
(Gluckman and Hanson 2004a,b). This work
should be extended to other domains—for
example, behavioral health. Just as childhood
obesity may result from mismatches between
children’s energy regulation and exposure to
energy-dense environments, it is possible that
the proliferation of childhood disorders like
attention deficit hyperactivity disorder, anxiety,
and learning disabilities may be a consequence
of mismatches between predictive adaptive
behavioral responses and the demands children
face in terms of executive functioning, emotional functioning, and learning in their home,
school, and other environments.
9
Principle 6: Thriving
9.1
What We Mean by “Thriving”
Optimal health development promotes survival,
enhances well-being, and protects against disease.
Health development bestows upon the individual
resources that have instrumental value, enabling an
individual to pursue goals and thrive (Seedhouse
2001; Blaxter 2004; Richman 2004; Forrest 2014).
N. Halfon and C.B. Forrest
36
It provides assets that individuals employ to pursue
the beings and doings (Sen 1999) that characterize
each person’s lived experiences. Thus, health development phenotypes are instrumental resources that
enable individuals to pursue desired goals and live
long, flourishing lives.
Health development phenotypes can be considered optimal according to the degree to which they
improve the chances of survival of individuals and
groups of individuals, the degree to which they
support transmission of heritable information to
successor generations, and the degree to which
they support physical robustness and psychological flourishing (what we term thriving) across time
and within the contexts of its environments.
On the other hand, the pathways by which
health development phenotypes are formed can
be perturbed to create suboptimal states that are
precursors to fully formed disease phenotypes.
These so-called endophenotypes represent intermediate, subclinical-phased transitions toward a
fully manifest phenotypic expression of a disease
or disorder (John and Lewis 1966; Gottesman
and Gould 2003). For example, the exposure to
unpredictable and uncontrollable stressors during
sensitive periods of neural development can
influence midbrain development and the functional development of attachment relationships,
the prefrontal cortex and the functional development of executive function, and the hypothalamic
pituitary axis and the regulation of stress
responses (Castellanos and Tannock 2002; Boyce
2016). Endophenotypes characterized by anxious
attachment, poor impulse control, and hyperactive stress response can impact health behaviors
and mental health and contribute to the development of many different chronic diseases including obesity, diabetes, and cardiovascular disease
(Duric et al. 2016).
9.2
Theories and Frameworks
Relevant to Thriving
The principle of health development articulated
the singularity of the concepts of health and
development. The principle of thriving further
clarifies the nature of health development by
explicitly characterizing its instrumental nature.
That is, health development provides a set of
resources that organisms draw on in order to pursue goals, such as surviving, achieving a state of
physical robustness and resilience, and psychological flourishing (Seedhouse 2001; Blaxter
2004; Committee on Evaluation of Children’s
Health 2004; Richman 2004; Forrest 2014).
Health development therefore enables the attainment of various beings (states of happiness, life
satisfaction, and meaning and purpose) and
doings (desired activities that an individual pursues) as individuals pursue their goals over the
life course (Sen 1999).
9.3
Implications of the Thriving
Principle
LCHD recognizes that phenotype is produced
by the continuous coactions of at least five factors: genome, epigenome, environment, developmental time, and life course stage. These
coactions do not merely produce single outcomes; instead they produce landscapes of
possibilities with peaks and valleys shaped by
an individual’s life history, evolutionary determined possibilities and constraints, and the
five-way interaction. Which “attractor” state
(i.e., health development phenotype) an individual settles in is the result of this complex,
nonlinear process. We know very little about
which attractor states are most likely to produce desirable outcomes (i.e., thriving) for
which individuals under which circumstances.
As we learn more about the interrelationships
among these variables, we will begin to forge
an ontology that specifies how health development variables interrelate with one another,
their subsystems and suprasystem environmental influences, and their consequences.
Research is needed that links health development phenotypes, singularly and collectively,
that enable individuals with varying personal
characteristics and environmental exposures to
lead long lives, avoid debilitating disease, and
achieve desirable goals and an optimal lived
experience.
The Emerging Theoretical Framework of Life Course Health Development
10
Principle 7: Harmony
10.1
What We Mean by “Harmony”
Health development results from the balanced
and coherent relations among molecular, physiological, behavioral, cultural, and evolutionary
processes.
Genetic modulations that occur in molecular
time frames measured in nanoseconds are linked
to biochemical modulation measured in milliseconds, homeostatic mechanisms measured in seconds to days, social norms that evolve or years and
decades, cultural processes that change from years
to centuries, and ecological processes that until
recently took millennia. Harmonious synchronization of these processes produces the rhythms and
variability that characterize health development.
Loss of coordination of these processes results in
less robustness of the human system, with resultant negative consequences. For example, the age
of menarche has decreased in response to a variety
of environmental changes that have resulted in
better health and nutrition. In traditional societies,
and until about 100 years ago, menarche coincided
with maturation of a repertoire of psychological
capabilities. Now, menarche precedes this process
of psychological maturation, which has also been
altered and extended by other cultural and environmental changes (Gluckman and Hanson
2006b). So the adaptive response of menarche to
better nutrition and health has led to a temporal
and functional uncoupling of biological and psychological capabilities that had previously been
harmonized. This has been associated with a range
of maladaptive outcomes (Mendle et al. 2007;
Ellis and Essex 2007).
10.2
Theories and Frameworks
Relevant to Harmony
The timing principle introduced the concepts of
time dependence, time sensitivity, and social
structuring of exposures. The harmony principle
extends these concepts by adding the notions of
harmonious and balanced relations of the various
37
biological, behavioral, environmental, and cultural systems that an individual is embedded
within. Extension of the classic evolutionary
modern synthesis—which assumes that phenotypic transitions occur through a series of small
steps that result in gradual evolutionary change—
has questioned and advanced core assumption
about pace of such change. Gradualism has given
way in the extended evolutionary synthesis to the
notion that evolution can manifest variable rates
of change, especially when mutations occur in
major regulatory control genes or when developmental process responds to environmental challenges with change in coordinated suites of traits
or via nonlinear threshold effects (Laland et al.
2015). Although key theories, frameworks, and
models in support of this principle come from
chronobiology (Kreitzman and Foster 2004),
developmental time (Kuzawa and Thayer 2011),
and adaptive developmental plasticity (Gluckman
et al. 2009), there is much more theoretical work
needed for this principle.
10.3
Implication of the Harmony
Principle
According to the principles of timing and plasticity, we know that there are periods of the life
course when environmental influences can have
particularly large effects on health development
plasticity. The tremendous plasticity of humans
contributes to the robustness and the ordinary
magic of child health development (Masten
2001). We need better descriptions and
conceptualizations of developmental time as it
affects all levels and dimensions of health development and how different time frames nest
together to produce coherent developmental
pathways and robustness and variability in phenotypic expression. This includes a better understanding of how molecular, physiologic,
developmental, historical, cultural, and evolutionary time frames independently and in harmony influence phenotypic variation, through
genetic, epigenetic, and yet to be determined
mechanisms and pathways. Because developmental time is uneven in its potency, intensity of
N. Halfon and C.B. Forrest
38
change, and accessibility to environmental influences, there is a great need to better characterize
and measure these temporal parameters (Boyce
et al. 2012).
11
Summary
Health and development are unified into a single
construct (health development principle) that adaptively unfolds over the life course (unfolding principle) according to the principles of complex
adaptive systems (complexity principle). Change in
health development results from time-specific processes (timing principle) that influence biobehavioral systems during sensitive periods when they are
most susceptible (plasticity principle), and the balanced alignment of molecular, biological, behavioral, cultural, and evolutionary process (harmony
principle) can result in developmental coherence.
Health development provides instrumental assets
that enable individuals and populations to pursue
desired lived experiences (thriving principle).
The Life Course Health Development framework organizes its seven principles into a coherent whole to enable the emergence of a new field
of science. The principles should not be viewed as
static, independent statements or claims. Instead,
they should be considered a set of nodes within a
highly interconnected knowledge producing and
testing network. We anticipate that these principles will change and evolve as the many fields
subsumed by the health development framework
themselves mature. Ultimately, we anticipate that
the framework will transform into a fully formed
theoretical model that enables explanation and
prediction of health development phenomena.
References
Alfred, T., Ben-Shlomo, Y., et al. (2012). A multi-cohort
study of polymorphisms in the GH/IGF Axis and
physical capability: The HALCyon Programme. PloS
One, 7(1), e29883.
Alwin, D. F. (2012). Integrating varieties of life course
concepts. The Journals of Gerontology. Series B,
Psychological Sciences and Social Sciences, 67(2),
206–220.
Antony, P. M., Balling, R., et al. (2012). From systems
biology to systems biomedicine. Current Opinion in
Biotechnology, 23(4), 604–608.
Arnesen, E., & Forsdahl, A. (1985). The Tromsø heart
study: Coronary risk factors and their association
with living conditions during childhood. Journal of
Epidemiology and Community Health, 39(3), 210–214.
Bale, T. L. (2015). Epigenetic and transgenerational
reprogramming of brain development. Nature Reviews
Neuroscience, 16(6), 332–344.
Baltes, P. B. (1983). Life-span developmental psychology:
Observations on history and theory revisited. In R. M.
Lerner (Ed.), Developmental Psychology: Historical
and philosophical perspectives (pp. 79–111).
Hillsdale: Erlbaum.
Baltes, P. B., Lindenberger, U., & Staudinger, U. M.
(2006). Life span theory in developmental psychology.
New York: John Wiley & Sons, Inc..
Baltes, P. B., Reese, H. W., et al. (1980). Life-span
Developmental Psychology. Annual Review of
Psychology, 31(1), 65–110.
Barker, D. J., Gluckman, P. D., et al. (1993). Fetal nutrition and cardiovascular disease in adult life. Lancet,
341(8850), 938–941.
Barker, D. J., Osmond, C., et al. (1989a). Growth in utero,
blood pressure in childhood and adult life, and mortality from cardiovascular disease. BMJ, 298(6673),
564–567.
Barker, D. J., Osmond, C., et al. (1989b). The intrauterine
and early postnatal origins of cardiovascular disease
and chronic bronchitis. Journal of Epidemiology and
Community Health, 43(3), 237–240.
Barker, D. J. P., Thornburg, K. L., et al. (2010). Beyond
birthweight: The maternal and placental origins of
chronic disease. Journal of Developmental Origins of
Health and Disease, 1(6), 360–364.
Bateson, P., & Gluckman, P. (2011). Plasticity, robustness,
development and evolution. Cambridge: Cambridge
University Press.
Ben-Shlomo, Y., & Kuh, D. (2002). A life course approach
to chronic disease epidemiology: Conceptual models,
empirical challenges and interdisciplinary perspectives.
International Journal of Epidemiology, 31(2), 285–293.
Berkman, L. F., & Syme, S. L. (1979). Social networks,
host resistance, and mortality: A nine-year follow-up
study of Alameda County residents. American Journal
of Epidemiology, 109(2), 186–204.
Blaxter, M. (2004). Health. Cambridge: Polity Press.
Borghol, N., Suderman, M., et al. (2012). Associations with
early-life socio-economic position in adult DNA methylation. International Journal of Epidemiology, 41(1), 62–74.
Bornstein, M. H., & Lamb, M. E. (Eds.). (2005).
Developmental science: An advanced textbook (5th
ed.). New York: Psychology Press.
Boyce, W. T., & Kobor, M. S. (2015). Development and
the epigenome: the ‘synapse’of gene–environment
interplay. Developmental science, 18(1), 1–23.
Boyce, W. T. (2016). Differential susceptibility of the
developing brain to contextual adversity and stress.
The Emerging Theoretical Framework of Life Course Health Development
Neuropsychopharmacology: Official Publication of
the American College of Neuropsychopharmacology,
41(1), 142–162.
Boyce, W. T., Sokolowski, M. B., et al. (2012). Toward
a new biology of social adversity. Proceedings of the
National Academy of Sciences of the United States of
America, 109(Suppl 2), 17143–17148.
Brody, H. (1973). The systems view of man: Implications
for medicine, science, and ethics. Perspectives in
Biology and Medicine, 17, 71–92.
Bronfenbrenner, U. (1976). The ecology of human
development: History and perspectives [The ecology
of human development: History and perspectives].
Psychologia Wychowawcza 19(5), 537–549.
Bronfenbrenner, U., & Morris, P. A. (2006). The
Bioecological model of human development. In R. M.
Lerner & W. Damon (Eds.), Handbook of child psychology: Theoretical models of human development
(Vol. 1, 6th ed., pp. 793–828). Hoboken: John Wiley
& Sons, Inc..
Cairns, R. B., Elder, G. H., et al. (1996). Developmental
science: Cambridge studies in social and emotional
development. New York: Cambridge University Press.
Caspi, A., Sugden, K., et al. (2003). Influence of life stress
on depression: Moderation by a polymorphism in the
5-HTT gene. Science, 301(5631), 386–389.
Cassel, J. (1964). Social science theory as a source of
hypotheses in epidemiological research. American
Journal of Public Health and the Nation's Health,
54(9), 1482–1488.
Castellanos, F. X., & Tannock, R. (2002). Neuroscience of
attention-deficit/hyperactivity disorder: The search for
endophenotypes. Nature Reviews Neuroscience, 3(8),
617–628.
Chen, F., Yang, Y., et al. (2010). Social change and socioeconomic disparities in health over the life course
in China a cohort analysis. American Sociological
Review, 75(1), 126–150.
Cicchetti, D., & Cohen, D. J. (1995). Developmental psychopathology: Theory and methods (Vol. 1). Oxford,
UK: John Wiley & Sons.
Cicchetti, D., & Rogosch, F. A. (1996). Equifinality
and multifinality in developmental psychopathology. Development and Psychopathology, 8(04),
597–600.
Clausen, J. A. (1986). The life course: A sociological perspective. Englewood Cliffs: Prentice-Hall.
Cohen, S., & Herbert, T. B. (1996). Health psychology:
Psychological factors and physical disease from the
perspective of human psychoneuroimmunology.
Annual Review of Psychology, 47, 113–142.
Committee on Evaluation of Children’s Health, N. R.
C. (2004). Children’s health, the Nation’s wealth:
Assessing and improving child health. Washington,
DC: National Academies Press.
Conti, G., & Heckman, J. J. (2013). The developmental approach to child and adult health. Pediatrics,
131(Suppl 2), S133–S141.
Cunliffe, V. T. (2015). Experience-sensitive epigenetic
mechanisms, developmental plasticity, and the bio-
39
logical embedding of chronic disease risk. Wiley
Interdisciplinary Reviews: Systems Biology and
Medicine, 7(2), 53–71.
Dannefer, D. (1984). The role of the social in life-span
developmental psychology, past and future: Rejoinder
to Baltes and Nesselroade. American Sociological
Review, 49(6), 847–850.
Darwin, C. (1859). On the origin of species by means of
natural selection, or the preservation of Favoured races
in the struggle for life. London, UK: John Murray.
Davey Smith, G. (2012). Epigenesis for epidemiologists: Does evo-devo have implications for population
health research and practice? International Journal of
Epidemiology, 41(1), 236–247.
Davies, J. (2014). Life unfolding: How the human body
creates itself. Oxford, UK: Oxford University Press.
Davila-Velderrain, J., Martinez-Garcia, J. C., & AlvarezBuylla, E. R. (2015). Modeling the epigenetic attractors landscape: Toward a post-genomic mechanistic
understanding of development. Frontiers in Genetics,
6, 160.
Dawber, T. R., Kannel, W. B., & Gordon, T. (1974).
Coffee and cardiovascular disease: Observations
from the Framingham study. New England Journal of
Medicine, 291(17), 871–874.
Del Giudice, M., Gangestad, S. W., & Kaplan, H. S.
(2015). Life history theory and evolutionary psychology. The handbook of evolutionary psychology. John
Wiley & Sons, Inc.
Diewald, M., & Mayer, K. U. (2009). The sociology of the
life course and life span psychology: Integrated paradigm or complementing pathways? Advances in Life
Course Research, 14(1), 5–14.
Duric, V., Clayton, S., Leong, M. L., & Yuan, L. L. (2016).
Comorbidity factors and brain mechanisms linking
chronic stress and systemic illness. Neural Plasticity,
8, 2016.
Egger, G., Liang, G., et al. (2004). Epigenetics in human
disease and prospects for epigenetic therapy. Nature,
429(6990), 457–463.
Elder, G. H., Jr. (1995). The life course paradigm: Social
change and individual development. In P. Moen, G. H.
Elder Jr., & K. Luscher (Eds.), Examining lives in
context: Perspectives on the ecology of human development (pp. 101–139). Washington, DC: American
Psychological Association.
Elder, G. H., Jr. (2000). The life course. In E. F. Borgatta
& R. J. V. Montgomery (Eds.), Encyclopedia of sociology (Vol. 3, pp. 1614–1622). New York: Macmillan
Reference.
Elder, G. H., & Shanahan, M. J. (2007). The life course
and human development. In Handbook of child psychology. New York: John Wiley & Sons, Inc.
Elder, G. H., Johnson, M. K., & Crosnoe, R. (2003). The
emergence and development of life course theory. In
Handbook of the life course (pp. 3–19). New York:
Springer US.
Elks, C. E., Loos, R. J. F., et al. (2012). Adult obesity susceptibility variants are associated with greater childhood weight gain and a faster tempo of growth: The
40
1946 British birth cohort study. American Journal of
Clinical Nutrition, 95(5), 1150–1156.
Ellis, B. J., & Essex, M. J. (2007). Family environments,
adrenarche, and sexual maturation: A longitudinal test
of a life history model. Child Development, 78(6),
1799–1817.
Engel, G. L. (1977). The need for a new medical model: A
challenge for biomedicine. Science, 196(4286), 129–136.
Enriquez, J., & Gullans, S. (2015). Evolving ourselves:
How unnatural selection and nonrandom mutation are
changing life on earth. New York: Penguin.
Entringer, S., Buss, C., et al. (2012). Fetal programming
of body composition, obesity, and metabolic function: The role of intrauterine stress and stress biology.
Journal of Nutrition and Metabolism, 2012, 632548.
Essex, M. J., Thomas Boyce, W., et al. (2011). Epigenetic
vestiges of early developmental adversity: Childhood
stress exposure and DNA methylation in adolescence.
Child Development, 84, 58–75.
Evans, G. W., & Schamberg, M. A. (2009). Childhood
poverty, chronic stress, and adult working memory.
Proceedings of the National Academy of Sciences of
the United States of America, 106(16), 6545–6549.
Evans, G. W., Chen, E., et al. (2012). How poverty
gets under the skin: A life course perspective. In
V. Maholmes & R. B. King (Eds.), The Oxford handbook of poverty and child development (pp. 13–100).
New York: Oxford University Press.
Featherman, D. L. (1983). Life span perspectives in
social science research. In P. B. Baltes & G. Brim
(Eds.), Life-span development and behavior (Vol. 5,
pp. 1–57). New York: Academic Press.
Forrest, C. B. (2014). A living systems perspective on
health. Medical Hypotheses, 82, 209–214.
Forsdahl, A. (1977). Are poor living conditions in childhood and adolescence an important risk factor for
arteriosclerotic heart disease? British Journal of
Preventive & Social Medicine, 31(2), 91–95.
Gatzweiler, F. W., & Baumüller, H. (2013). Marginality—A
framework for analyzing causal complexities of
poverty. In J. Braun von & F. W. Gatzweiler (Eds.),
Marginality: Addressing the nexus of poverty, exclusion and ecology (pp. 27–40). Dordrecht: Springer.
Geronimus, A. T. (2013). Deep integration: Letting the
epigenome out of the bottle without losing sight of
the structural origins of population health. American
Journal of Public Health, 103(S1), S56–S63.
Gilbert, S. F., Bosch, T. C., & Ledón-Rettig, C. (2015
Oct 1). Eco-Evo-Devo: Developmental symbiosis
and developmental plasticity as evolutionary agents.
Nature Reviews Genetics, 16(10), 611–622.
Gleick, J. (1987). Chaos: Making a new science.
New York: Penguin Books.
Gluckman, P. D., & Hanson, M. A. (2004a). Developmental
origins of disease paradigm: A mechanistic and evolutionary perspective. Pediatric Research, 56(3), 311–317.
Gluckman, P. D., & Hanson, M. A. (2004b). Living with
the past: Evolution, development, and patterns of disease. Science, 305(5691), 1733–1736.
N. Halfon and C.B. Forrest
Gluckman, P. D., & Hanson, M. A. (2004c). The developmental origins of the metabolic syndrome. Trends in
Endocrinology and Metabolism, 15(4), 183–187.
Gluckman, P. D., & Hanson, M. A. (2006a). Developmental
origins of health and disease. Cambridge, UK:
Cambridge University Press.
Gluckman, P. D., & Hanson, M. A. (2006b). Evolution,
development and timing of puberty. Trends in
Endocrinology and Metabolism, 17(1), 7–12.
Gluckman, P. D., Hanson, M. A., et al. (2008). Effect of
in utero and early-life conditions on adult health and
disease. New England Journal of Medicine, 359(1),
61–73.
Gluckman, P. D., Hanson, M. A., et al. (2009). Towards
a new developmental synthesis: Adaptive developmental plasticity and human disease. The Lancet,
373(9675), 1654–1657.
Gluckman, P. D., Hanson, M. A., et al. (2010). A conceptual framework for the developmental origins of
health and disease. Journal of Developmental Origins
of Health and Disease, 1(1), 6–18.
Goh, K. I., Cusick, M. E., et al. (2007). The human disease network. Proceedings of the National Academy of
Sciences, 104(21), 8685–8690.
Gottesman, I. I., & Gould, T. D. (2003). The endophenotype concept in psychiatry: Etymology and strategic intentions. The American Journal of Psychiatry,
160(4), 636–645.
Green, S., Fagan, M., & Jaeger, J. (2015). Explanatory
integration challenges in evolutionary systems biology. Biological Theory, 10(1), 18–35.
Greenberg, G., & Partridge, T. (2010). Biology, evolution,
and psychological development, In The Handbook of
life-span development (pp. 115–148). Hoboken: John
Wiley & Sons.
Günther, F., & Folke, C. (1993 Sep). Characteristics of
nested living systems. Journal of Biological Systems,
1(03), 257–274.
Hackman, D. A., & Farah, M. J. (2009). Socioeconomic
status and the developing brain. Trends in Cognitive
Sciences, 13(2), 65–73.
Hales, C. N., & Barker, D. J. P. (1992). Type 2 (noninsulin dependent) diabetes mellitus: The thrifty phenotype hypothesis. Diabetologia, 35, 595–601.
Halfon, N., & Hochstein, M. (2002). Life course healthdevelopment: An integrated framework for developing
health, policy, and research. The Milbank Quarterly,
80(3), 433–479. iii.
Halfon, N., Larson, K., Lu, M., Tullis, E., & Russ, S.
(2014). Lifecourse health development: Past, present
and future. Maternal and Child Health Journal, 18(2),
344–365.
Hanson, M. A., & Gluckman, P. D. (2014). Early developmental conditioning of later health and disease: physiology or pathophysiology? Physiological Reviews,
94(4), 1027–1076.
Hart, B., & Risley, T. R. (2003). The early catastrophe:
The 30 million word gap by age 3. American Educator,
27(1), 4–9.
The Emerging Theoretical Framework of Life Course Health Development
Haynes, S., Feinleib, M., & Kannel, W. B. (1980). The
relationship of psychosocial factors to coronary heart
disease in the Framingham Study. III. Eight-year incidence of coronary heart disease. American Journal of
Epidemiology, 111(1), 37–58.
Henrich, J. (2015). The secret of our success: how culture is driving human evolution, domesticating our
species, and making us smarter. Princeton: Princeton
University Press.
Hertzman, C. (1999). The biological embedding of early
experience and its effects on health in adulthood. Annals
of the New York Academy of Sciences, 896, 85–95.
Hertzman, C. (2012). Putting the concept of biological
embedding in historical perspective. Proceedings of
the National Academy of Sciences of the United States
of America, 109(Suppl 2), 17160–17167.
Hertzman, C., & Boyce, T. (2010). How experience gets
under the skin to create gradients in developmental
health. Annual Review of Public Health, 31, 329–347.
Hochberg, Z. E., Feil, R., Constancia, M., Fraga, M.,
Junien, C., Carel, J. C., Boileau, P., Le Bouc, Y., Deal,
C. L., Lillycrop, K., & Scharfmann, R. (2010). Child
health, developmental plasticity, and epigenetic programming. Endocrine Reviews, 32(2), 159–224.
Holland, J. H. (1998). Emergence: From chaos to order.
Cambridge, MA: Perseus Books.
Huang, S. (2009). Non-genetic heterogeneity of cells in
development: More than just noise. Development,
136(23), 3853–3862.
Huang, S. (2012). The molecular and mathematical basis
of Waddington's epigenetic landscape: A framework
for post-Darwinian biology? BioEssays: News and
Reviews in Molecular, Cellular and Developmental
Biology, 34(2), 149–157.
Huxley, J. (1942). Evolution: The modern synthesis.
London, UK: Allen & Unwin.
Jablonka, E., & Lamb, K. J. (2006). Evolution in four
dimensions: Genetic, epigenetic, behavioral, and symbolic variation in the history of life. Cambridge, MA:
MIT Press.
John, B., & Lewis, K. R. (1966). Chromosome variability and geographic distribution in insects. Science,
152(3723), 711–721.
Keating, D. P., & Hertzman, C. (Eds.). (1999).
Developmental health and the wealth of nations: Social,
biological, and educational dynamics. New York:
Guilford Publications.
Kitano, H. (2002). Systems biology: A brief overview.
Science, 295(5560), 1662–1664.
Konner, M. (2011). The evolution of childhood:
Relationships, emotion, mind. Cambridge, MA:
Belknap Press of Harvard University Press.
Kreitzman, L., & Foster, R. G. (2004). Rhythms of life:
The biological clocks that control the daily lives of
every living thing. New Haven: Yale University Press.
Krieger, N. (2001). Theories for social epidemiology in the
21st century: An ecosocial perspective. International
Journal of Epidemiology, 30(4), 668–677.
Kuh, D., & Ben-Shlomo, Y. (2004). Introduction. In
D. Kuh & Y. Ben-Shlomo (Eds.), A life course
41
approach to chronic disease epidemiology (pp. 3–14).
New York: Oxford University Press.
Kuh, D., & Davey Smith, G. (2004). The life course and
adult chronic disease: An historical perspective with
particular reference to coronary heart disease. In Life
course approach to chronic disease epidemiology
(pp. 15–37). Oxford University Press.
Kuh, D. J. L., & Wadsworth, M. E. J. (1993). Physical
health status at 36 years in a British national birth
cohort. Social Science & Medicine, 37(7), 905–916.
Kuzawa, C. W., & Thayer, Z. M. (2011). Timescales of
human adaptation: The role of epigenetic processes.
Epigenomics, 3(2), 221–234.
Laland, K. N., Uller, T., Feldman, M. W., Sterelny,
K., Müller, G. B., Moczek, A., Jablonka, E., &
Odling-Smee, J. (2015). The extended evolutionary synthesis: Its structure, assumptions and predictions. Proceedings. Royal Society B, 282(1813),
20151019.
Launay, J. M., Del Pino, M., et al. (2009). Smoking
induces long-lasting effects through a monoamineoxidase epigenetic regulation. PloS One, 4(11), e7959.
Lerner, D. R. M. (1984). On the nature of human plasticity. Cambridge: Cambridge University Press.
Lerner, R. M. (2006). Developmental science, developmental systems, and contemporary theories of human development. In W. Damon & R. M. Lerner (Eds.), Handbook
of child psychology, Vol. 1: Theoretical models of human
development (pp. 1–17). Hoboken: Wiley.
Lerner, R. M. (2012). Developmental science: Past, present, and future. International Journal of Developmental
Science, 6(1), 29–36.
Lerner, R. M., & Overton, W. F. (2008). Exemplifying the
integrations of the relational developmental system
synthesizing theory, research, and application to promote positive development and social justice. Journal
of Adolescent Research, 23(3), 245–255.
Lerner, R. M., Overton, W., et al. (2010). Life-span development: Concepts and issues. In The Handbook of lifespan development (pp. 1–29). Hoboken: Wiley.
Lester, B. M., Conradt, E., & Marsit, C. (2016).
Introduction to the special section on epigenetics.
Child Development, 87(1), 29–37.
Li, C. C., Maloney, C. A., et al. (2010). Epigenetic programming by maternal nutrition: Shaping future generations. Epigenomics, 2(4), 539–549.
Lillycrop, K. A., & Burdge, G. C. (2012). Epigenetic
mechanisms linking early nutrition to long term health.
Best Practice & Research Clinical Endocrinology &
Metabolism, 26(5), 667–676.
Lieberman, D. (2014). The story of the human body: evolution, health, and disease. New York: Vintage Books.
Lorenz, E. (1993). The essence of chaos. Seattle:
University of Washington Press.
Magnusson, D. (1995). Individual development: A holistic, integrated model. In A. Holistic, P. Moen, G. H.
Elder Jr., & K. Luscher (Eds.), Examining lives in
context: Perspectives on the ecology of human development (pp. 19–60). Washington, DC: American
Psychological Association.
42
Maier, S. F., Watkins, L. R., et al. (1994).
Psychoneuroimmunology - the Interface between
behavior, brain, and immunity. American Psychologist,
49(12), 1004–1017.
Marshall, P. J. (2014). Beyond different levels: embodiment and the developmental system. Frontiers in
Psychology, 5, 929.
Marmot, M. G., & Syme, S. L. (1976). Acculturation
and coronary heart disease in Japanese- Americans.
American Journal of Epidemiology, 104(3), 225–247.
Marmot, M. G., Adelstein, A. M., et al. (1978a). Changing
social-class distribution of heart disease. British
Medical Journal, 2(6145), 1109–1112.
Marmot, M. G., Rose, G., et al. (1978b). Employment
grade and coronary heart disease in British civil
servants. Journal of Epidemiology and Community
Health, 32(4), 244–249.
Martino, D., & Prescott, S. (2011). Epigenetics and prenatal influences on asthma and allergic airways disease.
Chest, 139(3), 640–647.
Masten, A. S. (2001). Ordinary magic. Resilience processes in development. The American Psychologist,
56(3), 227–238.
Masters, R. K., Hummer, R. A., et al. (2012).
Educational differences in U.S. adult mortality a
cohort perspective. American Sociological Review,
77(4), 548–572.
Mayer, K. U. (2009). New Directions in life course
research. Annual Review of Sociology, 35, 413–433.
McEwan, B. S. (1998). Protective and damaging effects of
stress mediators. New England Journal of Medicine,
338(3), 171–179.
McEwen, B. S. (2012). Brain on stress: How the social
environment gets under the skin. Proceedings of the
National Academy of Sciences of the United States of
America, 109(Suppl 2), 17180–17185.
McMichael, A. J. (1999). Prisoners of the proximate:
Loosening the constraints on epidemiology in an
age of change. American Journal of Epidemiology,
149(10), 887–897.
McMillen, I. C., & Robinson, J. S. (2005). Developmental origins of the metabolic syndrome: Prediction, plasticity, and
programming. Physiological Reviews, 85(2), 571–633.
Meaney, M. J. (2001). Maternal care, gene expression,
and the transmission of individual differences in
stress reactivity across generations. Annual Review of
Neuroscience, 24, 1161–1192.
Meaney, M. J. (2010). Epigenetics and the biological
definition of gene x environment interactions. Child
Development, 81(1), 41–79.
Meaney, M. J., Szyf, M., et al. (2007). Epigenetic mechanisms of perinatal programming of hypothalamicpituitary-adrenal function and health. Trends in
Molecular Medicine, 13(7), 269–277.
Mendle, J., Turkheimer, E., & Emery, R. E. (2007).
Detrimental psychological outcomes associated with
early pubertal timing in adolescent girls. Developmental
Review, 27(2), 151–171.
Miller, J. G. (1978). Living systems. New York: McGraw-Hill.
N. Halfon and C.B. Forrest
Molenaar, P. C. M., & Campbell, C. G. (2009). The new
person-specific paradigm in psychology. Current
Directions in Psychological Science, 18(2), 112–117.
Molenaar, P. C. M., Huizenga, H. M., & Nesselroade,
J. R. (2003). The relationship between the structure of
interindividual and intraindividual variability: A theoretical and empirical vindication of developmental systems theory. In Understanding Human Development
(pp. 339–360). New York: Springer.
Mortimer, J. T., & Shanahan, M. J. (2007). Handbook
of the life course New York: Springer Science &
Business Media.
Noble, K. G., Houston, S. M., et al. (2012). Neural correlates of socioeconomic status in the developing human
brain. Developmental Science, 15(4), 516–527.
Okasha, S. (2006). Evolution and the levels of selection.
New York: NY, Oxford University Press.
Overton, W. F. (2007). A coherent Metatheory for dynamic
systems: Relational Organicism- Contextualism.
Human Development, 50(2–3), 154–159.
Oyama, S. (1985). The ontogeny of information:
Developmental systems and evolution. Cambridge,
UK: Cambridge University Press.
Perera, F., Tang, W. Y., et al. (2009). Relation of DNA methylation of 5′-CpG island of ACSL3 to transplacental
exposure to airborne polycyclic aromatic hydrocarbons
and childhood asthma. PloS One, 4(2), e4488.
Power, C., Graham, H., et al. (2005a). The contribution of
childhood and adult socioeconomic position to adult obesity and smoking behaviour: An international comparison.
International Journal of Epidemiology, 34(2), 335–344.
Power, C., Hypponen, E., et al. (2005b). Socioeconomic
position in childhood and early adult life and risk of
mortality: A prospective study of the mothers of the
1958 British birth cohort. American Journal of Public
Health, 95(8), 1396–1402.
Relton, C. L., & Davey Smith, G. (2012). Is epidemiology ready for epigenetics? International Journal of
Epidemiology, 41(1), 5–9.
Roberts, E. A. (2012). Using metalloproteomics to investigate the cellular physiology of copper in hepatocytes.
Metallomics, 4(7), 633–640.
Roberts, E. (2015). System-driven research: Legitimate
experimental design for biological/biomedical
research. Accessed 2017 https://dalspace.library.dal.
ca/bitstream/handle/10222/56275/Roberts-Eve-PhDPHIL-May-2015.pdf?sequence=1&isAllowed=y
Relton, C. L., & Davey Smith, G. (2012). Is epidemiology ready for epigenetics? International Journal of
Epidemiology, 41(1), 5–9.
Repetti, R. L., Robles, T. F., et al. (2011). Allostatic
processes in the family. Development and
Psychopathology, 23(3), 921–938.
Repetti, R. L., Taylor, S. E., et al. (2002). Risky families:
Family social environments and the mental and physical health of offspring. Psychological Bulletin, 128(2),
330–366.
Richman, K. A. (2004). Ethics and the metaphysics of
medicine. Cambridge, MA: The MIT Press.
The Emerging Theoretical Framework of Life Course Health Development
Richardson, P. J., & Boyd, R. (2005). Not by genes alone:
How culture transformed human evolution. University
of Chicago Press.
Sameroff, A. (1975). Transactional models in early social
relations. Human Development, 18(1–2), 65–79.
Sameroff, A. (2010). A unified theory of development:
A dialectic integration of nature and nurture. Child
Development, 81(1), 6–22.
Sapolsky, R. M., Krey, L. C., & McEWEN, B. R. U. C.
E. S. (1985). Prolonged glucocorticoid exposure
reduces hippocampal neuron number: Implications for
aging. The Journal of Neuroscience, 5(5), 1222–1227.
Schadt, E. E., & Bjorkegren, J. L. (2012). NEW: Networkenabled wisdom in biology, medicine, and health care.
Science Translational Medicine, 4(115), 115rv111.
Schlotz, W., & Phillips, D. I. (2009). Fetal origins of mental health: Evidence and mechanisms. Brain, Behavior,
and Immunity, 23(7), 905–916.
Seedhouse, D. D. (2001). Health: The foundations for
achievement. Hoboken: Wiley.
Seeman, J. (1989). Toward a model of positive health. The
American Psychologist, 44(8), 1099–1109.
Seeman, T. E. (1997). Price of adaptation--allostatic
load and its health consequences. MacArthur studies
of successful aging. Archives of Internal Medicine,
157(19), 2259–2268.
Seeman, T. E., McEwen, B. S., et al. (2001). Allostatic
load as a marker of cumulative biological risk:
MacArthur studies of successful aging. Proceedings of
the National Academy of Sciences, 98(8), 4770–4775.
Sen, A. (1999). Commodities and capabilities. Oxford:
Oxford University Press.
Skinner, M. K., Anway, M. D., et al. (2008). Transgenerational
epigenetic programming of the brain transcriptome and
anxiety behavior. PloS One, 3(11), e3745.
Spencer, J. P., Thomas, M. S. C., et al. (Eds.). (2009).
Toward a unified theory of development: Connectionism
and dynamic systems theory re-considered. New York:
Oxford University Press.
Starfield, B. (1973). Health services research: A working
model. The New England Journal of Medicine, 289(3),
132–136.
43
Starfield, B., Katz, H., Gabriel, A., et al. (1984). Morbidity
in childhood - a longitudinal view. The New England
Journal of Medicine, 310(13), 824–829.
Stearns, S. (1992). The evolution of life histories.
New York: Oxford.
Stewart, G. T. (1968). Limitations of the germ theory. The
Lancet, 291(7551), 1077–1081.
Syme, S. L., & Berkman, L. F. (1976). Social class,
susceptibility and sickness. American Journal of
Epidemiology, 104(1), 1–8.
Szyf, M., Weaver, I. C. G., et al. (2005). Maternal programming of steroid receptor expression and phenotype through DNA methylation in the rat. Frontiers in
Neuroendocrinology, 26(3–4), 139–162.
Tamaki, M., Bang, J. W., Watanabe, T., & Sasaki, Y.
(2016). Night watch in one brain hemisphere during
sleep associated with the first-night effect in humans.
Current Biology, 26(9), 1190–1194.
Thayer, Z. M., & Kuzawa, C. W. (2011). Biological memories of past environments: Epigenetic pathways to
health disparities. Epigenetics: Official Journal of the
DNA Methylation Society, 6(7), 798–803.
Turecki, G., & Meaney, M. J. (2016). Effects of the social
environment and stress on glucocorticoid receptor
gene methylation: A systematic review. Biological
Psychiatry, 79(2), 87–96.
von Bertalanffy, L. (1968). General system theory:
Foundations, development, application. New York,
NY: George Braziller.
Waddington, C. H. (1942). The Epigenotype. Endeavour,
1, 18–20.
Wen, X., Kleinman, K., et al. (2012). Childhood body
mass index trajectories: Modeling, characterizing,
pairwise correlations and socio-demographic predictors of trajectory characteristics. BMC Medical
Research Methodology, 12, 38.
West-Eberhard, M. J. (2003). Developmental plasticity
and evolution. New York: Oxford University Press.
Worthman, C. M., & Kuzara, J. (2005). Life history and
the early origins of health differentials. American
Journal of Human Biology: The Official Journal of the
Human Biology Council, 17(1), 95–112.
Open Access This chapter is licensed under the terms of the Creative Commons Attribution 4.0 International License
(http://creativecommons.org/licenses/by/4.0/), which permits use, sharing, adaptation, distribution and reproduction in
any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license and indicate if changes were made.
The images or other third party material in this chapter are included in the chapter’s Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the chapter’s Creative Commons license
and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain
permission directly from the copyright holder.
Part II
Life Stages
Preconception and Prenatal
Factors and Metabolic Risk
Guoying Wang, Tami R. Bartell, and Xiaobin Wang
1
Introduction
There is growing evidence that early life may have
a profound impact on health and disease in later life
(Gluckman et al. 2008; Hales et al. 1991). Because
this growing evidence about the prenatal and preconception origins of health and disease is vast, this
chapter will primarily focused on early life origins
of metabolic risk for obesity and type 2 diabetes
(T2DM). Obesity and type 2 diabetes affect all age
groups including mothers and young children
(International Diabetes Federation 2013), especially in poor minority populations (Al-Rubeaan
2015). Increasing evidence points to a profound
impact of early life factors (e.g., maternal obesity,
G. Wang, MD, PhD
Department of Population, Family and Reproductive
Health, Center on the Early Life Origins of Disease,
Johns Hopkins University Bloomberg School of
Public Health, Baltimore, MD, USA
T.R. Bartell, BA
Stanley Manne Children’s Research Institute, Ann &
Robert H Lurie Children’s Hospital of Chicago,
Chicago, IL, USA
X. Wang, MD, MPH, ScD (*)
Center on the Early Life Origins of Disease,
Department of Population, Family and Reproductive
Health, Johns Hopkins University Bloomberg School
of Public Health, Baltimore, MD, USA
e-mail: xwang82@jhu.edu
diabetes, and unhealthy diet) on offspring metabolic
risk (Barbour 2014a), leading to a transgenerational
amplification of obesity and diabetes. The period
from conception to birth is a time of very rapid
growth, cellular differentiation, and functional maturation of organ systems. This early life period is
particularly sensitive to alterations of the intrauterine environment including the metabolic milieu.
Epigenomic variations (regulation of gene expression) are largely established in utero (Bogdarina
et al. 2004) and are particularly sensitive to prenatal
environmental factors that may have a lifelong
impact on health and disease. Moreover, babies that
are large at birth are more likely to be overweight or
obese in childhood, with these conditions persisting
into adulthood (Knittle et al. 1979; Rolland-Cachera
et al. 2006). Recent data suggest that elevated insulin levels may also originate in utero and persist into
early childhood (Wang et al. 2014a). Taken together,
the prenatal period is a critical developmental stage
for obesity and metabolic outcomes (Wang et al.
2014b; Dietz 2004). In light of the global obesity
and T2DM epidemic and growing evidence of early
life origins of obesity and diabetes, early identification of individuals at high risk and early prevention
of obesity and metabolic syndrome are a key to
achieve primary prevention and reverse the trends
of the obesity and T2DM epidemics.
As illustrated in Fig. 1, human health is interconnected from conception to fetal life to childhood and on into adulthood and influenced by
multilevel factors from gene to society. This
© The Author(s) 2018
N. Halfon et al. (eds.), Handbook of Life Course Health Development,
DOI 10.1007/978-3-319-47143-3_3
47
G. Wang et al.
48
Environmental Exposure Across Life Span
(physical, chemical, behavioral, social)
G×E
G×E
Parental
Genome
Epigenome
Fetal
Genome
Epigenome
Long-term metabolic risk
G×E
In uterine
environment
Fetal development
programming
Birth outcomes
Clinical outcomes:
Overweight or obesity
Insulin resistance
Dyslipidemia
Hypertension
Subclinical outcomes:
Inflammatory
Adipokines
Metabolites profile
Placental
structure
function
Fig. 1 Conceptual framework for preconceptional and prenatal factors and metabolic risk
chapter will discuss the impact of important
preconceptional and prenatal factors, including
maternal obesity and/or diabetes, gestational
weight gain, and maternal micronutrient status,
on in utero and lifelong metabolic outcomes and
the possible gene–environment interactions and
epigenetic mechanisms underlying early life origins of metabolic risk. Finally, it will provide
perspectives on current knowledge gaps and recommendations to advance the field.
2
The Effects of Maternal
Preconceptional Obesity
and/or Diabetes
and Micronutrient Status
on Offspring Metabolic
Outcomes
Evidence is growing about the associations
between early life exposures and later health.
Major exposures encompass multilevel and multifactorial influences from the societal level
through to individual lifestyle and biological factors. While a discussion of all of the possible
risks or protective factors is beyond the scope of
this chapter, below we highlight some important
preconceptional and prenatal factors, including
prepregnancy obesity, excessive weight gain during pregnancy, preexistent or gestational diabetes, and maternal micronutrient status.
2.1
Maternal Prepregnancy
Obesity and Gestational
Weight Gain
In parallel with the global obesity epidemic, the
prevalence of obesity among women of childbearing age has also increased. In 2009–2010,
the National Health and Nutrition Examination
Survey (NHANES) found that 56% of US women
aged 20–39 were overweight or obese (body
mass index (BMI) ≥ 25 kg/m2), and in particular,
32% were obese (BMI ≥ 30.0 kg/m2) (Flegal
et al. 2012). Thus, more than half of women starting their pregnancy are already overweight or
obese, and most of them remain overweight or
obese during their entire pregnancy. To further
complicate things, women who are overweight or
obese going into pregnancy are at an increased
risk for developing metabolic disorders, such as
gestational diabetes mellitus (GDM) (Torloni
et al. 2009), hypertensive disorders of pregnancy
(Bautista-Castano et al. 2013), and excessive
gestational weight gain (GWG) (Chu et al.
Preconception and Prenatal Factors and Metabolic Risk
2009). More important, maternal obesity and its
relevant metabolic disorders may impact offspring metabolic risk in later life.
Excessive maternal prepregnancy weight and
GWG are consistent risk factors for offspring
obesity and cardiometabolic risk (Lawlor 2013;
Hochner et al. 2012). In the Jerusalem Perinatal
Family Follow-Up Study, greater maternal prepregnancy BMI, independent of GWG and confounders, was significantly associated with
higher offspring blood pressures, serum insulin
and triglyceride concentrations, BMI, waist circumference, and lower high-density lipoprotein
cholesterol (Hochner et al. 2012). Of note, the
associations between maternal BMI and offspring BP, insulin, and lipids appeared to be
largely mediated by offspring concurrent body
size (both BMI and waist circumference)
(Hochner et al. 2012). This finding emphasizes
the impact that maternal adiposity may have
through offspring adiposity on various predictors
of subclinical and clinical disease, including diabetes mellitus and cardiovascular diseases. A
large US cohort study reported that excessive
maternal GWG was independently associated
with a 46% increased risk of overweight or obesity in offspring at 2–5 years of age (Sridhar et al.
2014). In a retrospective cohort study, excessive
maternal GWG had an adverse impact on the risk
of childhood overweight and abdominal adiposity (Ensenauer et al. 2013). Kaar et al. further
reported that maternal prepregnancy BMI was
not only associated with increased general adiposity (BMI) and abdominal adiposity (waist circumference) in offspring but visceral adipose
tissue at age 10 years (Kaar et al. 2014). A recent
study points to an association between maternal
excess weight in pregnancy and offspring BMI
increase from adolescence to adulthood
(Lawrence et al. 2014). Early pregnancy obesity
has also been associated with an increased risk of
premature death in adult offspring (Reynolds
et al. 2013a). To further the negative impact,
maternal prepregnancy BMI was also associated
with increased offspring insulin resistance at age
10 years (Kaar et al. 2014) and an increased risk
of developing T2DM (Dabelea et al. 2008).
49
2.2
Preexistent and Gestational
Diabetes
In parallel with the obesity epidemic is a diabetes
pandemic, which includes an increasing number
of women with type 1 diabetes (T1D), T2DM, and
GDM (Torloni et al. 2009). A body of studies has
established a link between exposure to maternal
diabetes in utero and metabolic risk in later life. In
a multiethnic population aged 6–13 years, exposure to maternal GDM was associated with higher
BMI, waist circumference, and more abdominal
fat (Crume et al. 2011). Adjustment for socioeconomic factors, birthweight, gestational age, maternal smoking during pregnancy, diet, and physical
activity did not alter the associations; however,
adjustment for maternal prepregnancy BMI attenuated all associations (Crume et al. 2011), suggesting that maternal obesity is an important mediator.
The studies in Pima Indians of the long-term
effects of diabetic pregnancy on offspring revealed
that the offspring of women with preexistent diabetes and GDM were more obese and had higher
glucose concentrations and more diabetes than the
offspring of nondiabetic women or women who
developed diabetes after pregnancy (Pettitt et al.
1993). In the Chicago Diabetes in Pregnancy
study, offspring of mothers with preexistent diabetes and GDM had significantly higher 2-h blood
glucose and insulin levels and rate of impaired glucose tolerance than the control group of nondiabetic mothers (Silverman et al. 1995). In the
SEARCH Case–Control Study (Dabelea et al.
2008), maternal diabetes and obesity were associated with 5.7 times and 2.8 times the risk of T2DM
in young offspring aged 10–22 years, respectively.
Notably, combined prenatal exposure to maternal
diabetes and obesity could explain about 47% of
the offspring risk of T2DM (Dabelea et al. 2008).
Among Pima Indian adults, individuals whose
mothers had diabetes during pregnancy had a 40%
lower acute insulin response to a 25 g intravenous
glucose challenge than those whose mothers
developed diabetes at an early age but after the
birth of the child (Gautier et al. 2001).
One important mediator is maternal blood glucose level during pregnancy. The Hyperglycemia
G. Wang et al.
50
and Adverse Pregnancy Outcome (HAPO) study
demonstrated that maternal glucose levels below
those diagnostic of diabetes were positively associated with increased birthweight and cord blood
C-peptide levels (Metzger et al. 2008). Poor glycemic control in women with pregestational diabetes increased the risk of congenital
malformations and spontaneous abortions
(Kendrick 2004; Miller et al. 1981). Although
T1D and T2DM have different pathogeneses, the
rates of pregnancy loss were similar (Cundy et al.
2007). Another factor related to diabetes is the
comorbidities. Diabetic gastropathy, a form of
diabetic neuropathy, not only worsens nausea and
vomiting but also can cause difficulty with glucose control during pregnancy (Kitzmiller et al.
2008). Reece et al. reviewed the literature and
reported that maternal diabetic nephropathy is
complicated by hypertension (60%), preeclampsia (41%), premature delivery (22–30%), and
fetal growth restriction (16%) (Reece et al. 1998).
Taken together, better control of blood glucose
levels and diabetic complications may improve
prenatal outcomes.
2.3
Maternal Micronutrient Status
Growing evidence suggests that maternal nutrition, through its impact on the fetal intrauterine
environment, has a profound and lifelong influence on later health (Barker et al. 1993). Among
a number of specific nutrients that have been
implicated, folate is particularly important.
Folate is an essential vitamin B involved in
nucleic acid synthesis, DNA methylation, cellular growth, differentiation, and repair (Crider
et al. 2012; Kim 2000). The demand for folate
increases during pregnancy due to the need for
fetal and placental growth and uterus enlargement (Greenberg et al. 2011). The causal role of
folate deficiency in fetal neural tube defects
(NTDs) is well established (De Wals et al. 2007).
A recent study showed that low maternal folate
concentration was associated with higher BMI in
offspring at 5–6 years (Krikke et al. 2016). Our
study also showed that maternal low folate was
not only independently associated with offspring
overweight or obesity but also worsened the
adverse effects of maternal prepregnancy obesity
on offspring metabolic risk (Wang et al. 2016).
On the other hand, excess maternal folate status may also link to offspring adverse metabolic
outcomes. A study in an Indian population
reported a positive association between maternal
folate level and offspring homeostatic model
assessment-insulin resistance (HOMA-IR)
(Krishnaveni et al. 2014). What we gather from
the combined results of these studies is that optimizing folate nutrition in women, and especially
OWO women, may offer a safe, simple, and
effective way to decrease the risk of transgenerational obesity and diabetes.
2.4
Adverse Birth Outcomes
Birthweight reflects cumulative growth in utero.
A body of studies has suggested that maternal
obesity is associated with a risk for large for gestational age (LGA) or macrosomia in the offspring (Bautista-Castano et al. 2013). In addition,
Brumbaugh et al. demonstrated that newborns of
obese mothers with GDM showed increased
intrahepatic fat at birth (Brumbaugh et al. 2013).
Prepregnancy obesity also has been shown to
increase the risk of Cesarean section (Martin
et al. 2015; Dzakpasu et al. 2014) and preterm
birth (Cnattingius et al. 2013). In a case–control
study of 914 women with pregestational diabetes
and 4000 controls, diabetes was associated with
an increased risk of preterm birth (Cnattingius
et al. 1994). Maternal diabetes has also been
linked to macrosomia and Cesarean section
(Barbour 2014b; Koyanagi et al. 2013).
These adverse birth outcomes are also associated with obesity and metabolic syndrome
(including hypertension, dyslipidemia, obesity,
and insulin resistance/diabetes) in later life.
Hypertension: Previous studies demonstrated
that preterm birth was associated with hypertension in childhood (Sipola-Leppanen et al. 2014)
and adulthood (Irving et al. 2000; Johansson
et al. 2005). In a prospective study, Irving et al.
found that preterm birth was associated with the
risk of hypertension and hyperglycemia in adult-
Preconception and Prenatal Factors and Metabolic Risk
51
hood (Irving et al. 2000). A meta-analysis of 27
studies further confirmed that preterm birth was
associated with cardiovascular risk factors, such
as higher systolic and diastolic blood pressure
and low-density lipoprotein cholesterol in adulthood (Parkinson et al. 2013). Even more
interesting is that the difference in blood pressure
between those born preterm and those born at
term may be greater in women than in men
(Parkinson et al. 2013). Dyslipidemia: A large
cohort study reported that boys born early preterm had 6.7% higher total cholesterol, 11.7%
higher low-density lipoprotein cholesterol (LDLC), and 12.3% higher apolipoprotein B concentrations than their term peers (Sipola-Leppanen
et al. 2014). Obesity: As fat deposition occurs
largely during the third trimester of pregnancy,
preterm babies are born with low levels of body
fat (Rigo et al. 1998; Uthaya et al. 2005).
Postnatally, preterm babies are more likely to
gain excessive weight, a known risk factor of
childhood obesity (Ong et al. 2000), and tend to
have a higher proportion of central adiposity
(Uthaya et al. 2005). Insulin resistance/diabetes:
Like children who were born at term and small
for gestational age (SGA), children born preterm
have lower insulin sensitivity (Wang et al. 2014a;
Hofman et al. 2004). Hovi et al. reported that preterm birth itself, independent of birth size, may
contribute to insulin resistance, elevated blood
glucose, and higher blood pressure (BP) in young
adulthood (Hovi et al. 2007). In addition, a
Swedish study found that preterm birth was associated with later T2DM: the hazard ratio for
T2DM comparing very preterm (≤32 weeks of
gestation) with term birth was 1.67 (Kaijser et al.
2009). There is a particular lack of large-scale
longitudinal birth cohort studies to examine the
effects of preterm birth on metabolic outcomes
over critical developmental windows. One prospective birth cohort study found that preterm
birth is associated with elevated plasma insulin
levels (indirect evidence of insulin resistance) at
birth that persist to age 6.5 years (Wang et al.
2014a), suggesting that insulin resistance originates in utero and persists into later life. There is
also evidence from a mechanistic study, which
revealed that preterm birth increased the risk of
T2DM via diminished insulin sensitivity
(Pilgaard et al. 2010), and yet another study
showed that preterm birth was associated with
changes in the cord blood adipokine profile that
may contribute to the impairment of glucose
metabolism (Bhargava et al. 2004).
3
Mechanisms/Pathways
Underlying Early Life Origins
of Metabolic Risk
Compelling evidence suggests that prenatal experiences influence metabolic alterations in late life
via multiple pathways, including genetic, in utero
environment, gene–environment interaction, and
epigenetic and shared familial socioeconomic
and lifestyle factors (Fig. 1).
3.1
Genetics
Several lines of evidence support that genetics
play a key role in the long-term effects of maternal obesity and diabetes on offspring metabolic
risk. First, obesity and diabetes tend to aggregate
among families. We also know that GDM is associated with a history of T2DM. One study showed
that, compared to women with nondiabetic parents, women with any parental history of T2DM
experienced a 2.3-fold increased risk of GDM,
suggesting that the risk of GDM was positively
associated with parental history of T2DM
(Williams et al. 2003). Second, the extensive
study of genetic variation in obesity and diabetes
has led to the identification of numerous candidate genes. Although currently identified genetic
markers only explain a small proportion of metabolic risks, twin studies reveal that BMI, body fat,
and insulin sensitivity are all highly heritable
(Zhang et al. 2009; Ouyang et al. 2010). Finally,
some gene variants related to adult diseases have
been linked to offspring outcomes. A epidemiological study showed that a genetic risk score
(GRS) comprised of SNPs associated with adult
obesity-related traits may provide an approach for
G. Wang et al.
52
predicting LGA birth and newborn adiposity
beyond established risk factors (Chawla et al.
2014). A genome-wide association study of
women of European descent established that
common variants near the melanocortin-4 receptor (MC4R) influence fat mass, weight and obesity risk and over transmission of the risk allele to
obese offspring (Loos et al. 2008). GDM is not
only associated with later risk of diabetes in mothers but also with metabolic changes that may lead
to the development of diabetes in their offspring
(Silverman et al. 1998). Some genes and their
interactions in functional networks between the
mother and fetus may also play a role in organ
development (Charalambous et al. 2007).
Additionally related is the fetal insulin hypothesis proposed by Drs. Hattersley and Tooke,
which highlights that fetal genetic factors affect
not only insulin-mediated fetal growth by regulating either fetal insulin secretion or insulin sensitivity but also insulin resistance in childhood
and adulthood (Hattersley and Tooke 1999). One
example is the case in which a mother transmits
mutations and pleiotropic effects in the glucokinase gene to her child. The glucokinase gene
codes for the glycolytic enzyme glucokinase,
which acts as the pancreatic beta-cell glucose
sensor (Matschinsky et al. 1993). Such mutations
result in mild beta-cell dysfunction with slightly
elevated fasting blood glucose concentrations,
which is present in early childhood and shows
little change with age (Hattersley 1998).
3.2
Intrauterine Environment
Based on observational research, Barker et al. proposed the fetal programming hypothesis, which
conceptualized that intrauterine experiences modify fetal systems and influence health in later life
(Hales and Barker 1992). Fetal development
responds to changes in the in utero environment in
response to changing metabolism, hormone production, and tissue sensitivity to hormones
(Gluckman et al. 2008). These adaptive changes
may influence the relative development of various
organs, leading to persistent alterations in physiologic and metabolic homeostasis. Maternal pre-
pregnancy BMI: In a large cohort of 4,091
mother–father–offspring trios in Britain, researchers found that the association between parental
prepregnancy BMI and offspring adiposity at ages
9–11 years was stronger among mother–offspring
pairs than among father–offspring pairs (Lawlor
et al. 2008). Given that maternal and paternal prepregnancy BMI are markers of genetic, behavioral, and environmental factors but that only the
maternal prepregnancy BMI was reflected in the
intrauterine environment, this finding provides
some support for the role of the in utero environment in lifelong metabolic risk beyond genetic and
shared environmental contributions. Pregestational
and Gestational Diabetes: A sibling study of Pima
Indians showed that the BMI of siblings born after
the mother developed T2DM was significantly
higher than that of siblings born before their
mother developed T2DM (Dabelea et al. 2000).
Offspring born to mothers with T2DM or GDM
had up to a sixfold higher risk of developing
T2DM as young adults compared to offspring
born to mothers before they developed T2DM
(Dabelea and Crume 2011). Other strong evidence
comes from a study in which comparisons were
made between sibling groups where one sibling
was born before the mother experienced dramatic
weight loss after bariatric surgery and the other
was born after the surgery (Kral et al. 2006).
Siblings born before maternal bariatric surgery
were at a much greater risk for obesity than siblings born after the weight loss surgery (Kral et al.
2006). Moreover, infants of mothers who had bariatric surgery before pregnancy were at a reduced
risk of being LGA at birth (Johansson et al. 2015)
and developing obesity and insulin resistance in
childhood and adolescence (Smith et al. 2009).
Collectively, these findings support the role of the
intrauterine environment in transmitting a risk for
obesity beyond that of genetics.
3.3
Gene and Environment
Interaction and Epigenetics
Epigenetic mechanisms are critical to normal
human development and play an important role
in complex human diseases (O’Neill et al. 2014;
Preconception and Prenatal Factors and Metabolic Risk
53
Reynolds et al. 2013b; Dolinoy et al. 2007). Both
animal models and human studies implicate the
intrauterine period as a sensitive time for the
establishment of epigenetic variability (Heijmans
et al. 2008; Cook et al. 2007; Tang et al. 2012;
Radford et al. 2014; Janssen et al. 2013), which
in turn influences risk for a range of disorders
that develop later in life. In the Boston Birth
Cohort, we observed a wide range of interindividual variations in genome-wide methylation at
birth (Wang et al. 2012). One possible mechanism by which maternal obesity affects offspring
OWO is via alterations in fetal DNA methylation
induced by maternal obesity.
Animal studies have shown that certain transient environmental influences in utero can produce persistent changes in epigenetic marks that
have lifelong consequences (Sinclair et al. 2007;
Anway et al. 2005). In humans, our previous
study showed an association between maternal
prepregnancy BMI and alterations in fetal DNA
methylation (Liu et al. 2014). Other studies also
showed the link between maternal obesity and
fetal epigenome alterations (Lesseur et al. 2013;
Yang et al. 2013). In addition, the use of methyl
donor supplementation has been shown to prevent transgenerational obesity (Waterland et al.
2008). A recent study indicated lower DNA
methylation plasticity in skeletal muscle among
low birthweight vs. normal birthweight men,
which potentially contributes to our understanding of the link between low birthweight and an
increased risk of T2DM (Jacobsen et al. 2014).
Genes related to metabolic risk are regulated
by epigenetic alteration: Evidence suggests that
epigenetic mechanisms are involved in the risk of
obesity. Current evidence supports the important
role of epigenetic regulation in key genes involved
in the control of adipogenesis, glucose homeostasis, inflammation, and/or insulin signaling, including leptin (Milagro et al. 2009), peroxisome
proliferator-activated receptor gamma (Noer et al.
2007), insulin (Yang et al. 2011), glucose transporter (Yokomori et al. 1999), proliferatoractivated receptor-γ (Fujiki et al. 2009), lipoprotein
lipase (Noer et al. 2007), and fatty acid-binding
protein 4 (Noer et al. 2007). In addition, greater
methylation in the promoter region of the retinoid
X receptor-a gene was also related to greater adiposity in childhood (Godfrey et al. 2011).
Genes subject to genomic imprinting are predominantly expressed from one of the two parental chromosomes and are often clustered in the
genome and epigenetically regulated. The role of
imprinted genes in growth control has been apparent since the discovery of imprinting in the early
1980s. A related study found altered methylation
at multiple imprint regulatory regions in children
born to obese parents compared with children born
to nonobese parents (Soubry et al. 2015).
4
Preconception and Prenatal
Care
Maternal health is not only important for the
mother but also for the fetus and neonate, which
makes it critical for women to be and stay healthy
during their reproductive years. It is well recognized that optimizing a woman’s health and
knowledge before planning and conceiving a
pregnancy may eliminate or reduce health risks to
her and her baby. As emerging clinical and scientific advances come to be realized through a life
course health development approach to health
optimization, the AAP/ACOG Guidelines for
Perinatal Care have shifted from framing preconceptional care as appropriately targeted toward
prospective parents who are contemplating pregnancy to an emphasis on the integration of preconception health promotion throughout the
lifespan. Taking this approach, all women and
men can benefit from preconception health,
whether or not they plan to have a baby one day.
The implementation of effective interventions
that prioritize risk factors and the provision of
quality health services during prepregnancy and
pregnancy are recommended (Bilano et al. 2014).
In a multicenter randomized trial, 1108 overweight (BMI ≥ 25 kg/m2) women were randomized to a comprehensive dietary and lifestyle
intervention. Infants born to women who had
received lifestyle advice were significantly less
likely to have a birthweight above 4000 g; treatment effect (95%CI):0.82, 0.68–0.99; P = 0.04)
(Dodd et al. 2014). All women with T2DM should
G. Wang et al.
54
be advised regarding safe, effective contraception
and the benefits of optimal glycemic control, folic
acid supplementation, and avoidance of potentially harmful medications before attempting
pregnancy. Adequate glucose control in a woman
with diabetes before conception and throughout
pregnancy can decrease maternal morbidity,
spontaneous abortion, fetal malformation, fetal
macrosomia, intrauterine fetal death, and neonatal
morbidity (American College of Obstetricians
and Gynecologists 2005).
5
Recommendations
for Future Study
and Perspectives
5.1
Major Themes and Findings
Growing evidence indicates that preconception
and prenatal risk factors may play an important
role both in fetal metabolic reprogramming and
long-term metabolic disorders in later life.
Although the onset of obesity and diabetes may
begin later in childhood or adulthood, programming at the earliest ages may contribute a latent
susceptibility. In addition to genetic susceptibility, epigenetic alterations may be important
molecular mechanisms underlying gene–environment interactions in early life origins of disease. Early identification and intervention may
improve long-term health outcomes and help to
reverse the obesity and diabetes epidemics during
the most sensitive developmental stages, when
interventions are likely to be most cost-efficient.
Particular attention is required for US urban lowincome minority populations that have been disproportionally affected by the obesity and
diabetes epidemics, and most likely fall into
vicious cycle of transgenerational obesity and
diabetes.
5.2
Research Priorities
5.2.1 Epidemiologic research
Despite the notion of early life origins of obesity
and T2DM, there is a particular lack of wellpowered prospective birth cohort studies to exam-
ine to what degree and how early life factors affect
pregnancy and infant and child health outcomes
across multiple developmental windows in a life
course framework, particularly in high-risk US
urban low-income minority populations. Findings
from this line of research will have important
research, clinical, and public health implications.
5.2.2 Mechanism research
In light of growing recognition that epigenetic
mechanisms may play an important role in mediating early life origins of diseases and that epigenome is potentially modifiable and reversible,
well-designed large-scale prospective birth
cohort studies are needed to trace the pathways
from early life factors to adverse pregnancy outcomes to postnatal long-term metabolic outcomes and to better understand how epigenome
changes from fetal to childhood to adolescence in
response to environmental exposures. The role of
social and environmental adversity in obesity and
diabetes among urban low-income minorities has
taken on new urgency given that these populations are disproportionally affected by the obesity and diabetes epidemics. In addition to
genetics and epigenetics, the field may also leverage and benefit from the latest advances in other
“omics” such as metabolomics and microbiome
as well as system sciences and bioinformatics.
5.2.3 Translational research
A particular challenge in preventing childhood
obesity is to identify important and modifiable
early life risk and protective factors to design
safe, effective, and sustainable interventions.
Evidence is needed to inform clinical guidelines
regarding the optimal age to screen for obesity in
children. Conflicting recommendations have
been proposed for the starting age: age 6 by the
US Preventive Services Task Force (Barton 2010)
vs. age 2 by the Expert Committee (Barlow
2007). While low birthweight has been included
in current clinical assessment of future metabolic
risk, there is a need to consider preterm birth as
an important risk factor of future metabolic risk.
The American Diabetes Association included
histories of SGA and maternal history of diabetes
or GDM during the child’s gestation as part of
their diabetes risk assessment for children
Preconception and Prenatal Factors and Metabolic Risk
(American Diabetes Association 2014). To date,
preterm birth has not been included in the diabetes risk assessment guidelines in children and
adults, an area that requires more research given
growing evidence linking preterm with subsequent metabolic risk.
5.3
Data and Methods
Development Priorities
Advanced analytical methods are needed to comprehensively examine temporal and causal links
of multilevel early life factors with metabolic
outcomes. Future studies need to fully capture
the complex interplay of broad environmental
factors, genome, epigenome, metabolome, and
microbiome that affect metabolic outcomes
across lifespan and generations. While some systems models exist to help characterize particular
sub-systems of the complex set of factors that
influences children’s bodyweight, none have
tried to comprehensively represent the relationship between early life factors and the subsequent
development of childhood metabolic risk across
critical developmental stages.
6
Conclusions
There is growing evidence that preconceptional
and prenatal factors play an important role in fetal
metabolic programming and metabolic risk in later
life. More research is needed to identify important
and modifiable early life risk and protective factors and underlying mechanisms, which will pave
the road for developing cost-effective early screening, prevention, and treatment strategies to halt
and reverse the obesity and T2DM epidemics in
the US, in particular among the most vulnerable
populations (urban low-income minorities).
7
Source of Funding
Drs. Xiaobin Wang and Guoying Wang are supported in part by the National Institutes of Health
(NIH) grant R01HD086013 (PI: X Wang).
55
References
Al-Rubeaan, K. (2015). National surveillance for type 1,
type 2 diabetes and prediabetes among children and
adolescents: A population-based study (SAUDI-DM).
Journal of Epidemiology and Community Health.
doi:10.1136/jech-2015-205710.
American College of Obstetricians and Gynecologists.
(2005). ACOG Practice Bulletin. Clinical Management
Guidelines for Obstetrician-Gynecologists. Number
60, March 2005. Pregestational diabetes mellitus.
Obstetrics and Gynecology, 105(3), 675–685.
American Diabetes Association. (2014). Diagnosis and
classification of diabetes mellitus. Diabetes Care,
37(Suppl 1), S81–S90.
Anway, M. D., Cupp, A. S., Uzumcu, M., & Skinner,
M. K. (2005). Epigenetic transgenerational actions
of endocrine disruptors and male fertility. Science,
308(5727), 1466–1469.
Barbour, L. A. (2014a). Changing perspectives in preexisting diabetes and obesity in pregnancy: Maternal
and infant short- and long-term outcomes. Current
Opinion in Endocrinology, Diabetes, and Obesity,
21(4), 257–263.
Barbour, L. A. (2014b). Changing perspectives in preexisting diabetes and obesity in pregnancy: Maternal
and infant short- and long-term outcomes. Current
Opinion in Endocrinology, Diabetes, and Obesity,
21(4), 257–263.
Barker, D. J., Gluckman, P. D., Godfrey, K. M., Harding,
J. E., Owens, J. A., & Robinson, J. S. (1993). Fetal
nutrition and cardiovascular disease in adult life.
Lancet, 341(8850), 938–941.
Barlow, S. E. (2007). Expert committee recommendations
regarding the prevention, assessment, and treatment of
child and adolescent overweight and obesity: Summary
report. Pediatrics, 120(Suppl 4), S164–S192.
Barton, M. (2010). Screening for obesity in children
and adolescents: US Preventive Services Task Force
recommendation statement. Pediatrics, 125(2),
361–367.
Bautista-Castano, I., Henriquez-Sanchez, P., AlemanPerez, N., et al. (2013). Maternal obesity in early pregnancy and risk of adverse outcomes. PLoS One, 8(11),
e80410.
Bhargava, S. K., Sachdev, H. S., Fall, C. H., et al. (2004).
Relation of serial changes in childhood body-mass
index to impaired glucose tolerance in young adulthood. The New England Journal of Medicine, 350(9),
865–875.
Bilano, V. L., Ota, E., Ganchimeg, T., Mori, R., & Souza,
J. P. (2014). Risk factors of pre-eclampsia/eclampsia
and its adverse outcomes in low- and middle-income
countries: A WHO secondary analysis. PLoS One,
9(3), e91198.
Bogdarina, I., Murphy, H. C., Burns, S. P., & Clark, A. J.
(2004). Investigation of the role of epigenetic modification of the rat glucokinase gene in fetal programming. Life Sciences, 74(11), 1407–1415.
56
Brumbaugh, D. E., Tearse, P., Cree-Green, M., et al.
(2013). Intrahepatic fat is increased in the neonatal
offspring of obese women with gestational diabetes.
The Journal of Pediatrics, 162(5), 930–936; e931.
Charalambous, M., da Rocha, S. T., & Ferguson-Smith,
A. C. (2007). Genomic imprinting, growth control and
the allocation of nutritional resources: Consequences
for postnatal life. Current Opinion in Endocrinology,
Diabetes, and Obesity, 14(1), 3–12.
Chawla, R., Badon, S. E., Rangarajan, J., et al. (2014).
Genetic risk score for prediction of newborn adiposity and large-for-gestational-age birth. The Journal
of Clinical Endocrinology and Metabolism, 99(11),
E2377–E2386.
Chu, S. Y., Callaghan, W. M., Bish, C. L., D’Angelo, D.
(2009). Gestational weight gain by body mass index
among US women delivering live births, 2004-2005:
Fueling future obesity. American Journal of Obstetrics
and Gynecology,. 200(3), 271; e271–277.
Cnattingius, S., Berne, C., & Nordstrom, M. L. (1994).
Pregnancy outcome and infant mortality in diabetic patients in Sweden. Diabetic Medicine, 11(7),
696–700.
Cnattingius, S., Villamor, E., Johansson, S., et al. (2013).
Maternal obesity and risk of preterm delivery. JAMA,
309(22), 2362–2370.
Cook, J. D., Davis, B. J., Goewey, J. A., Berry, T. D.,
& Walker, C. L. (2007). Identification of a sensitive
period for developmental programming that increases
risk for uterine leiomyoma in Eker rats. Reproductive
Sciences, 14(2), 121–136.
Crider, K. S., Yang, T. P., Berry, R. J., & Bailey, L. B.
(2012). Folate and DNA methylation: A review of
molecular mechanisms and the evidence for folate’s
role. Advances in Nutrition, 3(1), 21–38.
Crume, T. L., Ogden, L., West, N. A., et al. (2011).
Association of exposure to diabetes in utero with adiposity and fat distribution in a multiethnic population
of youth: The Exploring Perinatal Outcomes among
Children (EPOCH) Study. Diabetologia, 54(1), 87–92.
Cundy, T., Gamble, G., Neale, L., et al. (2007). Differing
causes of pregnancy loss in type 1 and type 2 diabetes.
Diabetes Care, 30(10), 2603–2607.
Dabelea, D., & Crume, T. (2011). Maternal environment
and the transgenerational cycle of obesity and diabetes. Diabetes, 60(7), 1849–1855.
Dabelea, D., Hanson, R. L., Lindsay, R. S., et al. (2000).
Intrauterine exposure to diabetes conveys risks for
type 2 diabetes and obesity: A study of discordant sibships. Diabetes, 49(12), 2208–2211.
Dabelea, D., Mayer-Davis, E. J., Lamichhane, A. P., et al.
(2008). Association of intrauterine exposure to maternal diabetes and obesity with type 2 diabetes in youth:
The SEARCH Case-Control Study. Diabetes Care,
31(7), 1422–1426.
De Wals, P., Tairou, F., Van Allen, M. I., et al. (2007).
Reduction in neural-tube defects after folic acid fortification in Canada. The New England Journal of
Medicine, 357(2), 135–142.
G. Wang et al.
Dietz, W. H. (2004). Overweight in childhood and adolescence. The New England Journal of Medicine, 350(9),
855–857.
Dodd, J. M., Turnbull, D., McPhee, A. J., et al. (2014).
Antenatal lifestyle advice for women who are overweight or obese: LIMIT randomised trial. BMJ, 348,
g1285.
Dolinoy, D. C., Weidman, J. R., & Jirtle, R. L. (2007).
Epigenetic gene regulation: Linking early developmental environment to adult disease. Reproductive
Toxicology, 23(3), 297–307.
Dzakpasu, S., Fahey, J., Kirby, R. S., et al. (2014).
Contribution of prepregnancy body mass index and
gestational weight gain to caesarean birth in Canada.
BMC Pregnancy and Childbirth, 14, 106.
Ensenauer, R., Chmitorz, A., Riedel, C., et al. (2013).
Effects of suboptimal or excessive gestational
weight gain on childhood overweight and abdominal adiposity: Results from a retrospective cohort
study. International Journal of Obesity, 37(4),
505–512.
Flegal, K. M., Carroll, M. D., Kit, B. K., & Ogden, C. L.
(2012). Prevalence of obesity and trends in the distribution of body mass index among US adults, 19992010. JAMA, 307(5), 491–497.
Fujiki, K., Kano, F., Shiota, K., & Murata, M. (2009).
Expression of the peroxisome proliferator activated
receptor gamma gene is repressed by DNA methylation in visceral adipose tissue of mouse models of diabetes. BMC Biology, 7, 38.
Gautier, J. F., Wilson, C., Weyer, C., et al. (2001). Low
acute insulin secretory responses in adult offspring
of people with early onset type 2 diabetes. Diabetes,
50(8), 1828–1833.
Gluckman, P. D., Hanson, M. A., Cooper, C., & Thornburg,
K. L. (2008). Effect of in utero and early-life conditions on adult health and disease. The New England
Journal of Medicine, 359(1), 61–73.
Godfrey, K. M., Sheppard, A., Gluckman, P. D., et al.
(2011). Epigenetic gene promoter methylation at birth
is associated with child's later adiposity. Diabetes,
60(5), 1528–1534.
Greenberg, J. A., Bell, S. J., Guan, Y., & Yu, Y. H. (2011).
Folic Acid supplementation and pregnancy: More
than just neural tube defect prevention. Reviews in
Obstetrics & Gynecology, 4(2), 52–59.
Hales, C. N., & Barker, D. J. (1992). Type 2 (non-insulindependent) diabetes mellitus: The thrifty phenotype
hypothesis. Diabetologia, 35(7), 595–601.
Hales, C. N., Barker, D. J., Clark, P. M., et al. (1991).
Fetal and infant growth and impaired glucose tolerance at age 64. BMJ, 303(6809), 1019–1022.
Hattersley, A. T. (1998). Maturity-onset diabetes of the
young: Clinical heterogeneity explained by genetic
heterogeneity. Diabetic Medicine, 15(1), 15–24.
Hattersley, A. T., & Tooke, J. E. (1999). The fetal insulin
hypothesis: An alternative explanation of the association of low birthweight with diabetes and vascular disease. Lancet, 353(9166), 1789–1792.
Preconception and Prenatal Factors and Metabolic Risk
57
Heijmans, B. T., Tobi, E. W., Stein, A. D., et al. (2008).
Persistent epigenetic differences associated with prenatal exposure to famine in humans. Proceedings of
the National Academy of Sciences of the United States
of America, 105(44), 17046–17049.
Hochner, H., Friedlander, Y., Calderon-Margalit, R., et al.
(2012). Associations of maternal prepregnancy body
mass index and gestational weight gain with adult
offspring cardiometabolic risk factors: The Jerusalem
Perinatal Family Follow-up Study. Circulation,
125(11), 1381–1389.
Hofman, P. L., Regan, F., Jackson, W. E., et al. (2004).
Premature birth and later insulin resistance. The New
England Journal of Medicine, 351(21), 2179–2186.
Hovi, P., Andersson, S., Eriksson, J. G., et al. (2007).
Glucose regulation in young adults with very low
birth weight. The New England Journal of Medicine,
356(20), 2053–2063.
International Diabetes Federation. IDF Diabetes Atlas, 5th
ed. 2013.; http://www.idf.org/diabetesatlas. Accessed
July 19, 2013.
Irving, R. J., Belton, N. R., Elton, R. A., & Walker, B. R.
(2000). Adult cardiovascular risk factors in premature
babies. Lancet, 355(9221), 2135–2136.
Jacobsen, S. C., Gillberg, L., Bork-Jensen, J., et al.
(2014). Young men with low birthweight exhibit
decreased plasticity of genome-wide muscle DNA
methylation by high-fat overfeeding. Diabetologia,
57(6), 1154–1158.
Janssen, B. G., Godderis, L., Pieters, N., et al. (2013).
Placental DNA hypomethylation in association with
particulate air pollution in early life. Particle and
Fibre Toxicology, 10, 22.
Johansson, S., Iliadou, A., Bergvall, N., Tuvemo, T.,
Norman, M., & Cnattingius, S. (2005). Risk of high
blood pressure among young men increases with the
degree of immaturity at birth. Circulation, 112(22),
3430–3436.
Johansson, K., Cnattingius, S., Naslund, I., et al. (2015).
Outcomes of pregnancy after bariatric surgery. The
New England Journal of Medicine, 372(9), 814–824.
Kaar, J. L., Crume, T., Brinton, J. T., Bischoff, K. J.,
McDuffie, R., & Dabelea, D. (2014). Maternal obesity,
gestational weight gain, and offspring adiposity: The
exploring perinatal outcomes among children study.
The Journal of Pediatrics, 165(3), 509–515.
Kaijser, M., Bonamy, A. K., Akre, O., et al. (2009).
Perinatal risk factors for diabetes in later life. Diabetes,
58(3), 523–526.
Kendrick, J. M. (2004). Preconception care of women
with diabetes. The Journal of Perinatal & Neonatal
Nursing, 18(1):14–25; quiz 26–17.
Kim, Y. I. (2000). Methylenetetrahydrofolate reductase
polymorphisms, folate, and cancer risk: A paradigm of
gene-nutrient interactions in carcinogenesis. Nutrition
Reviews, 58(7), 205–209.
Kitzmiller, J. L., Block, J. M., Brown, F. M., et al.
(2008). Managing preexisting diabetes for pregnancy:
Summary of evidence and consensus recommendations for care. Diabetes Care, 31(5), 1060–1079.
Knittle, J. L., Timmers, K., Ginsberg-Fellner, F., Brown,
R. E., & Katz, D. P. (1979). The growth of adipose
tissue in children and adolescents. Cross-sectional and
longitudinal studies of adipose cell number and size.
The Journal of Clinical Investigation, 63(2), 239–246.
Koyanagi, A., Zhang, J., Dagvadorj, A., et al. (2013).
Macrosomia in 23 developing countries: An analysis
of a multicountry, facility-based, cross-sectional survey. Lancet, 381(9865), 476–483.
Kral, J. G., Biron, S., Simard, S., et al. (2006). Large
maternal weight loss from obesity surgery prevents
transmission of obesity to children who were followed
for 2 to 18 years. Pediatrics, 118(6), e1644–e1649.
Krikke, G. G., Grooten, I. J., Vrijkotte, T., van Eijsden,
M., Roseboom, T. J., & Painter, R. C. (2016). Vitamin
B12 and folate status in early pregnancy and cardiometabolic risk factors in the offspring at age 5-6 years:
Findings from the ABCD multi-ethnic birth cohort.
BJOG, 123(3), 384–392.
Krishnaveni, G. V., Veena, S. R., Karat, S. C., Yajnik,
C. S., & Fall, C. H. (2014). Association between
maternal folate concentrations during pregnancy and
insulin resistance in Indian children. Diabetologia,
57(1), 110–121.
Lawlor, D. A. (2013). The Society for Social Medicine
John Pemberton Lecture 2011. Developmental overnutrition--an old hypothesis with new importance?
International Journal of Epidemiology, 42(1), 7–29.
Lawlor, D. A., Timpson, N. J., Harbord, R. M., et al. (2008).
Exploring the developmental overnutrition hypothesis
using parental-offspring associations and FTO as an
instrumental variable. PLoS Medicine, 5(3), e33.
Lawrence, G. M., Shulman, S., Friedlander, Y., et al.
(2014). Associations of maternal pre-pregnancy
and gestational body size with offspring longitudinal change in BMI. Obesity (Silver Spring), 22(4),
1165–1171.
Lesseur, C., Armstrong, D. A., Paquette, A. G., Koestler,
D. C., Padbury, J. F., & Marsit, C. J. (2013). Tissuespecific Leptin promoter DNA methylation is associated with maternal and infant perinatal factors.
Molecular and Cellular Endocrinology, 381(1–2),
160–167.
Liu, X., Chen, Q., Tsai, H. J., et al. (2014). Maternal preconception body mass index and offspring cord blood
DNA methylation: Exploration of early life origins of
disease. Environmental and Molecular Mutagenesis,
55(3), 223–230.
Loos, R. J., Lindgren, C. M., Li, S., et al. (2008). Common
variants near MC4R are associated with fat mass,
weight and risk of obesity. Nature Genetics, 40(6),
768–775.
Martin, K. E., Grivell, R. M., Yelland, L. N., & Dodd,
J. M. (2015). The influence of maternal BMI and
gestational diabetes on pregnancy outcome. Diabetes
Research and Clinical Practice, 108(3), 508–513.
Matschinsky, F., Liang, Y., Kesavan, P., et al. (1993).
Glucokinase as pancreatic beta cell glucose sensor and
diabetes gene. The Journal of Clinical Investigation,
92(5), 2092–2098.
58
Metzger, B. E., Lowe, L. P., Dyer, A. R., et al. (2008).
Hyperglycemia and adverse pregnancy outcomes.
The New England Journal of Medicine, 358(19),
1991–2002.
Milagro, F. I., Campion, J., Garcia-Diaz, D. F.,
Goyenechea, E., Paternain, L., & Martinez, J. A.
(2009). High fat diet-induced obesity modifies the
methylation pattern of leptin promoter in rats. Journal
of Physiology and Biochemistry, 65(1), 1–9.
Miller, E., Hare, J. W., Cloherty, J. P., et al. (1981).
Elevated maternal hemoglobin A1c in early pregnancy
and major congenital anomalies in infants of diabetic
mothers. The New England Journal of Medicine,
304(22), 1331–1334.
Noer, A., Boquest, A. C., & Collas, P. (2007). Dynamics
of adipogenic promoter DNA methylation during
clonal culture of human adipose stem cells to senescence. BMC Cell Biology, 8, 18.
O’Neill, R. J., Vrana, P. B., & Rosenfeld, C. S. (2014).
Maternal methyl supplemented diets and effects on
offspring health. Frontiers in Genetics, 5, 289.
Ong, K. K. L., Ahmed, M. L., Emmett, P. M., Preece,
M. A., Dunger, D. B., & Pregnancy, A. L. S. (2000).
Association between postnatal catch-up growth and
obesity in childhood: Prospective cohort study. British
Medical Journal, 320(7240), 967–971.
Ouyang, F., Christoffel, K. K., Brickman, W. J., et al.
(2010). Adiposity is inversely related to insulin sensitivity in relatively lean Chinese adolescents: A population-based twin study. The American Journal of
Clinical Nutrition, 91(3), 662–671.
Parkinson, J. R., Hyde, M. J., Gale, C., Santhakumaran,
S., & Modi, N. (2013). Preterm birth and the metabolic syndrome in adult life: A systematic review and
meta-analysis. Pediatrics, 131(4), e1240–e1263.
Pettitt, D. J., Nelson, R. G., Saad, M. F., Bennett, P. H.,
& Knowler, W. C. (1993). Diabetes and obesity in the
offspring of Pima Indian women with diabetes during
pregnancy. Diabetes Care, 16(1), 310–314.
Pilgaard, K., Faerch, K., Carstensen, B., et al. (2010). Low
birthweight and premature birth are both associated
with type 2 diabetes in a random sample of middleaged Danes. Diabetologia, 53(12), 2526–2530.
Radford, E. J., Ito, M., Shi, H., et al. (2014). In utero
effects. In utero undernourishment perturbs the adult
sperm methylome and intergenerational metabolism.
Science, 345(6198), 1255903.
Reece, E. A., Leguizamon, G., & Homko, C. (1998).
Pregnancy performance and outcomes associated
with diabetic nephropathy. American Journal of
Perinatology, 15(7), 413–421.
Reynolds, R. M., Allan, K. M., Raja, E. A., et al. (2013a).
Maternal obesity during pregnancy and premature
mortality from cardiovascular event in adult offspring: Follow-up of 1 323 275 person years. BMJ,
347, f4539.
Reynolds, R. M., Jacobsen, G. H., & Drake, A. J. (2013b).
What is the evidence in humans that DNA methyla-
G. Wang et al.
tion changes link events in utero and later life disease?
Clinical Endocrinology, 78(6), 814–822.
Rigo, J., Nyamugabo, K., Picaud, J. C., Gerard, P.,
Pieltain, C., & De Curtis, M. (1998). Reference values
of body composition obtained by dual energy X-ray
absorptiometry in preterm and term neonates. Journal
of Pediatric Gastroenterology and Nutrition, 27(2),
184–190.
Rolland-Cachera, M. F., Deheeger, M., Maillot, M., &
Bellisle, F. (2006). Early adiposity rebound: Causes
and consequences for obesity in children and adults.
International Journal of Obesity, 30(Suppl 4), S11–S17.
Silverman, B. L., Metzger, B. E., Cho, N. H., & Loeb,
C. A. (1995). Impaired glucose tolerance in adolescent
offspring of diabetic mothers. Relationship to fetal
hyperinsulinism. Diabetes Care, 18(5), 611–617.
Silverman, B. L., Rizzo, T. A., Cho, N. H., & Metzger,
B. E. (1998). Long-term effects of the intrauterine
environment. The Northwestern University Diabetes
in Pregnancy Center. Diabetes Care, 21(Suppl 2),
B142–B149.
Sinclair, K. D., Allegrucci, C., Singh, R., et al. (2007).
DNA methylation, insulin resistance, and blood pressure in offspring determined by maternal periconceptional B vitamin and methionine status. Proceedings
of the National Academy of Sciences of the United
States of America, 104(49), 19351–19356.
Sipola-Leppanen, M., Vaarasmaki, M., Tikanmaki, M.,
et al. (2014). Cardiovascular risk factors in adolescents born preterm. Pediatrics, 134(4), e1072–e1081.
Smith, J., Cianflone, K., Biron, S., et al. (2009). Effects
of maternal surgical weight loss in mothers on intergenerational transmission of obesity. The Journal
of Clinical Endocrinology and Metabolism, 94(11),
4275–4283.
Soubry, A., Murphy, S. K., Wang, F., et al. (2015).
Newborns of obese parents have altered DNA methylation patterns at imprinted genes. International
Journal of Obesity, 39(4), 650–657.
Sridhar, S. B., Darbinian, J., Ehrlich, S. F., et al. (2014).
Maternal gestational weight gain and offspring risk for
childhood overweight or obesity. American Journal of
Obstetrics and Gynecology, 211(3):259; e251–258.
Tang, W. Y., Morey, L. M., Cheung, Y. Y., Birch, L., Prins,
G. S., & Ho, S. M. (2012). Neonatal exposure to estradiol/bisphenol A alters promoter methylation and
expression of Nsbp1 and Hpcal1 genes and transcriptional programs of Dnmt3a/b and Mbd2/4 in the rat
prostate gland throughout life. Endocrinology, 153(1),
42–55.
Torloni, M. R., Betran, A. P., Horta, B. L., et al. (2009).
Prepregnancy BMI and the risk of gestational diabetes: A systematic review of the literature with metaanalysis. Obesity Reviews, 10(2), 194–203.
Uthaya, S., Thomas, E. L., Hamilton, G., Dore, C. J.,
Bell, J., & Modi, N. (2005). Altered adiposity after
extremely preterm birth. Pediatric Research, 57(2),
211–215.
Preconception and Prenatal Factors and Metabolic Risk
59
Wang, D., Liu, X., Zhou, Y., et al. (2012). Individual variation and longitudinal pattern of genome-wide DNA
methylation from birth to the first two years of life.
Epigenetics: Official Journal of the DNA Methylation
Society, 7(6), 594–605.
Wang, G., Divall, S., Radovick, S., et al. (2014a). Preterm
birth and random plasma insulin levels at birth and in
early childhood. JAMA, 311(6), 587–596.
Wang, G., Chen, Z., Bartell, T., & Wang, X. (2014b).
Early Life Origins of Metabolic Syndrome: The Role
of Environmental Toxicants. Current Environmental
Health Reports. doi:10.1007/s40572-013-0004-6.
Wang, G., Hu, F., Mistry, K., et al. (2016). Maternal
Prepregnancy BMI, Plasma Folate Level and Child
Metabolic Risk. JAMA Pediatrics. 170(8), e160845.
Waterland, R. A., Travisano, M., Tahiliani, K. G., Rached,
M. T., & Mirza, S. (2008). Methyl donor supplementation prevents transgenerational amplification
of obesity. International Journal of Obesity, 32(9),
1373–1379.
Williams, M. A., Qiu, C., Dempsey, J. C., & Luthy, D. A.
(2003). Familial aggregation of type 2 diabetes and
chronic hypertension in women with gestational diabetes mellitus. The Journal of Reproductive Medicine,
48(12), 955–962.
Yang, B. T., Dayeh, T. A., Kirkpatrick, C. L., et al. (2011).
Insulin promoter DNA methylation correlates negatively with insulin gene expression and positively
with HbA(1c) levels in human pancreatic islets.
Diabetologia, 54(2), 360–367.
Yang, Q. Y., Liang, J. F., Rogers, C. J., Zhao, J. X., Zhu,
M. J., & Du, M. (2013). Maternal obesity induces epigenetic modifications to facilitate Zfp423 expression
and enhance adipogenic differentiation in fetal mice.
Diabetes, 62(11), 3727–3735.
Yokomori, N., Tawata, M., & Onaya, T. (1999). DNA
demethylation during the differentiation of 3T3-L1
cells affects the expression of the mouse GLUT4 gene.
Diabetes, 48(4), 685–690.
Zhang, S., Liu, X., Brickman, W. J., et al. (2009).
Association of plasma leptin concentrations with adiposity measurements in rural Chinese adolescents. The
Journal of Clinical Endocrinology and Metabolism,
94(9), 3497–3504.
Open Access This chapter is licensed under the terms of the Creative Commons Attribution 4.0 International License
(http://creativecommons.org/licenses/by/4.0/), which permits use, sharing, adaptation, distribution and reproduction in
any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license and indicate if changes were made.
The images or other third party material in this chapter are included in the chapter’s Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the chapter’s Creative Commons license
and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
Early Childhood Health
and the Life Course: The State
of the Science and Proposed
Research Priorities
A Background Paper for the MCH Life
Course Research Network
W. Thomas Boyce and Clyde Hertzman
But Laura lay awake a little while, listening to Pa’s fiddle softly playing and to the
lonely sound of the wind in the Big Woods. She looked at Pa sitting on the bench by
the hearth, the firelight gleaming on his brown hair and beard and glistening on the
honey-brown fiddle. She looked at Ma, gently rocking and knitting.
She thought to herself, ‘This is now.’
She was glad that the cozy house, and Pa and Ma and the firelight and the music,
were now. They could not be forgotten, she thought, because now is now. It can never
be a long time ago.
Little House in the Big Woods
Laura Ingalls Wilder (1932)
1
Introduction: What’s
So Special About the Early
Years?
In these lines from the Little House book series
recalling her early years in a nineteenth-century
pioneer family, Laura Ingalls Wilder evoked
what we deem a timeless and evocative, if modestly sentimental, image of childhood: children’s
dependency upon the reliable and protective
presence of parents, children’s uniquely vulnera-
W. Thomas Boyce (*)
Departments of Pediatrics and Psychiatry, University
of California San Francisco, San Francisco, CA, USA
e-mail: istom.boyce@ucsf.edu
C. Hertzman
Human Early Learning Partnership, School of
Population and Public Health, University of British
Columbia, Vancouver, BC, Canada
ble and consequential sensibilities, their openness to the character and “feel” of the physical
and social worlds, and their singular immersion
in the present, the moment at hand. The overarching goals of this chapter are to survey extant
literature examining evidence for these special
susceptibilities of children to social-environmental conditions, to show how variation in life
course health development is attributable to
interactive differences in constitutional susceptibility and contextual exposure, and to summarize
lacunae in our collective vision of how early
adversity becomes biologically embedded in the
course of individual health development.
Wilder’s portrait of early childhood is the same
image promulgated in the eighteenth century
writings of Jean Jacque Rousseau, who affirmed
children’s inherent goodness and receptivity to
the social world and educational institutions’
responsibility never to impede children’s natural
independence and curiosity (see Émile, or Treatise
© The Author(s) 2018
N. Halfon et al. (eds.), Handbook of Life Course Health Development,
DOI 10.1007/978-3-319-47143-3_4
61
62
on Education). Such an image of childhood is also
wholly commensurate with twenty-first century
visions of youth as a season of special vulnerability within the human life course health development—a developmental discontinuity from the
less susceptible, more defended reaches of adulthood and life in an adult society. Some historians
of childhood have claimed that these visions of
children’s special sentience and fragility are social
constructions—products of the temporal and cultural epoch in which we now live. Philippe Ariès
in his Centuries of Childhood (Ariès 1962) and
Edward Shorter in The Making of the Modern
Family (Shorter 1975) argued, for example, that
prior to the seventeenth and eighteenth centuries,
fewer distinctions were made between children
and their adult counterparts in their needs, capacities, and proclivities. They further held that, as a
consequence of the extraordinary rates of infant
mortality at the time (25–50%; King 2007), parents were less likely to make strong emotional
investments in children. Other scholars have
judged such claims hyperbolic, however, discerning evidence for qualitative differentiation
between children and adults in ancient Egyptian
songs (“Hast thou come to kiss the child?/ I will
not let thee kiss him!/ Hast thou come to silence
him?/ I will not let thee set silence over him!”
(Hrdy 1999)), in the writings of Renaissance Italy
and Reformation Germany (King 2007) and in the
plays of Shakespeare (“Those that do teach young
babes/ Do it with gentle means and easy tasks/ He
might ha’ chid me so; for, in good faith,/ I am a
child to chiding.” Romeo and Juliet) (Chedgzoy
et al. 2007). Cultural historian Peter Stearns
(Stearns 2011) thus maintained that a special
affection for and attentiveness to children and
children’s vulnerability are no inventions or fabrications of modernity.
Nonetheless, ethnogeographer Jared Diamond
points to the dramatic variation that exists, even
in the contemporary world, among parenting
practices emanating from this differentiated and
particular view of childhood (Diamond 2012).
Both Stearns and Diamond cite, for example, the
historical and contemporary practice of swaddling infants as an iconic parenting act that is
grounded in deep cultural beliefs about the nature
W. Thomas Boyce and C. Hertzman
of infancy and childhood, about the relationship
of children to parents, and about the fundamental
nature of the human child. Hrdy shows how
extraordinary differences in mothering behavior
have occurred, historically and contemporaneously, as a consequence of sociocultural and
economic conditions, despite strong, evolutionarily conserved commonalities in innate maternal
responses (Hrdy 1999). Further, Barr et al. (1991)
demonstrate that developmental and temporal
patterns of infant crying are invariant between
North American and !Kung San infants, but
parental responses to such crying are highly
divergent. Though parenting practices come and
go and beliefs regarding the consequences of
such practices vary by geography and cultural
era, most historical and present human societies
have attributed to young children a special susceptibility to the character of the social and physical contexts in which they are reared.
There have also been abiding cultural convictions—crossing geography and time—that the
special sensibilities of young children, when
exposed to psychological and physical adversities, can produce forms of morbidity extending
well beyond the exposure itself, into the domain
of adult life. We have strongly held beliefs in the
continuities—for better and for worse—between
childhood and adulthood. John Milton (1608–
1674) wrote in Paradise Regained that “The
childhood shews the man/ As morning shews the
day” and William Wordsworth (1770–1850)
observed, in My Heart Leaps Up When I Behold,
that “The Child is father of the Man.” Marcel
Proust (1871–1922) described, in Remembrance
of Things Past, the sudden, flooding and vivid
awareness of a childhood memory in an ordinary
moment of adult life:
When from a long distant past nothing subsists,
after the people are dead, after the things are broken and scattered, taste and smell alone, more fragile but enduring, more unsubstantial, more
persistent, more faithful, remain poised a long
time, like souls, remembering, waiting, hoping,
amid the ruins of all the rest.
In the early and mid-twentieth century, these
long held beliefs about childhood’s capacity for
intrusion into the experiences of adult life began
Early Childhood Health and the Life Course: The State of the Science and Proposed Research Priorities
to find their way into the borderlands of emerging
science. Freud famously asserted that neurotic
behavior and obsessions manifesting in midadulthood were often linked, without conscious
awareness, to experiences of trauma sustained
decades before, in very early life (Freud 1940).
Biologist and scholar René Dubos invoked “biological Freudianism” to argue that the adverse
exposures of childhood can produce lasting neurobiological predispositions that persist even
when such exposures are later abated or gone
(Dubos et al. 1966). Jerome Kagan (e.g., Brim
and Kagan 1980) observed the ways in which
human health development is characterized by
both constancy and sometimes remarkable discontinuity across developmental periods. Though
the experiences of childhood may not overdetermine or dominate the strengths and liabilities of
adult life, our cultural heritage avows not only a
child’s special receptivity to the provisions and
predations of its early world but the lifelong
echoes of that receptivity, as well.
2
Susceptibility
and Continuity: The Echoes
of Childhood
The now thriving science of how early social
experience influences lifelong health and development might be legitimately portrayed as building a scientific foundation for these two
longstanding tenets of cultural belief. These are
the claims (a) that childhood is imbued with a
distinctive susceptibility to the character of the
social and physical world and (b) that, partially
as a consequence of the former, the exposures of
early life—those both damaging and sustaining—continue to exert their effects on health and
well-being long beyond the childhood years.
John Bowlby thus concluded that the infants of
all primate species show an evolutionarily conserved predisposition to form powerful emotional attachments to their mothers, striving to
stay close to her side at all times and thereby
reducing the risks of abandonment or endangerment (Bowlby 1969). Harlow, Suomi, and col-
63
leagues, in the nonhuman primate equivalent of
Bowlby’s work with human children, showed
that the drive toward early attachment is so
strong that, in the mother’s absence, an infant
macaque will physically and emotionally bond
to an inanimate surrogate but with subsequent
behavioral aberrations that extend through the
infant’s lifetime (Harlow et al. 1971). Over the
last two decades, Barker and colleagues produced a large body of findings suggesting that
chronic, life-threatening cardiovascular disease
in mid- to late life is derived from nutritional
deficits and impaired growth occurring in very
early, even prenatal life (see, e.g., Barker 1990;
Barker and Bagby 2005; Bock and Whelan
1991). Kuh and Ben-Shlomo (2004) outlined a
“life course approach” to chronic disease epidemiology, assembling convergent evidence for
temporally distant relations between markers of
childhood deprivation and compromise and indices of chronic morbidity in adult life. The Human
Development Program of the Canadian Institute
for Advanced Research (CIFAR) showed how
population health and developmental science
coincide to reveal powerful societal effects on
child development and health and how such
effects are transcribed into lifetimes of socially
partitioned differences in adult disease (Keating
and Hertzman 1999). The American Academy of
Pediatrics has recently called public attention to
the issue of significant, “toxic” stress and adversity in the lives of young children and asserted
that many of the chronic disorders of adult life
should now be regarded as “developmental disorders,” stemming as they do from adverse
childhood experiences and events. Abstracting
these and other programs of research, three
major reports—in the USA (Shonkoff and
Phillips 2000), the UK (Marmot 2010), and
Canada (Boivin and Hertzman 2012)—have
sought and achieved a multidisciplinary consensus that the experiences of early life, dramatically partitioned by aspects of socioeconomic
status (SES) and social position, result in societies with widely divergent developmental and
health outcomes. Such societies produce visibly
and persistently disadvantaged social groups
W. Thomas Boyce and C. Hertzman
64
whose children follow blighted developmental
trajectories, acquire chronic disease and disability more frequently and severely, learn and
achieve less well, and live substantially shorter,
less adaptive lives.
3
Constitution, Context,
and the Nonrandom
Distribution of Morbidity
Although strong, multinational consensus has
been reached on the power and pervasiveness of
epidemiologic findings in linking early experience
to lifelong disparities in health development and
health, urgent and challenging new research questions remain. Among these are the following:
• What is the source of the extensive variability
in associations between early adversity and
health? Why are some children profoundly
affected by early stressors and disadvantage,
while others appear to thrive in the face of
great risk?
• How does it happen? That is, what are mechanisms and mediators by which early exposures
become biologically embedded in development, yielding lifelong differences in health?
How does adversity “get into the body,” altering long-term pathogenic processes that predispose to disease and disorder?
• What role, if any, do constitutional differences
in vulnerability—genetic variation, neurobiological response biases, or enduring temperamental attributes—play in the consequences
of early life adversity?
• Does the timing, frequency, and persistence of
exposure matter? That is, is there unevenness
in the effects of adversity by the age of the
child or by the intensity and duration of the
exposure?
• Are there transgenerational effects of early
social adversity? Are the “iniquities of the
father visited upon their children, to the second and third generation” (Exodus 34:7)? Are
the traumas of a single generation transmitted
into the lives of the next?
Such questions are not just scientifically compelling; they do not simply appeal to our twentyfirst century zeal to understand nuance,
contingency, and mechanism. Conceiving and
implementing effective societal interventions to
mitigate health disparities will be necessarily
conditional upon clear and instructive answers.
As an organizational approach to these and other
life course health development questions, we
propose a useful convergence of two fundamental
principles emerging from this new science of
early childhood experience and development.
These are:
Principle 1: Most developmental and health
endpoints in human populations are derived from
causal interactions between contextual and constitutional factors.
Principle 2: Disordered development and
health are nonrandomly distributed by population, space, and time.
Principle 1 affirms a now broadly accepted
refutation of earlier claims that maladaptive outcomes are principally genetically or environmentally determined. Incontrovertible evidence now
rejects both environmental and genetic determinism and views the genesis of disease as involving
a dynamic interplay between environments and
genes. Even the simple partitioning of developmental variance into environmental and genetic
components falls short of a truly interactive view
of the conjoint operation of nurture and nature
(Boyce et al. 2012b; Meaney 2010). Importantly,
our assertion that causal interactions between contextual and constitutional differences will predominate in the pathogenesis of human morbidities
does not entail a claim that no main effects of such
differences will be found. Far more research is
needed to conclude confidently that gene-environment interplay predominates in the pathogenesis
of most human diseases, but such interactive processes appear almost certainly more prevalent and
common that was imagined even a decade ago
(Dunn et al. 2011; Wright and Christiani 2010).
Principle 2 contends, as did Bronfenbrenner’s bioecological model (e.g., Bronfenbrenner and Morris
2006), that disorders of health and development
are neither evenly nor stochastically distributed
Early Childhood Health and the Life Course: The State of the Science and Proposed Research Priorities
within human populations and that such disorders
are instead informatively clustered by population
(both within and across populations), space (both
geography and jurisdiction), and time (both developmental and historical). The maldistribution of
disorder within human populations signals important and reliable differences in susceptibility to
various forms of morbidity and their causal agents.
The aggregation of morbidities by space and time
extends beyond conventional understandings of
epidemicity to include geographic and mobility
effects on morbidity and critical periodicity in
developmental processes.
A cross-referencing of these Principles 1 and
2 produces the table below, which divides supportive evidential territory into six domains of
research: I. social stratification, poverty, and subordination; II. vulnerability, resilience, and neurobiological susceptibility; III. the topography of
vulnerability; IV. marginalization and scapegoating; V. history and affliction; and VI. critical periodicity. These six domains codify the contextual
and constitutional sources of disease maldistribution, aggregated by population, space, and
time. In the sections that follow, we abstract and
review each of these arenas of research, which
together serve to summarize current and emerging findings on the lifetime effects of early childhood exposures. The crosscutting, confluent cell
below the six domains designates a section on
interactions between context and constitution, in
the form of gene-environment interplay, and their
role in the biological embedding of early social
adversities and the emergence of what has been
65
called “the social brain.” Finally, as indicated in
table’s bottom cell, we conclude with a specification of missing areas of study, unaddressed questions, and an agenda for future work on the
developmental origins of life morbidities
(Table 1).
4
Six Domains of Early
Developmental Research
4.1
Social Stratification, Poverty,
and Subordination
Among the most well-replicated, populationlevel findings in all of the child health services
research is illustrated in Fig. 1. Within childhood
populations from a broad diversity of settings,
cultures, and societies, a subgroup of 15–20%
sustain over half of the populations’ morbidities,
both biomedical and psychiatric, and are responsible for over half of the health-care utilization
(Boyce 1985; Diaz et al. 1986). The public health
implications of this finding are self-evident: if we
could explain and address the disproportionate
morbidity carried by this small subgroup of
childhood populations, we might prevent over
half of the population’s diseases, injuries, and
disorders.
The search for an explanation for this highly
nonrandom distribution of human morbidity
begins and ends with the phenomenon of social
stratification, that is, the hierarchical social organization that characterizes much of invertebrate to
Table 1 Domains of existing, anticipated, and needed research, organized by Principles 1 and 2
Principle 2: Disordered
outcomes are
nonrandomly
distributed by
Population
Space
Time
Principle 1: Developmental and health outcomes stem from
interactions between
Context
Constitution
I. Social stratification, poverty,
II. Vulnerability, resilience, and
and subordination
neurobiological susceptibility
III. The topography of
IV. Marginalization and
vulnerability
scapegoating
V. History and affliction
VI. Critical periodicity
Interactions between context and constitution,
gene-environment interplay
An Agenda for Future Research
66
W. Thomas Boyce and C. Hertzman
Fig. 1 The nonrandom distribution of biomedical and psychiatric morbidities in child populations
vertebrate phylogeny and manifests as SES within
human societies. SES constitutes the single, most
powerful epidemiologic determinant of virtually
all forms of morbidity, a predictor so potent that
we question the authenticity of other associations
failing to adjust for SES (Adler et al. 1993, 1994).
Beginning early in life, impoverished children
and families sustain higher rates of virtually every
form of human malady: from low birth weight
(Blumenshine et al. 2010) to traumatic injury
(Brown 2010), from infectious disease (Dowd
et al. 2009) to dental caries (Boyce et al. 2010),
and from developmental disability (Msall et al.
1998) to poorer academic achievement (Kawachi
et al. 2010). Socioeconomic stratification of
developmental psychopathology, moreover,
which often emerges in preclinical form in the
middle childhood years, exerts lasting influences
on academic achievement, employment success,
interpersonal relationships, and lifelong wellbeing (Offord 1995; U.S. Department of Health
and Human Services 2011). Figures 2 and 3 show
how poor and neglected populations bear disproportionate burdens of disease and disorder
throughout the life span (see Marmot 2010)—Fig.
2 showing the graded association between SES
and socioemotional adjustment from middle
childhood through adolescence and Fig. 3 revealing the same relation with chronic disease at each
stage of the life course.
Despite this universality and potency of societal disparities in health, it is only recently that
SES has become itself a focus of serious scientific study (Syme 2008). New studies of the SES
antecedents of population morbidities have recognized the extended influence of childhood
social status on adult disease, even after controlling for SES in adulthood (Cohen et al. 2010;
Galobardes et al. 2004; Lawlor et al. 2006). Such
evidence for lifelong effects of early disadvantage is rendered still more compelling by research
documenting the developmental origins of adult
health and disease (Gluckman et al. 2005, 2009)
and by epidemiologic work revealing systematic
differences in nutrition (Khan and Bhutta 2010),
access to medical care (Houweling and Kunst
2010), and physical environmental exposures
(Gump et al. 2007) among children of differing
social classes.
Although these and other material factors
undoubtedly play roles in the origins of health
disparities (Hackman et al. 2010), another prominent and increasingly persuasive explanation for
such disparities is the SES-stratified differences
in early exposures to family adversities and
stressors (Brown et al. 2009; Hillis et al. 2004;
Wadsworth et al. 2008). Low-SES children live
with significantly more family chaos (Evans et al.
2005), sustain more frequent and severe psychological stressors (Evans 2004; Evans and Kim
Early Childhood Health and the Life Course: The State of the Science and Proposed Research Priorities
67
Percent poor
adjustment
25
20
15
10
5
0
I/II
Aged 7
IIINM
IIIM
IV/V
Social class at birth
Aged 11
Aged 16
Source: 1958 National Child Development
Study64
Fig. 2 Rates of poor social/emotional adjustment at ages 7, 11, and 17, by father’s social class at birth (Marmot 2010)
2007), and experience fewer supportive parentchild communications (Hart and Risley 1995),
relative to their higher SES peers. Low SES children also show greater neurobiological sensitivity to aversive social contexts, in the form of
heightened reactivity within the stress-responsive,
neuroendocrine pathways, i.e., the hypothalamicpituitary-adrenocortical (HPA) axis and the
autonomic nervous system (ANS) (Boyce et al.
2010; Evans and Kim 2007; Lupien et al. 2001;
Steptoe et al. 2002). Further, the exaggerated
reactivity of these peripheral stress response systems among disadvantaged youth is subserved by
socially partitioned differences in central neural
circuits, which also have profound influences on
health development, cognitive function, and educational attainment (Curley et al. 2011; Cushing
and Kramer 2005; Gianaros and Manuck 2010;
Hackman and Farah 2009; MacDonald and
Roskams 2009; McEwen and Gianaros 2010).
Influences of childhood SES on health and
neurobiological endpoints extend beyond concur-
rent effects on health and responsivity during
childhood to longer-term relations with health status in adult life. Adverse experiences in early
childhood, for example, may carry risk effects for
coronary artery disease in adult life before the age
of 50 years and effects mediated by some form of
biological embedding (Kittleson et al. 2006). One
important set of candidate mechanisms for such
temporally distant effects is the constellation of
pro-inflammatory changes that have been shown
associated with childhood disadvantage. Programs
of work by Miller and Chen (Miller et al. 2009a;
Miller et al. 2009b), Cole (Cole 2009; Cole et al.
2007) and others (Danese et al. 2008; McDade
2012), for example, have demonstrated systematic differences in cytokine signaling pathways,
transcriptional control, and risk for inflammationand immune-mediated disease. These studies suggest that changes in immune responsivity and the
molecular processes involved in inflammation
may be one category of mediating events linking
childhood social adversity to adult disease.
W. Thomas Boyce and C. Hertzman
68
(a) Males
Percent
60
Socioeconomic classification (NS-SEC)
50
40
30
20
10
0
0-15
16-44
45-64
65 & over
45-64
65 & over
Age
(b) Females
Percent
60
Socioeconomic classification (NS-SEC)
50
40
30
20
10
0
0-15
16-44
Age
Large employers and higher managerial
Small employers and own account
Higher professional
Lower supervisory and technical
Lower managerial and professional
Semi-routine
Intermediate
Routine
Note: NS-SEC=National Statistics Socioeconomic Classification
Source: Office for National Statistics
General Household Survey52
Fig. 3 Percentage of males and females with limiting long-term illness, by age and socioeconomic classification
(Marmot 2010)
Early Childhood Health and the Life Course: The State of the Science and Proposed Research Priorities
Finally, there is emerging evidence that experiences of social subordination per se, themselves
prevalent sources of human adversity (Keltner
et al. 2003), may be related to disorders of mental
and physical health, educational failure, and maladaptive behavior, especially in the young. The
works of Pellegrini (Pellegrini et al. 2007, 2010),
Hawley (Hawley 1999), and Fehr (Fehr et al.
2008) and their colleagues, describing the developmental emergence, behavioral signs, and
socioemotional sequelae of childhood dominance
and egalitarianism, have offered important “down
payments” on such a research agenda. More
recently, we have examined linkages between
subordination and maladaptive health outcomes
in a socioeconomically and ethnically diverse
sample of kindergarten children, assessing associations between experiences of dominancesubordination and patterns of maladaptive
behavior (Boyce et al. 2012a). As shown in
Fig. 4, we found that indicators of mental health
in 5-year-old children were stratified within
classroom hierarchies in a manner parallel to that
seen in larger, adult societies. Children occupying subordinate positions within their classrooms
had more depressive symptoms, more frequent
episodes of inattention, fewer positive peer relationships, and less evidence of prosocial behavior. Further, we found that the children most
likely to show low or diminishing health status
Fig. 4 Depression by
social position and
learner-centered
pedagogical practices
(LCPs) at half-SD
increments from the
mean (−1, −0.5, mean,
+0.5, +1 SD) (Boyce
et al. 2012a)
69
were those occupying subordinate social positions who also came from low SES families, and
those least likely to show a diminution in health
came from classrooms with teachers highly
invested in learner-centered pedagogical practices (LCPs). Indeed, as shown in Fig. 4, the link
between rank and behavior nearly disappeared in
classrooms with strong teacher LCPs, suggesting
that classrooms with egalitarian, student-centered
“cultures” produced diminished mental health
effects of hierarchical social ranks.
Such findings—that social subordination is
associated with decrements in mental health,
even within hierarchical, early childhood
groups—are commensurate, moreover, with prior
findings that subjective estimates of social class
may be a stronger predictor of health outcomes
than objective indicators, such as job status,
income, or wealth (Adler et al. 2000; Goodman
et al. 2003; Ostrove et al. 2000), and that dominance status in primate social hierarchies is similarly associated with health, even among captive
animals with equal access to food, open environments, and veterinary care (Abbott et al. 2003;
Cohen et al. 1997; Kaplan et al. 1982; Sapolsky
2005).
A growing body of research thus demonstrates powerful, graded effects of childhood
social stratification and SES on development and
health, both in children and their adult counter-
W. Thomas Boyce and C. Hertzman
70
parts. Such effects may account, at least in part,
for the strikingly uneven distribution of morbidities within human populations. Nonetheless, as
noted by Kessler, Duncan, and colleagues (e.g.,
Kessler et al. 2014), results of large-scale human
experiments challenge assumptions that simply
augmenting financial resources or moving children out of high-poverty neighborhoods will
have salutary effects on health development.
Childhood SES effects are likely mediated by a
broad array of factors that include diet, exposures to toxic environmental agents, differences
in parenting, and access to health care, but substantial evidence also now implicates differential
exposures to stress and adversity as a major
mechanism of SES influence. Acquired differences in neurobiological reactivity to stressors
and pro-inflammatory shifts in immune signaling pathways may also play an important mediating role in SES-health relations. Lastly,
research has also begun to document effects of
social subordination per se on childhood health
outcomes, independent of family or neighborhood SES. Thus, relations between social position within classroom peer hierarchies and
subsequent health outcomes show the same
graded associations with morbidity found in
studies of nations and large human populations.
4.2
Vulnerability, Resilience,
and Neurobiological
Susceptibility
The aggregation of ill health within human populations appears also attributable to constitutional
differences underlying a remarkable variation in
susceptibility to social-environmental influence.
There is now growing evidence for a generalized
sensitivity to social contexts within a human subgroups, i.e., highly sensitive or environmentally
“permeable” individuals showing maladaptive
outcomes in conditions of adversity but unusually positive outcomes in settings characterized
by support, predictability, and protection (see,
e.g., Aron et al. 2012; Belsky et al. 2007; Boyce
2016; Boyce and Ellis 2005; Ellis et al. 2011a).
Such individuals thus show either the least or
most adaptive outcomes within the population,
depending upon the character of the proximal
social contexts in which they are reared. Studies
demonstrating this greater susceptibility of neurobiologically responsive children to both positive and negative aspects of their environments
have implicated a wide variety of:
• Stressors and adversities, including paternal
depression (Cummings et al. 2007), marital
conflict (El-Sheikh 2005; El-Sheikh et al.
2007), parental psychopathology (Shannon
et al. 2007), and overall family distress
(Obradovic et al. 2010)
• Positive environmental features, including
parental warmth (Ellis et al. 1999), beneficial
experiences and exposures (Pluess and Belsky
2013), and supportive interventions (BakermansKranenburg et al. 2008a)
• Defining biological parameters, including
physiological reactivity (e.g., Alkon et al.
2006; Boyce et al. 1995), differences in brain
circuitry (Whittle et al. 2010), and gene polymorphisms (Bakermans-Kranenburg et al.
2008b; Knafo et al. 2011; Manuck et al. 2011)
Most importantly, highly susceptible children
(and adults) show bidirectional effects on outcomes in contrasting highly supportive and
highly stressful settings—not simply an attenuation of negative effects in low stress
circumstances.
Such
differences
in
neurobiological
sensitivity—likely based, in part, on genetic and
epigenetic variation (Boyce and Kobor 2015)—
are figuring prominently in the field’s exploration
of the biological embedding of early stress.
Findings suggesting differential susceptibility
have been replicated in many samples of children, a troop of rhesus macaques (Boyce et al.
1998), and in a series of randomized controlled
trials of socially supportive interventions
(Bakermans-Kranenburg and Van Ijzendoorn
2015; Quas et al. 2004). Neurobiological sensitivity has also been the focus of a ten-paper special section of Development and Psychopathology
Early Childhood Health and the Life Course: The State of the Science and Proposed Research Priorities
71
Fig. 5 Incidence of
violent injuries in free
ranging macaques by
confinement stress and
reactivity status (Boyce
et al. 1998)
Fall Baseline Daily Cortisol
Expression
Met carrier
Val/Val
1.0
0.7
0.4
0.1
-0.2
-0.5
-0.8
- 1 SD
+1 SD
Family Income
Fig. 6 Daily baseline cortisol expression by family SES
and BDNF polymorphism (N. Bush, personal communication, unpublished data, 2017)
(see Ellis et al. 2011a). As shown in the accompanying Figs. 5 and 6, examples have included the
differential rates of violent injuries among high
and low reactivity rhesus macaques before and
during a prolonged period of confinement stress
(Fig. 5) and the level of HPA activation among
kindergartners by allelic variants in the BDNF
gene and family income (Fig. 6). In both cases,
the “risky endophenotype” (i.e., high reactivity
status or being a Met carrier in the BDNF polymorphism) showed high levels of maladaptive
outcomes (injuries or basal cortisol secretion)
under stressful conditions but remarkably low
levels (lower even than their low-risk counterparts) in positive, low stress settings.
Importantly, strong evolutionary arguments
have been made, albeit not without dissension
(e.g., Bateson and Gluckman 2011; Overton
and Lerner 2014; West-Eberhard 2003) for the
conservation of environmental sensitivity
within rearing environments of evolutionary
adaptedness. Slavich and Cole, for example,
have summarized recent evidence on how
social-environmental exposures can regulate
gene expression, thereby calibrating individual
sensitivity to social context (Slavich and Cole
2013). Such sensitivity appears to influence not
only the rates and severity of morbidity but also
the timing and pace of important developmental transitions, such as puberty (Ellis et al.
2011b). This characteristic, which probably
becomes increasingly trait-like over the course
of development, appears to emerge as a conditional adaptation, garnering signals from the
early social environment about the inherent
levels of threat, adversity, support, and nurturance that the growing child is likely to encounter and calibrating stress-responsive biological
systems to optimize survival, health, and developmental well-being (Ellis et al. 2006, 2011a;
Hane and Fox 2006). Understanding phenotypic variation in environmental susceptibility
might logically also play a role in the conceptualization and development of newly individualized approaches to “precision medicine”
(Committee on a Framework for Development
a New Taxonomy of Disease 2011).
W. Thomas Boyce and C. Hertzman
72
4.3
The Topography
of Vulnerability
In addition to their aggregation within population
subgroups, developmental vulnerability and ill
health are also clustered by spatial geography. It
is well known, for example, that of the more than
7 million of the world’s children less than 5 years
of age that die each year, the majority live in the
regions of sub-Saharan Africa and South Asia
(Liu et al. 2012). Equally familiar is the epidemiologic, small area clustering of neurodevelopmental disorders such as autism spectrum
disorder (Mazumdar et al. 2010). Less well
known, but equally compelling, are the dramatic
differences in developmental vulnerability at primary school entry—differences with major
implications for learning and academic success—that are found within major national jurisdictions, such as the Canadian province of British
Columbia (see Fig. 7).
Between 2000 and 2011, the provincial BC
government, with the assistance and oversight of
the Human Early Learning Partnership at UBC
Vancouver, completed four population-based
assessments of developmental health. Assessments
were completed during the kindergarten year, and
developmental health was measured using the
Early Development Instrument (EDI), a kindergarten teacher-completed checklist for each child
based on five scaled measures of development:
physical well-being, social competence, emotional maturity, language and cognitive development, and communication and general knowledge
(Janus and Offord 2000). The EDI yields, for each
child and each scale, a score as “vulnerable” or
“not vulnerable.” The designation as “vulnerable”
is not given directly to the family, however; rather,
rates of EDI vulnerability are computed and
mapped according to residential neighborhoods
where children live. Mapping is done by neighborhoods because local geography defines unique
combinations of factors that support or undermine
early child development.
Across Canadian neighborhoods, vulnerability
rates on one or more of the EDI scales range from
Fig. 7 Neighborhood variation in developmental vulnerability in British Columbia on the early development instrument (http://earlylearning.ubc.ca/interactive-map/)
Early Childhood Health and the Life Course: The State of the Science and Proposed Research Priorities
as low as 4% to as high as 69%, such that there is
a more than 17-fold inequality in developmental
vulnerability in Canada at the level of the neighborhood. This range is much larger than would be
predicted on the basis of random sample surveys
of child development, which rarely demonstrate
social gradients larger than threefold.
Neighborhoods aggregate family environments,
reflect broader environments, and have emergent
properties, such as safety and social cohesion, that
influence developing children. They tend to
include families of similar socioeconomic status,
unique mixes of relational characteristics (i.e., the
factors that shape a family’s social identity), and
similar levels of access or barriers to programs
and services. For analytical purposes, random
sample surveys aggregate children from different
geographical locales into statistical (rather than
real) neighborhoods according to a small number
of grouping factors (e.g., median family income
or proportion of adults with high school graduation). Such “neighborhood effect” analyses from
random sample surveys do not capture the unique
circumstances of real neighborhoods. Thus, the
17-fold variation in EDI-estimated developmental health validates this spatial geographic
approach to local differences, since only this tactic captures the circumstances of real neighborhoods (Hertzman 2010; Kershaw et al. 2009).
Such an approach has been used, for example, to
reveal that optimal, area level EDI scores occur
within neighborhoods with mixed, rather than uniformly affluent, SES representation, i.e., those
with relatively equal proportions of affluent and
disadvantaged families (Carpiano et al. 2009).
Adopted on an even larger scale, such methods
are capable of offering social ecological perspectives on the physical, socioeconomic, and social
political conditions of children on a global scale
(Panter-Brick et al. 2012).
4.4
Marginalization
and Scapegoating
Interpersonal marginalization and scapegoating can
also contribute to uneven distributions of morbidity
and maladaptive development, often based on targeting by individual, constitutional characteristics.
73
Victimization is a well-represented and elemental
human behavior, even within groups of very young
children. Described by Lord Byron as “the longest
pleasure” (Don Juan, 13th Canto: “Now hatred is
by far the longest pleasure; Men love in haste but
they detest at leisure.”), hatred and stigmatization
have remained, for all of history, a sadly defining
feature of human societies. Family systems theory
has regarded scapegoating and the designation of an
“identified patient” as an effective if personally
costly means of securing family solidarity (Vogel
and Bell 1961), much as the vilification of Simon in
William Golding’s Lord of the Flies served the
cohesion of a marooned band of lost school boys.
Behavioral vulnerability stemming from physical
weakness, anxiety, poor social skills, or chronic illness has been shown to engender victimization in
groups of children (Egan and Perry 1998; Sentenac
et al. 2012), and within nonhuman primate troops,
the active targeting of highly reactive, more sensitive members has been described during periods of
intensive group stress (Boyce et al. 1998). Far from
restricted to Western, individualist societies, bullying and victimization are also highly prevalent in
more collectivist cultures (Kim et al. 2004). Among
human children and youth, such victimization can
undermine mental health and lead to poor academic
performance (Glew et al. 2005), psychosomatic disorders (Arseneault et al. 2006; Gini and Pozzoli
2009), suicidal ideation behavior (Kim et al. 2009;
Turner et al. 2012), and later criminality (Sourander
et al. 2007). Among the survivors of populationlevel genocide, specific genetic variants have been
linked to traumatic memory retention and the level
of risk for post-traumatic stress disorder (PTSD) (de
Quervain et al. 2012). Abusive experiences in children, whether at the hands of family or peers,
become itself a social determinant of adult mental
and physical health (Greenfield 2010), resulting in
the sequestering of morbidities within both perpetrators and their victims.
4.5
History and Affliction
As Kuzawa and Thayer noted (Kuzawa and
Thayer 2011), human adaptation to environmental conditions takes place at a variety of timescales, ranging from homeostatic changes that can
W. Thomas Boyce and C. Hertzman
74
occur over seconds or minutes to developmental
plasticity emerging over months or years to conserved genetic changes that operate on a timescale
of millennia. Thus, historical contexts can also
concentrate and skew distributions of disease and
disorder over epochs of time. For example, prenatal exposures to a natural disaster, such as the
1998 Québec Ice Storm, can contribute to the risk
for childhood obesity, even after adjustment for
breastfeeding, SES, obstetrical complications,
and smoking during pregnancy (Dancause et al.
2012). During historical periods of war, elevated
lifetime risks for PTSD are found among children
exposed to violent or traumatic events (Javidi and
Yadollahie 2012). Researchers have also studied
how early life exposures to famine can influence
adult health, using historical cohort data from the
Finnish crop failure famine of 1866–1868, the
Dutch Hunger Winter of 1944–1945, the Siege of
Leningrad in 1944, the seasonal famines in the
Gambia between 1949 and 1994, and the Chinese
Great Leap Forward famine of 1959–1961.
Outcomes examined have included adult height
and weight, glucose metabolism, blood pressure,
lipid profiles, metabolic syndrome, cardiovascular outcomes, self-reported health, mental performance and cognition, mental disorders, and adult
mortality (Falconi et al. 2017; Lumey et al. 2011).
Exposure episodes related to war, such as the
1944–1945 Dutch Famine of World War II,
resulted in elevated odds of developing coronary
heart disease in adult life (Painter et al. 2005;
Roseboom et al. 2000) and in systematic differences in the epigenetic modification of genes such
as insulin-like growth factor (IGF-2) (Heijmans
et al. 2008) and aspects of risk that appear capable
of crossing into unexposed generations (Painter
et al. 2008). Genocide, whether historical in the
case of the Holocaust or contemporary, as in the
Rwandan atrocities, exerts powerful, lasting damage on children’s prospects for health and wellbeing, both contemporaneous and longitudinal
(Hazani and Shasha 2008; Oberg 2008). Each of
these examples illustrates how time, in the form
of historical period, and context, in the form of
broad socioenvironmental perturbations and
assaults, converge to influence and bias life course
health development.
4.6
Critical Periodicity
Finally, events and exposures within developmental time, rather than historical time, also create constraints and opportunities for healthy
development and the avoidance of disorder. The
function of neuronal circuits in the brain is guided
by experiences in postnatal life that regulate the
maturation of inhibitory connections and interneurons (Hensch 2005). Critical periods exist, for
example, for the acquisition of language and discrimination of speech sounds in human infants
(Weikum et al. 2012), and exposure to music can
change auditory preferences in young mice
through changes in prelimbic and infralimbic
medial prefrontal cortex during an early critical
period (Yang et al. 2012). Adult brain plasticity
appears to become restricted by structural and
functional developmental “brakes,” such as perineuronal nets and myelin that inhibit neurite
growth and the balance between excitatory and
inhibitory circuitries (Bavelier et al. 2010).
Genetic, pharmacological, and environmental
influences can alter such plasticity, suggesting
future opportunities for reopening closed critical
periods, such as those for language acquisition or
vision, or enhancing recovery from traumatic or
cerebrovascular brain injury.
5
Interactions
Between Context
and Constitution:
The Biological Embedding
of Early Adversity
These arenas of research, defining the contextual
and constitutional origins of disease maldistribution by population, space, and time, converge in a
new and growing body of work examining the
interplay between genes and environments in
determining developmental and health endpoints
over the life course. Such research comprises
studies of (a) true gene-environment interactions,
in which allelic variation moderates primary
associations between environmental factors
and outcomes, and (b) epigenetic processes, in
which exposure-related chemical/structural
Early Childhood Health and the Life Course: The State of the Science and Proposed Research Priorities
modifications of chromatin up- or downregulate
the transcriptional expression of functionally
salient genes. Together, such studies and larger
territories of research have begun to advance
serious answers, at a variety of analytic levels, to
the questions surrounding how socially partitioned early experience “gets under the skin,”
affecting broad life course trajectories of development and health (Evans et al. 2012; Hertzman
and Boyce 2010; Slavich and Cole 2013).
5.1
Chromatin Modification
and the Molecular Biology
of Epigenesis
As shown in Fig. 8, chromatin is the structural
packaging of DNA and its associated proteins
into nucleosomes: strands of DNA wrapped
around histone protein octamers, like thread
around a spool. Nucleosomes in turn form a
loosely or tightly wound chromatin “necklace,”
arrayed like beads on a string. Epigenetics has
been defined as a functional modification of DNA
that does not involve an alteration of sequence
(Meaney 2010). The differentiation of embryonic
stem cells into specific tissue lineages, for example, is epigenetically guided by patterns of downregulation and activation of lineage-specific
Fig. 8 Chromatin and
nucleosome structure
(Courtesy of Joseph
Roland© 2003 – www.
cytographica.com)
75
genes (Wu and Sun 2006). Figure 9 shows how
DNA methylation changes with development
throughout the sequential ontogeny of germ cell
development, fertilization, and embryogenesis.
The more general processes of gene-environment
interplay involve important functional distinctions between gene sequence, epigenetic modifications, and gene expression—variations in each
of which may influence individual responses to
environmental exposures. Gene sequence variation, for example, has been linked to vulnerability to a variety of maladaptive or disordered
health outcomes (Caspi et al. 2002, 2003).
Differences in gene expression have also been
associated with disease susceptibility (Liew et al.
2006; Mohr and Liew 2007) and are, in turn,
related to a variety of epigenetic, molecular processes, which include DNA methylation, histone
modification, production of noncoding microRNAs, presence of various transcription factors, and
nucleosome remodeling (Gilbert and Epel 2009).
Given the complexities of these chemical mechanisms, however, it is important to understand that
(a) DNA methylation is only one form of epigenetic mark, (b) DNA methylation is not the equivalent of gene expression, and (c) it is sometimes
unclear whether DNA methylation is a precursor
of gene silencing or a marker of such silencing.
For purposes of simplicity, the discussion here
W. Thomas Boyce and C. Hertzman
76
Fig. 9 Epigenetic methylation events in early human gestation (Russo et al. 1999)
focuses primarily upon true gene-environment
interactions and a single form of epigenetic modification, the methylation of cytosine-phosphateguanine (CpG) dinucleotide sites in DNA. The
relative stability of DNA methylation suggests
that, among epigenetic marks, it may be responsible for longer-term, more developmentally important changes in gene expression.
As shown in Fig. 10, genes have both promoter and coding regions, with the latter demarcated by start and stop sequences recognized by
RNA polymerase. RNA polymerase, in association with facilitating transcription factors (i.e., a
“transcription complex”), transcribes the DNA
coding region into messenger RNA, which in
turn undergoes ribosomal translation into proteins that alter cellular function. Social experiences can result in epigenetic marks, such as
CpG methylation, within a gene’s promoter
region, and the degree of such methylation regulates gene expression, like a dimmer switch regulates light in a room. Transcription start sites and
promoters are known to be enriched with CpG
sites (areas called “CpG islands”) (Turner et al.
2008), and experience-induced methylation of
these sites is known to tighten chromatin structure, rendering transcription factor binding sites
physically less accessible and interfering with
RNA polymerase attachment to the coding start
sequence. Thus, the experience-induced methylation of a promoter CpG can effectively silence
the gene, altering cellular function and changing
physiological processes ranging from neurotransmitter reuptake in neuronal synapses to blood
glucose regulation and sympathetic nervous system reactivity.
5.2
The Epigenetics of Social
Adversity
Although the study of epigenetics began in the
field of cancer biology, research has more
recently begun to explore epigenetic processes by
which socially stratified exposures to early adversities may condition long-term risks to development and health (Gluckman et al. 2007; Jirtle and
Skinner 2007; Tsankova et al. 2007). There is
evidence for DNA methylation-associated regulation of adversity-responsive genes in both
Early Childhood Health and the Life Course: The State of the Science and Proposed Research Priorities
77
Fig. 10 Simplified representation of epigenetic modification and transcription regulation. CH3 methyl group, Tag TF
transcription factor, RNA plmrs RNA polymerase, mRNA messenger RNA
laboratory animals (McGowan et al. 2008) and
human infants (Oberlander et al. 2008) and for
stressor-associated differences in mRNA expression in human subjects (Cole et al. 2007; Morita
et al. 2005). Further, the biochemical “markings”
of genomic DNA and its associated, nucleosomal
histone proteins are the epigenetic mechanisms
by which the expression of neuroregulatory genes
is controlled (Isles and Wilkinson 2008; Meaney
and Ferguson-Smith 2010; Mehler 2008).
Epigenetic marks are capable, through this governing of neuroregulatory gene expression, of
guiding both neurodevelopment and the function
of peripheral and central neural pathways (Liu
et al. 2008; MacDonald and Roskams 2009).
The work of Meaney, Szyf, and colleagues
(Szyf et al. 2008; Weaver 2009; Weaver et al.
2004; Zhang and Meaney 2010), for example,
has demonstrated in the rat pup how maternal
licking and grooming in the first several days of
life downregulate pups’ long-term HPA reactivity through increases in hippocampal glucocorticoid receptor (GR, NR3C1) expression and
serotonergic tone. Licking and grooming triggers
increases in serotonin in the hippocampus, and
activation of the 5-HT receptor induces expression of a transcription factor, nerve growth factorinducible protein-A (NGFI-A). Licking and
grooming also facilitates NGFI-A’s association
with the exon 17 GR gene promoter by demethylating a CpG site located in the NGFI-A binding
region of this exon. The resulting increased
expression of GR results in a downregulation of
corticotropin-releasing hormone (CRH) and
diminished activation of the HPA axis. Readers
are referred to (Hackman et al. 2010) for more
detailed description of these molecular events.
Other work in rat models of early infant maltreatment has demonstrated enduring methylation of
the gene coding for brain-derived neurotrophic
factor (BDNF), reduced BDNF expression, and
extension of both epigenetic marks and maltreatment of young into a subsequent generation
(Roth et al. 2009). Because BDNF is a key mediator of neural plasticity in brain regions such as
the prefrontal cortex and hippocampus, such
findings might present a legitimate model of the
neurocognitive sequelae of transgenerational
78
neglect and abuse of human children. Champagne
and colleagues (Champagne et al. 2006) have
also shown, in the same model, that the increased
estrogen receptor-α (ER-α) expression that
occurs in the medial preoptic area of high licking
and grooming mothers is associated with cytosine methylation of the ER-α gene promoter.
Nonhuman primate work by Kinnally et al.
(Kinnally et al. 2010a, 2010b, 2011) has explored,
in infant macaques, interrelations among early
stressors (social group vs. nursery rearing and
variable foraging vs. control conditions), allelic
variation in the 5-HTT serotonin transporter gene
promoter, and both genome-wide and 5-HTTspecific differential methylation in peripheral
blood mononuclear cells (PBMCs). Among their
findings with direct relevance to the research
considered here are the following:
• Higher 5-HTT CpG methylation, but not
sequence variation in the serotonin transporter
promoter region, exacerbates the effects of
early life stress on behavioral stress
reactivity.
• Both greater 5-HTT and whole-genome methylation level (i.e., 5-methylcytosine content) is
associated with enhanced stress reactivity
across the subsequent life span.
• When brain tissue is unavailable for methylation analysis, peripheral blood methylation
levels may serve as useful surrogates (see also
Cupello et al. 2009; Uebelhack et al. 2006).
The Meaney-Szyf research group has also
recently advanced evidence, in the postmortem
hippocampi of human suicide victims, for hypermethylation of the ribosomal RNA gene and
NR3C1 gene promoters and epigenetic marks
suggesting dysregulation in cellular protein synthesis machinery and HPA responsivity
(McGowan et al. 2008, 2009). Further, two prospective human studies have now documented
differential patterns of DNA methylation conditional upon various aspects of early childhood
adversity. First, genome-wide, blood DNA methylation analysis in low- and high-SES subjects
from the 1958 British Birth Cohort revealed a
cluster of probes obtained from the 500 most
W. Thomas Boyce and C. Hertzman
variable promoters that was enriched with highSES individuals, confirming that SES differences
contributed to overall epigenetic variation.
Methylation levels for 1,252 gene promoters
were associated with childhood SES versus 545
promoters for adulthood SES (Borghol et al.
2012). Second, in a subsample of adolescents
from the Wisconsin Study of Family and Work
birth cohort, Essex et al. (Essex et al. 2013) found
differential methylation in 170 CpG sites among
participants whose parents reported high levels of
stress during their children’s early lives. Maternal
stressors in infancy and paternal stressors in the
preschool years were most strongly predictive of
differential methylation, and the patterning of
such epigenetic marks varied by children’s gender. Both birth cohort studies suggest that epigenetic signatures detectable in adolescence to
mid-life are related to early experiences with
stressful environments. Other human research
has documented:
• Altered DNA methylation of the insulin-like
growth factor gene (IGF2) in individuals
exposed prenatally to the Dutch Famine of
1944–1945 (Heijmans et al. 2008)
• Increased NR3C1 methylation at an NGFI-A
binding site in newborns exposed to mothers’
depressed mood in the third trimester of pregnancy (Oberlander et al. 2008)
• Highly variable methylation patterns within
the five GR promoters activated in PBMCs,
possibly reflecting epigenetic fine-tuning of
GR expression by early life experiences
(Turner et al. 2008)
• Pro-inflammatory shifts in cytokine expression among young adults with low-SES childhood histories, via transcriptional signaling
changes in adrenergic and adrenocortical
pathways (Chen et al. 2011; Miller et al.
2009a)
• Both between-tissue and between-individual
discordance in epigenetic marks at the insulinlike growth factor, IGF2/H19, locus in multiple tissues at birth, even among monozygotic
twins (Ollikainen et al. 2010)
• Differences in risk for psychopathology
related to concurrent allelic and epigenetic
Early Childhood Health and the Life Course: The State of the Science and Proposed Research Priorities
variation, such that trauma-related 5-HTT
promoter methylation operates as a moderator
of the association between the 5-HTT polymorphism and psychological problems (van
IJzendoorn et al. 2010)
Taken together, these observations offer evidence for social-environmental regulation of
mammalian and human gene expression, at the
levels of both whole-genome and candidate gene
methylation (Cole 2009). The findings also suggest that divergences in methylation and gene
expression begin early in development, even during intrauterine life, and that socially partitioned
stress and adversity may be transmuted into mental health risks through molecular modifications
of the epigenome. Differential exposures to environmental events may explain, for example, the
diverging trajectories of epigenetic profiles found
among monozygotic twins over time (Fraga et al.
2005). More broadly, epigenetic remodeling in
response to developmentally potent environmental events may serve as a mechanism for neural
plasticity and the conditional production of individual differences in attributes and risks for psychopathology (Bagot and Meaney 2010).
5.3
Gene-Environment Interplay
in Brain Development
Indeed, there is already emerging evidence that
brain processes and neural plasticity may function as key mediators in the linkages between
G-E interplay and developmental and health endpoints. Bush et al., for example, have recently
shown that the BDNF Val66Met polymorphism
confers an increased neuroendocrine sensitivity
to socioeconomic context, with Met carriers having the highest and lowest cortisol expression
levels, depending on SES (N. Bush, personal
communication, unpublished data, 2017). Other
findings in the nascent science of “imaging
genomics”—a field merging high-throughput
genotyping with new brain imaging technologies—are revealing how variations in DNA
sequence are associated with structural and functional connectivity in specific brain regions
79
(Thompson et al. 2010). Recent fMRI studies, for
example, have demonstrated the heritability of
patterns of task-related brain region activation
and shown how the COMT val108/158met functional polymorphism is associated with systematic differences in prefrontal cortical physiology
and function (Egan et al. 2001). Consistent with
the same general hypothesis, Hariri and colleagues (Hariri et al. 2005; Hariri and Weinberger
2003) have found that subjects carrying the less
efficient short allele of the 5-HTT gene promoter
had an increased amygdalar response to fearful
stimuli in comparison with subjects homozygous
for the long allele. Even more to the point in
regard to the development of brain structures subserving socioemotional development, Alexander
et al. (Alexander et al. 2012) recently reported a
significant GxE interaction effect on neural
response patterns and functional amygdalahypothalamus
connectivity—circuitry
that
appears closely tied to emotional control and the
interpretation of social processes (Blakemore
2010; Norman et al. 2012). Such findings begin
to reveal possible neural substrates for previously
observed, epidemiological observations linking
functional polymorphisms, early stressful events,
and vulnerability to psychopathology (Caspi
et al. 2003; Moffitt et al. 2005).
Also coming into view through the work of a
range of investigators (Adolphs 2009; Blakemore
2010, 2012; Lesch 2007; Norman et al. 2012;
Robinson et al. 2005, 2008) is a new corpus of
research describing and exploring the social
brain. Such work has begun to address a neurogenomic basis for complex social cognitions,
including the capacities for inferences about others’ thoughts and emotions (a cognitive ability
referred to in the child development literature as
“theory of mind”), recognition of self, processing
of facial information, differentiation of social
opportunities and challenges, and control of
socially evoked emotion. The substrates for these
capacities are already known to lie, at the neural
circuit level, in functional connectivity between
limbic structures, such as the amygdala, hippocampus, and basal ganglia, and prefrontal cortical regions, including the dorsolateral and
orbitofrontal cortex, the anterior cingulate and
W. Thomas Boyce and C. Hertzman
80
the posterior superior temporal sulcus at the temporoparietal junction. At the molecular level,
there is evidence for perturbations in the functionality of such circuits related to allelic and epigenetic variation in genes such as those coding
for the serotonin transporter (Lesch 2007), oxytocin (Norman et al. 2012), and endorphins
(Keverne et al. 1989). There is to date no single
definition of the social brain, but a consensus
appears to be forming that a subset of autonomic,
neuroendocrine, neural, and genomic processes
influence and are influenced by aspects of social
cognition and behavior (see Robinson et al. 2005,
2008). Although it will be important to avoid a
simplistic attribution of complex cognitive and
perceptual events to discrete neural, endocrine, or
cellular structures (Norman et al. 2012), there is
clearly much to be learned about the nature of
sociality, the mechanisms that underpin it, the
degree to which these mechanisms are conserved
in animal phylogeny, and the implications of perturbations in its development.
In sum, a burgeoning research enterprise is
producing evidence, in both animal models and
human studies, that many if not most human morbidities have their points of origin in early childhood (Shonkoff and Garner 2012), are the
products of gene-environment interplay (Rutter
2006), and influence developing neural circuits
that are directly linked to long-term trajectories
of health, disease, and life achievement. Although
the fields of social epigenetics and developmental neuroscience are yet in their relative infancies,
promising advances in both suggest a scientific
frontier in which functional interactions among
social environments, genetic and epigenetic variation, and the functionality of neural circuits will
offer compelling new knowledge of how developmental variation emerges.
6
Summary: What We Know
and What We Don’t
Deep cultural traditions affirming the special sensibilities of children and the echoing of childhood difficulties and stress into the decades of
adult life are becoming now powerfully grounded
in a new science describing the biological embedding of early social adversity (Boivin and
Hertzman 2012; Boyce et al. 2012a; Hertzman
and Boyce 2010). No longer relegated to categories of either unsupported belief or speculative
hypothesis, the science of early child development has become a multidisciplinary landscape
of novel findings documenting prospective, longitudinal associations, powerful and increasingly
causal understandings of mechanism and mediation, and new levels of observation made possible
by dramatic technological advances. Among the
discrete, foundational discoveries that have
become the products of this science are the
following:
1. Epidemiologic and population-based studies
have soundly documented the propensity for
morbidities and difficulties of all kinds—biomedical diseases, psychiatric disorders, injuries, and academic, professional, and personal
underachievement—to
aggregate
by
population, space, and time, thereby disproportionately affecting small subgroups of
individuals, during constrained periods of historical or developmental time. Thus, especially during periods of strife, war, or
economic decline, those people whose health
and well-being are most harshly and persistently vulnerable are likely to issue from
impoverished, alienated, or otherwise marginalized groups. Misfortune is neither stochastically nor evenly distributed within human
populations, and the inequalities that ensue
from maldistribution are serious, ethical, and
public health challenges to society’s commitments to justice and health equity. Our societal aspirations to create a more “empathic
civilization” (Rifkin 2009) will demand a
more human practice of medicine (Halpern
2001), a broader allegiance to unbiased opportunity (Robert Wood Johnson Foundation
Commission to Build a Healthier America
2009), and a deeper commitment to the rich
pluripotentiality of young lives (Shonkoff and
Phillips 2000).
2. Studies examining the developmental biology
of social adversity have positioned this new
Early Childhood Health and the Life Course: The State of the Science and Proposed Research Priorities
science on the cusp of deep mechanistic
explanations for the important, earlier insights
of Waddington (Waddington 1959, 2012),
Gottlieb (Gottlieb 1991), and others (Gilbert
2002; Karmiloff-Smith 2007; Meaney
2010)—insight that organismic development
is guided by the combined, interactive influences of genes and experiences. We now know
that development includes not only gene
expression regulation through experiential
modification of chromatin structure but also
by real GxE interactions and the joint, interactive effects of allelic variation and chromatin
modification together (see, e.g., Lam et al.
2012; Pezawas et al. 2005; Rutter 2012). As
asserted here and elsewhere, the simple partitioning of developmental variance into genetic
and environmentally determined components
now falls far short of a truly interactive view
of the operation of nature and nurture (Meaney
2010). Coming now steadily into view are the
actual molecular mechanisms by which constitution and context conjointly determine differences in health and development, both in
childhood and over the lifespan.
3. The origins, dimensions, and consequences of
individual differences in phenotype are emerging as essential components in a full understanding of the biology of social adversity.
Biology is replete with both between- and
within-species variation that bears convincing
witness to the evolutionary uses of diversity,
and elegant ethological and epigenetic work,
such as that by Meaney (Meaney 2010), Szyf
(McGowan and Szyf 2010), and Suomi
(Suomi 2006), reveals the adaptive benefits of
phenotypic diversification. There is the nearly
tautological reality that the operation of natural selection demands the genetic diversity
achieved through spontaneous mutation, but
beyond such self-evident principles, there are
clear examples of how species diversification
enhances survival and fitness. Thus, maternally and epigenetically regulated differentiation of rat pups’ adrenocortical responsivity
produces a range of low to high reactivity phenotypes, each of which may maximize survival and fitness within particular, early and
81
later life environments (Weaver et al. 2004).
Similarly, neither the aggressively uninhibited
nor the shy, neophobic phenotypes of young
rhesus macaques can be warranted as “normal” or optimal; rather, each has adaptive
value within specific social and physical contexts (Cirulli et al. 2009; Stevens et al. 2009).
What is salient and important about phenotypic variants is their capacity for enhancing
fitness within the diversity of species-typical
environments encountered. One illustrative
variant are those children and young nonhuman primates that evince an exceptional neurobiological susceptibility to aspects of social
settings, a phenotype likely representing a
conditional adaptation to early environmental
signals (Barr et al. 2004; Belsky 2005; Boyce
and Ellis 2005; Ellis et al. 2011a). Such individuals show a heightened risk of morbidity
under conditions of stress and adversity but
exceptionally good health and positive
development in settings characterized by support, nurturance, and stability.
4. Increasingly visible within the emerging literature on the neurobiological consequences
of early life adversity is a new collection of
studies and papers examining the social brain
(Adolphs 2009; Blakemore 2010, 2012; Lesch
2007; Norman et al. 2012; Robinson et al.
2005, 2008). The brain has evolved to acquire
specific organizational circuits, cortical
regions, and subcortical structures dedicated
to the detection of survival threats, and social
experiences of loneliness, abandonment, or
neglect may chronically activate such circuitry, creating long-term perceptual and
affective biases and fundamentally altering a
child’s view of the social world (Eisenberger
and Cole 2012). Such circuitry is also represented in the periphery, with both animal
(Insel 2010) and human (Norman et al. 2012)
studies documenting effects on social affiliative processes mediated in part by hormonal
events such as the expression of oxytocin and
vasopressin.
5. As also discussed, a remarkable diversity of
early, social environmental dimensions has
been linked to important differences in mental
W. Thomas Boyce and C. Hertzman
82
and physical health, trajectories of development, and individual differences in behavior.
Such dimensions include, but are not limited
to, acute and chronic stressors, poverty and
subjective social marginalization, and the
absence of positive contextual factors, such as
good parenting or a child-supportive community. Of apparently particular importance to
social epidemiologic perspectives on early
development are studies describing and documenting the effects of social hierarchies,
structural subordination, bullying, discrimination, and victimization. The hierarchical and
networked social structures found across phylogeny—literally from fruit flies (fish and primates) to human kindergartners (see Boyce
et al. 2012a; Fernald and Maruska 2012;
Schneider et al. 2012)—suggest an evolved
predisposition with implications for survival,
reproduction, and safety. Although such hierarchies may be a heritable legacy from our
evolutionary past, promising new work
addresses approaches to minimizing the health
effects of subordinate social positions at the
levels of societies, communities, and schools.
Such work also attests to the individual and
societal benefits of fostering the development
of empathy, altruism, and sociality.
6. The central role of time—evolutionary, historical, developmental, neurogenomic, and
neurophysiological—in determining phenotypic variation is another recurrent if often
implicit theme in emerging developmental
science. Much of the biological embedding of
current social contexts reflects response predispositions established, selectively and epigenetically, through adaptations to the
temporally distant environments of early
hominids (Dubos 1965; Nesse and Young
2000). Social disparities in health—products,
in part, of the social, economic, and health
policies of contemporary societies—wax and
wane within historical time according to the
era’s dominant sociopolitical philosophies
(Beckfield and Krieger 2009; Krieger 2001).
Further, developmental time is strikingly
uneven in its potency, intensity of change, and
accessibility to environmental influence.
Thus, at quite different levels of temporal resolution, time and timing appear to play crucial, but not yet fully explored, roles in guiding
societal, organismic, and neurobiological
responses to the conditions of early life.
Taken together, these points of emerging
empirical evidence form a constellation of discoveries defining a new and lively research field
and the social and developmental biology of early
adversity and its influences on life course development and health. Although substantial recent
progress has been made within a broad assortment of disciplines, much new research, conceptual integration, and thought remains to be
accomplished. The promise of this new field,
however, lies in a stunning prospect that understanding the social determinants of morbidities in
childhood might unlock new approaches to the
prevention and treatment of disorders over the
entire life course. A final agenda for future
research now outlines a set of proposals for new,
promising, and possibly heuristic directions of
investigation.
7
An Agenda for Future
Research
1. As the fields reporting various forms of interplay between genes and environments exponentially grow, what is now needed are
programs of research examining the questions
of how and by what mechanisms genes and
early social contexts co-determine trajectories
of behavioral and biological development.
With respect to differences in complex behavior and its disorders, a focus on proximal, neurobiological processes must come strongly to
the foreground. Although pursuing the epigenetic pathogenesis of early psychiatric and
other disorders is a powerful first step
(Docherty and Mill 2008; Robison and Nestler
2011), it must be followed by studies examining the brain structures and neural processes
that mediate GxE associations with mental
and physical health (Ladd-Acosta et al. 2007;
Turecki et al. 2012). As new knowledge of the
Early Childhood Health and the Life Course: The State of the Science and Proposed Research Priorities
interactive genetic and environmental influences on social brain development becomes
available, a major challenge will be the integration of mechanistic observations across
levels of analysis and scale (Norman et al.
2012).
2. A systematic and useful biology of social
adversity will necessarily involve not only a
search for the mechanisms (i.e., mediators)
underpinning associations among stress,
development, and morbidity but also the
effects of modifiers (i.e., moderators) that
reveal when, at what ages, or in what subgroups such associations hold (Baron and
Kenny 1986; Kraemer et al. 2001, 2008).
Understanding the mechanistic processes by
which an environmental exposure is linked to
disordered development can be a powerful aid
to elucidating pathogenesis (e.g., the role of
high-density lipoprotein transport of cholesterol as a mediator of the association between
dietary fat and coronary heart disease (Lloyd
et al. 2012)) and imagining novel interventions (e.g., changing mother-infant relationships as a means of altering the association
between poverty and child development (Olds
et al. 2010)). On the other hand, grasping
mediational linkages may be a necessary but
insufficient condition for understanding causation, and the parsing of populations into
subgroups of varying exposure susceptibility,
through the discovery of moderator variables,
can also advance comprehension and the tractability of a given association (e.g., changes in
the potency of stress-illness associations by
differences in individual sensitivity to social
contextual effects (Belsky and Pluess 2009;
Boyce and Ellis 2005; Ellis et al. 2011a)).
3. As attention to epigenetic development grows,
the capacity to place a finer, more exacting
point on the specific kinds of environments
that interact with particular allelic and epigenetic variants will be important and likely illuminating. Evolutionary perspectives on social
adversity will also be essential to understand
how difficult “environments of evolutionary
adaptedness” shaped human and infra-human
biology (e.g., Boyce and Ellis 2005; Sapolsky
83
2003), how contextual stressors might have
generated species diversity (Wolf et al. 2005),
and how adversity may have contributed to the
emergence of social cooperation (Andras
et al. 2007). We need far finer and more precise renditions of the social-environmental
dimensions that interact with genes and produce negative and positive outcomes salient to
population and public health. An important
subtext to such work is the extraordinary value
of cross-species, animal-human, comparative
studies, which together not only inform the
evolutionary biology of social environments
and its consequences but also enable experimental studies capable of bolstering the causal
inferences disallowed by human research.
4. Successfully pursuing a new developmental
science of childhood adversity will surely also
involve the perspective of complex adaptive
systems. Social causation is nonlinear, rather
than Newtonian, in character. Traditional
epidemiologic strategies for understanding
the health effects of social-environmental factors involve the ascertainment of such factors’
“independent” influences on a health endpoint
through the use of multiple hierarchical
regression models (Diez Roux 2007).
Although such an approach allows estimation
of the isolated effects of single independent
variables, it belies the reality that most human
disorders are etiologically complex, with multiple interacting “causes.” Even detecting GxE
interactions almost certainly underserves the
true complexity of pathogenic processes,
because allelic variation in a single gene likely
interacts with polymorphisms in many other
genes, and multiple dimensions of the environment may also interactively influence outcomes. In such circumstances—circumstances
that may eventually prove to predominate in
disease causation—the use of more sophisticated models and analytic tools may be
required to understand the multiply interactive
networks of risk factors involved in the ontogeny of disordered development and health
(Kauffman 1993; Koopman and Lynch 1999).
If so, one such approach with increasingly
demonstrable efficacy is the use of complex
W. Thomas Boyce and C. Hertzman
84
systems analysis, involving descriptive inventories of system components, nonlinear mathematical modeling, and the construction of
agent-based models of causal networks (Diez
Roux 2007; Galea et al. 2010).
5. Finally, as highlighted by Garner, Shonkoff,
and others (Garner et al. 2012; Shonkoff
2012), prevention science is powerfully in
need of new ideas for and approaches to the
design of interventions based on the emerging
science of early development. The NurseFamily Partnership program conceived and
elegantly studied by David Olds and colleagues (see http://www.nursefamilypartnership.org), for example, departed dramatically
from prior, outmoded early childhood enrichment programs by envisioning the motherinfant dyad as the unit of intervention and by
using nurses as home visitors to effectively
place the intervention within a medical frame
of reference. Nonetheless, even more radical
departures are needed from the conventions of
traditional early development programs,
including careful consideration of how complex dynamic systems thinking might be wed
to novel, preventive interventions. Attending
closely to discoveries emanating from the disciplines and sciences surveyed here could
potentially prompt approaches to targeting
and conceiving imaginative interventions with
far greater efficacy and whole population
effects. However complex and challenging
that task might be, all such efforts could be
potentially rewarded with a level of health and
well-being for our children and grandchildren
that is more enduring, robust, and enabling.
The new developmental biology of early life
effects on health development over the life course
is a now flourishing science, nearing readiness to
transform conventional understandings of disease ontogeny, to foster clearer and more vivid
perspectives on the temporally distant effects of
childhood events, and to broaden and transform
approaches to ensuring the health of our children
and the developmental sturdiness of their futures.
Although much careful and innovative thought
will be a prerequisite for such a demanding journey, the road ahead looks promising, indeed.
References
Abbott, D. H., Keverne, E. B., Bercovitch, F. B., Shively,
C. A., Mendoza, S. P., Saltzman, W., et al. (2003). Are
subordinates always stressed? A comparative analysis
of rank differences in cortisol levels among primates.
Hormones and Behavior, 43(1), 67–82.
Adler, N. E., Boyce, W. T., Chesney, M. A., Folkman, S.,
& Syme, S. L. (1993). Socioeconomic inequalities in
health: No easy solution. JAMA, 269(24), 3140–3145.
Adler, N. E., Boyce, W. T., Chesney, M. A., Cohen, S.,
Folkman, S., Kahn, R. L., & Syme, S. L. (1994).
Socioeconomic status and health: The challenge of the
gradient. The American Psychologist, 49(1), 15–24.
Adler, N. E., Epel, E. S., Castellazzo, G., & Ickovics, J. R.
(2000). Relationship of subjective and objective social
status with psychological and physiological functioning: Preliminary data in healthy, White women. Health
Psychology, 19(6), 586–592.
Adolphs, R. (2009). The social brain: Neural basis of social
knowledge. Annual Review of Psychology, 60, 693–
716. doi:10.1146/annurev.psych.60.110707.163514.
Alexander, N., Klucken, T., Koppe, G., Osinsky, R., Walter,
B., Vaitl, D., et al. (2012). Interaction of the serotonin
transporter-linked polymorphic region and environmental adversity: Increased amygdala-hypothalamus
connectivity as a potential mechanism linking neural
and endocrine hyperreactivity. Biological Psychiatry,
72(1), 49–56. doi:10.1016/j.biopsych.2012.01.030.
Alkon, A., Lippert, S., Vujan, N., Rodriquez, M. E.,
Boyce, W. T., & Eskenazi, B. (2006). The ontogeny of
autonomic measures in 6- and 12-month-old infants.
Developmental Psychobiology, 48(3), 197–208.
doi:10.1002/dev.20129.
Andras, P., Lazarus, J., & Roberts, G. (2007).
Environmental adversity and uncertainty favour
cooperation. BMC Evolutionary Biology, 7, 240.
doi:10.1186/1471-2148-7-240.
Ariès, P. (1962). Centuries of childhood: A social history
of family life. New York: Alfred A. Knopf.
Aron, E. N., Aron, A., & Jagiellowicz, J. (2012). Sensory
processing sensitivity: A review in the light of the
evolution of biological responsivity. Personality
and Social Psychology Review, 16(3), 262–282.
doi:10.1177/1088868311434213.
Arseneault, L., Walsh, E., Trzesniewski, K., Newcombe,
R., Caspi, A., & Moffitt, T. E. (2006). Bullying victimization uniquely contributes to adjustment problems
in young children: A nationally representative cohort
study. Pediatrics, 118(1), 130–138. doi:10.1542/
peds.2005-2388.
Bagot, R. C., & Meaney, M. J. (2010). Epigenetics and
the biological basis of gene x environment interactions. Journal of the American Academy of Child and
Adolescent Psychiatry, 49(8), 752–771. doi:10.1016/j.
jaac.2010.06.001.
Bakermans-Kranenburg, M. J., & Van Ijzendoorn,
M. H. (2015). The hidden efficacy of interventions: Genexenvironment experiments from a
differential susceptibility perspective. Annual
Early Childhood Health and the Life Course: The State of the Science and Proposed Research Priorities
Review of Psychology, 66, 381–409. doi:10.1146/
annurev-psych-010814-015407.
Bakermans-Kranenburg, M. J., Van Ijzendoorn, M. H.,
Mesman, J., Alink, L. R., & Juffer, F. (2008a). Effects
of an attachment-based intervention on daily cortisol
moderated by dopamine receptor D4: A randomized
control trial on 1- to 3-year-olds screened for externalizing behavior. Development and Psychopathology,
20(3), 805–820. doi:10.1017/S0954579408000382.
Bakermans-Kranenburg, M. J., Van Ijzendoorn, M. H.,
Pijlman, F. T., Mesman, J., & Juffer, F. (2008b).
Experimental evidence for differential susceptibility: Dopamine D4 receptor polymorphism (DRD4
VNTR) moderates intervention effects on toddlers'
externalizing behavior in a randomized controlled
trial. Developmental Psychology, 44(1), 293–300.
doi:10.1037/0012-1649.44.1.293.
Barker, D. J. (1990). The fetal and infant origins of adult
disease. BMJ, 301(6761), 1111.
Barker, D. J., & Bagby, S. P. (2005). Developmental antecedents of cardiovascular disease: A historical perspective. Journal of the American Society of Nephrology,
16(9), 2537–2544. doi:10.1681/ASN.2005020160.
Baron, R., & Kenny, D. (1986). The moderator-mediator
variable distinction in social psychological research:
Conceptual, strategic, and statistical considerations.
Journal of Personality and Social Psychology, 51(6),
1173–1182.
Barr, R. G., Konner, M., Bakeman, R., & Adamson, L.
(1991). Crying in !Kung San infants: A test of the cultural specificity hypothesis. Developmental Medicine
and Child Neurology, 33(7), 601–610.
Barr, C. S., Newman, T. K., Lindell, S., Shannon, C.,
Champoux, M., Lesch, K. P., et al. (2004). Interaction
between serotonin transporter gene variation and rearing condition in alcohol preference and consumption
in female primates. Archives of General Psychiatry,
61(11), 1146–1152.
Bateson, P., & Gluckman, P. (2011). Plasticity, robustness, development and evolution. Cambridge CB2
8RU, UK: Cambridge University Press, The Edinburgh
Building.
Bavelier, D., Levi, D. M., Li, R. W., Dan, Y., & Hensch,
T. K. (2010). Removing brakes on adult brain plasticity: From molecular to behavioral interventions. The
Journal of Neuroscience, 30(45), 14964–14971.
Beckfield, J., & Krieger, N. (2009). Epi + demos + cracy:
Linking political systems and priorities to the magnitude of health inequities--evidence, gaps, and a
research agenda. Epidemiologic Reviews, 31, 152–
177. doi:10.1093/epirev/mxp002.
Belsky, J. (2005). Differential susceptibility to rearing
influence: An evolutionary hypothesis and some evidence. In B. J. Ellis & D. F. Bjorklund (Eds.), Origins
of the social mind: Evolutionary psychology and child
development (pp. 139–163). New York: Guilford.
Belsky, J., & Pluess, M. (2009). Beyond diathesis stress:
Differential susceptibility to environmental influences.
Psychological Bulletin, 135(6), 885–908. doi:10.1037/
a0017376.
85
Belsky, J., Bakermans Kranenburg, M. J., & Van
IJzendoorn, M. H. (2007). For better and for worse:
Differential susceptibility to environmental influences.
Current Directions in Psychological Science, 16(6),
300–304.
Blakemore, S. J. (2010). The developing social brain:
Implications for education. Neuron, 65(6), 744–747.
doi:10.1016/j.neuron.2010.03.004.
Blakemore, S. J. (2012). Development of the social brain in
adolescence. Journal of the Royal Society of Medicine,
105(3), 111–116. doi:10.1258/jrsm.2011.110221.
Blumenshine, P., Egerter, S., Barclay, C. J., Cubbin, C.,
& Braveman, P. A. (2010). Socioeconomic disparities in adverse birth outcomes: A systematic review.
American Journal of Preventive Medicine, 39(3),
263–272. doi:10.1016/j.amepre.2010.05.012.
Bock, G. R., & Whelan, J. (Eds.). (1991). The childhood
environment and adult disease. West Sussez: Ciba
Foundation.
Boivin, M., & Hertzman, C. (2012). Early childhood
development: Adverse experiences and developmental
health. Ottawa: Royal Society of Canada.
Borghol, N., Suderman, M., McArdle, W., Racine, A.,
Hallett, M., Pembrey, M., et al. (2012). Associations
with early life socio-economic position in adult DNA
methylation. International Journal of Epidemiology,
41(1), 62–74.
Bowlby, J. (1969). Attachment and loss. London: Hogarth.
Boyce, W. T. (1985). Stress and child health: An overview.
Pediatric Annals, 14(8), 539–542.
Boyce, W. T. (2016). Differential susceptibility of the
developing brain to contextual adversity and stress.
Neuropsychopharmacology Reviews, 41(1), 142–162.
Boyce, W. T., & Ellis, B. J. (2005). Biological sensitivity to context: I. An evolutionary-developmental theory of the origins and functions of stress reactivity.
Development and Psychopathology, 17(2), 271–301.
Boyce, W. T., & Kobor, M. S. (2015). Development and
the epigenome: The ‘synapse’ of gene-environment
interplay. Developmental Science, 18(1), 1–23.
doi:10.1111/desc.12282.
Boyce, W. T., Chesney, M., Alkon-Leonard, A., Tschann,
J., Adams, S., Chesterman, B., et al. (1995).
Psychobiologic reactivity to stress and childhood
respiratory illnesses: Results of two prospective studies. Psychosomatic Medicine, 57, 411–422.
Boyce, W. T., O'Neill-Wagner, P., Price, C. S., Haines,
M., & Suomi, S. J. (1998). Crowding stress and violent injuriesamong behaviorally inhibited rhesus
macaques. Health Psychology, 17(3), 285–289.
Boyce, W. T., Den Besten, P. K., Stamperdahl, J., Zhan,
L., Jiang, Y., Adler, N. E., & Featherstone, J. D.
(2010). Social inequalities in childhood dental caries:
The convergent roles of stress, bacteria and disadvantage. Social Science & Medicine, 71(9), 1644–1652.
doi:10.1016/j.socscimed.2010.07.045.
Boyce, W. T., Obradović, J., Bush, N., Stamperdahl, J.,
Kim, Y. S., & Adler, N. (2012a). Social stratification,
classroom 'climate' and the behavioral adaptation of
kindergarten children. Proceedings of the National
Academy of Sciences, 109(Suppl 2), 17168–17173.
86
Boyce, W. T., Sokolowski, M. B., & Robinson, G. E.
(2012b). Toward a new biology of social adversity.
Proceedings of the National Academy of Sciences of
the United States of America, 109(Suppl 2), 17143–
17148. doi:10.1073/pnas.1121264109.
Brim, O. G., & Kagan, J. (Eds.). (1980). Constancy and
change in human development. Cambridge, MA:
Harvard University Press.
Bronfenbrenner, U., & Morris, P. A. (2006). The bioecological model of human development. In R. M. Lerner
& W. Damon (Eds.), Handbook of child psychology:
Vol. 1. Theoretical models of human development (6th
ed., pp. 793–828). Hoboken: John Wiley & Sons, Inc.
Brown, R. L. (2010). Epidemiology of injury and
the impact of health disparities. Current Opinion
in Pediatrics, 22(3), 321–325. doi:10.1097/
MOP.0b013e3283395f13.
Brown, D. W., Anda, R. F., Tiemeier, H., Felitti,
V. J., Edwards, V. J., Croft, J. B., & Giles, W. H.
(2009). Adverse childhood experiences and the
risk of premature mortality. American Journal of
Preventive Medicine, 37(5), 389–396. doi:10.1016/j.
amepre.2009.06.021.
Carpiano, R. M., Lloyd, J. E., & Hertzman, C. (2009).
Concentrated affluence, concentrated disadvantage,
and children’s readiness for school: A populationbased, multi-level investigation. Social Science
& Medicine, 69(3), 420–432. doi:10.1016/j.
socscimed.2009.05.028.
Caspi, A., McClay, J., Moffitt, T. E., Mill, J., Martin, J.,
Craig, I. W., et al. (2002). Role of genotype in the
cycle of violence in maltreated children. Science,
297(5582), 851–854.
Caspi, A., Sugden, K., Moffitt, T. E., Taylor, A., Craig,
I. W., Harrington, H., et al. (2003). Influence of life
stress on depression: Moderation by a polymorphism
in the 5-HTT gene. Science, 301(5631), 386–389.
Champagne, F. A., Weaver, I. C., Diorio, J., Dymov, S.,
Szyf, M., & Meaney, M. J. (2006). Maternal care
associated with methylation of the estrogen receptoralpha1b promoter and estrogen receptor-alpha expression in the medial preoptic area of female offspring.
Endocrinology, 147(6), 2909–2915. doi:10.1210/
en.2005-1119.
Chedgzoy, K., Greenhalgh, S., & Shaughnessy, R.
(2007). Shakespeare and childhood. Cambridge, UK:
Cambridge University Press.
Chen, E., Miller, G. E., Kobor, M. S., & Cole, S. W.
(2011). Maternal warmth buffers the effects of low
early-life socioeconomic status on pro-inflammatory
signaling in adulthood. Molecular Psychiatry, 16(7),
729–737. doi:10.1038/mp.2010.53.
Cirulli, F., Francia, N., Berry, A., Aloe, L., Alleva, E.,
& Suomi, S. J. (2009). Early life stress as a risk factor for mental health: Role of neurotrophins from
rodents to non-human primates. Neuroscience and
Biobehavioral Reviews, 33(4), 573–585. doi:10.1016/j.
neubiorev.2008.09.001.
Cohen, S., Line, S., Manuck, S. B., Rabin, B. S., Heise,
E. R., & Kaplan, J. R. (1997). Chronic social stress,
social status, and susceptibility to upper respiratory
W. Thomas Boyce and C. Hertzman
infections in nonhuman primates [see comments].
Psychosomatic Medicine, 59(3), 213–221.
Cohen, S., Janicki-Deverts, D., Chen, E., & Matthews,
K. A. (2010). Childhood socioeconomic status and adult
health. Annals of the New York Academy of Sciences,
1186, 37–55. doi:10.1111/j.1749-6632.2009.05334.x.
Cole, S. W. (2009). Social regulation of human gene expression. Current Directions in Psychological Science, 18(3),
132–137. doi:10.1111/j.1467-8721.2009.01623.x.
Cole, S. W., Hawkley, L. C., Arevalo, J. M., Sung, C. Y.,
Rose, R. M., & Cacioppo, J. T. (2007). Social regulation of gene expression in human leukocytes. Genome
Biology, 8(9), R189. doi:10.1186/gb-2007-8-9-r189.
Committee on a Framework for Development a New
Taxonomy of Disease. (2011). Toward precision medicine: Building a knowledge network for biomedical
research and a new taxonomy of disease. Washington,
DC: National Academies Press.
Cummings, E. M., El-Sheikh, M., Kouros, C. D.,
& Keller, P. S. (2007). Children's skin conductance reactivity as a mechanism of risk in the context of parental depressive symptoms. Journal of
Child Psychology and Psychiatry, 48(5), 436–445.
doi:10.1111/j.1469-7610.2006.01713.x.
Cupello, A., Albano, C., Gatta, E., Scarrone, S., Villa, E., &
Zona, G. (2009). Binding of paroxetine to the serotonin
transporter in membranes from different cells, subcellular fractions and species. Neurochemical Research,
34(2), 255–259. doi:10.1007/s11064-008-9764-z.
Curley, J. P., Jensen, C. L., Mashoodh, R., & Champagne,
F. A. (2011). Social influences on neurobiology
and behavior: Epigenetic effects during development. Psychoneuroendocrinology, 36(3), 352–371.
doi:10.1016/j.psyneuen.2010.06.005.
Cushing, B. S., & Kramer, K. M. (2005). Mechanisms
underlying epigenetic effects of early social experience: The role of neuropeptides and steroids.
Neuroscience and Biobehavioral Reviews, 29(7),
1089–1105. doi:10.1016/j.neubiorev.2005.04.001.
Dancause, K. N., Laplante, D. P., Fraser, S., Brunet, A.,
Ciampi, A., Schmitz, N., & King, S. (2012). Prenatal
exposure to a natural disaster increases risk for obesity
in 5(1/2)-year-old children. Pediatric Research, 71(1),
126–131. doi:10.1038/pr.2011.18.
Danese, A., Moffitt, T. E., Pariante, C. M., Ambler, A.,
Poulton, R., & Caspi, A. (2008). Elevated inflammation levels in depressed adults with a history of childhood maltreatment. Archives of General Psychiatry,
65(4), 409–416.
de Quervain, D. J., Kolassa, I. T., Ackermann, S., Aerni,
A., Boesiger, P., Demougin, P., et al. (2012). PKCalpha
is genetically linked to memory capacity in healthy
subjects and to risk for posttraumatic stress disorder
in genocide survivors. Proceedings of the National
Academy of Sciences of the United States of America,
109(22), 8746–8751. doi:10.1073/pnas.1200857109.
Diamond, J. (2012). The world until yesterday: What
can we learn from traditional societies. New York:
Penguin Group.
Diaz, C., Starfield, B., Holtzman, N., Mellits, E. D.,
Hankin, J., Smalky, K., & Benson, P. (1986). Ill health
Early Childhood Health and the Life Course: The State of the Science and Proposed Research Priorities
and use of medical care: Community-based assessment of morbidity in children. Medical Care, 24(9),
848–856.
Diez Roux, A. V. (2007). Integrating social and biologic
factors in health research: A systems view. Annals
of Epidemiology, 17(7), 569–574. doi:10.1016/j.
annepidem.2007.03.001.
Docherty, S., & Mill, J. (2008). Epigenetic mechanisms as
mediators of environmental risks for psychiatric disorders. Psychiatry, 7(12), 500–506.
Dowd, J. B., Zajacova, A., & Aiello, A. (2009). Early origins of health disparities: Burden of infection, health,
and socioeconomic status in U.S. children. Social
Science & Medicine, 68(4), 699–707. doi:10.1016/j.
socscimed.2008.12.010.
Dubos, R. J. (1965). Man adapting. New Haven: Yale
University Press.
Dubos, R., Savage, D., & Schaedler, R. (1966). Biological
freudianism. Lasting effects of early environmental
influences. Pediatrics, 38(5), 789–800.
Dunn, E. C., Uddin, M., Subramanian, S. V., Smoller,
J. W., Galea, S., & Koenen, K. C. (2011). Research
review: Gene-environment interaction research in
youth depression – a systematic review with recommendations for future research. Journal of Child
Psychology and Psychiatry, 52(12), 1223–1238.
doi:10.1111/j.1469-7610.2011.02466.x.
Egan, S. K., & Perry, D. G. (1998). Does low self-regard
invite victimization? Developmental Psychology,
34(2), 299–309.
Egan, M. F., Goldberg, T. E., Kolachana, B. S., Callicott,
J. H., Mazzanti, C. M., Straub, R. E., et al. (2001).
Effect of COMT Val108/158 Met genotype on frontal
lobe function and risk for schizophrenia. Proceedings
of the National Academy of Sciences of the United
States of America, 98(12), 6917–6922. doi:10.1073/
pnas.111134598.
Eisenberger, N. I., & Cole, S. W. (2012). Social neuroscience and health: Neurophysiological mechanisms linking social ties with physical health. Nature
Neuroscience, 15(5), 669–674. doi:10.1038/nn.3086.
Ellis, B. J., McFadyen-Ketchum, S., Dodge, K. A., Pettit,
G. S., & Bates, J. E. (1999). Quality of early family
relationships and individual differences in the timing
of pubertal maturation in girls: A longitudinal test of
an evolutionary model. Journal of Personality and
Social Psychology, 77(2), 387–401.
Ellis, B. J., Jackson, J. J., & Boyce, W. T. (2006). The
stress response systems: Universality and adaptive
individual differences. Developmental Review, 26(2),
175–212.
Ellis, B. J., Boyce, W. T., Belsky, J., BakermansKranenburg, M. J., & van Ijzendoorn, M. H. (2011a).
Differential susceptibility to the environment: An evolutionary--neurodevelopmental theory. Development
and Psychopathology, 23(1), 7–28. doi:10.1017/
S0954579410000611.
Ellis, B. J., Shirtcliff, E. A., Boyce, W. T., Deardorff,
J., & Essex, M. J. (2011b). Quality of early family
relationships and the timing and tempo of puberty:
Effects depend on biological sensitivity to context.
87
Development and Psychopathology, 23(1), 85–99.
doi:10.1017/S0954579410000660.
El-Sheikh, M. (2005). Stability of respiratory sinus
arrhythmia in children and young adolescents: A longitudinal examination. Developmental Psychobiology,
46(1), 66–74. doi:10.1002/dev.20036.
El-Sheikh, M., Keller, P. S., & Erath, S. A. (2007). Marital
conflict and risk for child maladjustment over time:
Skin conductance level reactivity as a vulnerability
factor. Journal of Abnormal Child Psychology, 35(5),
715–727. doi:10.1007/s10802-007-9127-2.
Essex, M. J., Thomas Boyce, W., Hertzman, C., Lam,
L. L., Armstrong, J. M., Neumann, S. M., & Kobor,
M. S. (2013). Epigenetic vestiges of early developmental adversity: Childhood stress exposure and
DNA methylation in adolescence. Child Development,
84(1), 58–75. doi:10.1111/j.1467-8624.2011.01641.x.
Evans, G. W. (2004). The environment of childhood poverty. The American Psychologist, 59(2), 77–92.
Evans, G. W., & Kim, P. (2007). Childhood poverty and
health: Cumulative risk exposure and stress dysregulation. Psychological Science, 18(11), 953–957.
doi:10.1111/j.1467-9280.2007.02008.x.
Evans, G. W., Gonnella, C., Marcynyszyn, L. A., Gentile,
L., & Salpekar, N. (2005). The role of chaos in poverty and children's socioemotional adjustment.
Psychological Science, 16(7), 560–565.
Evans, G. W., Chen, E., Miller, G., & Seeman, T. (2012).
How poverty gets under the skin: A life-course perspective. In V. Maholmes & R. B. King (Eds.), The
Oxford handbook of poverty and child development
(pp. 13–36). Oxford: Oxford University Press.
Falconi, A. M., Catalano, R., & Boyce, W. T. (2017). Early
life predictors of late life health. In W. Satariano &
M. Maus (Eds.), Aging, Place, and Health: A Global
Perspective. Burlington: Jones & Bartlett Learning.
Fehr, E., Bernhard, H., & Rockenbach, B. (2008).
Egalitarianism in young children. Nature, 454(7208),
1079–1083. doi:10.1038/nature07155.
Fernald, R. D., & Maruska, K. P. (2012). How does social
information change the brain? PNAS.
Fraga, M. F., Ballestar, E., Paz, M. F., Ropero, S., Setien,
F., Ballestar, M. L., et al. (2005). Epigenetic differences arise during the lifetime of monozygotic twins.
Proceedings of the National Academy of Sciences of
the United States of America, 102(30), 10604–10609.
Freud, S. (1940). An outline of psychoanalysis. In
J. Strachey (Ed.), The standard edition of the complete
psychological works of Sigmund Freud (Vol. 23).
London: Hogarth Press.
Galea, S., Riddle, M., & Kaplan, G. A. (2010). Causal
thinking and complex system approaches in epidemiology. International Journal of Epidemiology, 39(1),
97–106.
Galobardes, B., Lynch, J. W., & Davey Smith, G. (2004).
Childhood socioeconomic circumstances and causespecific mortality in adulthood: Systematic review and
interpretation. Epidemiologic Reviews, 26, 7–21.
Garner, A. S., Shonkoff, J. P., Siegel, B. S., Dobbins,
M. I., Earls, M. F., Garner, A. S., et al. (2012). Early
childhood adversity, toxic stress, and the role of the
88
pediatrician: Translating developmental science
into lifelong health. Pediatrics, 129(1), e224–e231.
doi:10.1542/peds.2011-2662.
Gianaros, P. J., & Manuck, S. B. (2010). Neurobiological
pathways linking socioeconomic position and
health. Psychosomatic Medicine, 72(5), 450–461.
doi:10.1097/PSY.0b013e3181e1a23c.
Gilbert, S. F. (2002). The genome in its ecological context: Philosophical perspectives on interspecies epigenesis. Annals of the New York Academy of Sciences,
981, 202–218.
Gilbert, S. F., & Epel, D. (2009). Ecological developmental biology: Integrating epigenetics, medicine, and
evolution. Sunderland: Sinauer Associates.
Gini, G., & Pozzoli, T. (2009). Association between
bullying and psychosomatic problems: A metaanalysis. Pediatrics, 123(3), 1059–1065. doi:10.1542/
peds.2008-1215.
Glew, G. M., Fan, M. Y., Katon, W., Rivara, F. P., & Kernic,
M. A. (2005). Bullying, psychosocial adjustment, and
academic performance in elementary school. Archives
of Pediatrics & Adolescent Medicine, 159(11), 1026–
1031. doi:10.1001/archpedi.159.11.1026.
Gluckman, P. D., Hanson, M. A., & Pinal, C. (2005). The
developmental origins of adult disease. Maternal &
Child Nutrition, 1(3), 130–141.
Gluckman, P. D., Hanson, M. A., & Beedle, A. S. (2007).
Non-genomic transgenerational inheritance of disease
risk. BioEssays, 29(2), 145–154.
Gluckman, P. D., Hanson, M. A., Bateson, P., Beedle,
A. S., Law, C. M., Bhutta, Z. A., et al. (2009). Towards
a new developmental synthesis: Adaptive developmental plasticity and human disease. Lancet, 373(9675),
1654–1657. doi:10.1016/S0140-6736(09)60234-8.
Goodman, E., Adler, N. E., Daniels, S. R., Morrison, J. A.,
Slap, G. B., & Dolan, L. M. (2003). Impact of objective and subjective social status on obesity in a biracial cohort of adolescents. Obesity Research, 11(8),
1018–1026.
Gottlieb, G. (1991). Experiential canalization of
behavioral development: Theory. Developmental
Psychology, 27(1), 4–13.
Greenfield, E. A. (2010). Child abuse as a life-course
social determinant of adult health. Maturitas, 66(1),
51–55. doi:10.1016/j.maturitas.2010.02.002.
Gump, B. B., Reihman, J., Stewart, P., Lonky, E.,
Darvill, T., & Matthews, K. A. (2007). Blood lead
(Pb) levels: A potential environmental mechanism explaining the relation between socioeconomic status and cardiovascular reactivity in
children. Health Psychology, 26(3), 296–304.
doi:10.1037/0278-6133.26.3.296.
Hackman, D. A., & Farah, M. J. (2009). Socioeconomic
status and the developing brain. Trends in
Cognitive Sciences, 13(2), 65–73. doi:10.1016/j.
tics.2008.11.003.
Hackman, D. A., Farah, M. J., & Meaney, M. J. (2010).
Socioeconomic status and the brain: Mechanistic
insights from human and animal research. Nature
Reviews Neuroscience, 11(9), 651–659. doi:10.1038/
nrn2897.
W. Thomas Boyce and C. Hertzman
Halpern, J. (2001). From detached concern to empathy:
Humanizing medical practice. New York: Oxford
University Press.
Hane, A. A., & Fox, N. A. (2006). Ordinary variations in
maternal caregiving influence human infants' stress
reactivity. Psychological Science, 17(6), 550–556.
doi:10.1111/j.1467-9280.2006.01742.x.
Hariri, A. R., & Weinberger, D. R. (2003). Imaging
genomics. British Medical Bulletin, 65, 259–270.
Hariri, A. R., Drabant, E. M., Munoz, K. E., Kolachana,
B. S., Mattay, V. S., Egan, M. F., & Weinberger, D. R.
(2005). A susceptibility gene for affective disorders
and the response of the human amygdala. Archives of
General Psychiatry, 62(2), 146–152.
Harlow, H. F., Harlow, M. K., & Suomi, S. J. (1971). From
thought to therapy: Lessons from a primate laboratory.
American Scientist, 59(5), 538–549.
Hart, T., & Risley, T. R. (1995). Meaningful differences in
the everyday experience of young American children.
Baltimore, MD: Paul H. Brookes.
Hawley, P. H. (1999). The ontogenesis of social dominance: A strategy-based evolutionary perspective.
Developmental Review, 19, 97–132.
Hazani, E., & Shasha, S. M. (2008). Effects of the
Holocaust on the physical health of the offspring of
survivors. The Israel Medical Association Journal,
10(4), 251–255.
Heijmans, B. T., Tobi, E. W., Stein, A. D., Putter, H., Blauw,
G. J., Susser, E. S., et al. (2008). Persistent epigenetic
differences associated with prenatal exposure to famine in humans. Proceedings of the National Academy
of Sciences of the United States of America, 105(44),
17046–17049. doi:10.1073/pnas.0806560105.
Hensch, T. K. (2005). Critical period plasticity in local
cortical circuits. Nature Reviews. Neuroscience,
6(11), 877–888. doi:10.1038/nrn1787.
Hertzman, C. (2010). Social geography of developmental
health in the early years. Healthcare Quarterly, 14(1),
32–40.
Hertzman, C., & Boyce, W. T. (2010). How experience
gets under the skin to create gradients in developmental health. Annual Review of Public Health, 31,
329–347. 323p following 347. doi:10.1146/annurev.
publhealth.012809.103538.
Hillis, S. D., Anda, R. F., Dube, S. R., Felitti, V. J.,
Marchbanks, P. A., & Marks, J. S. (2004). The association between adverse childhood experiences and
adolescent pregnancy, long-term psychosocial consequences, and fetal death. Pediatrics, 113(2), 320–327.
Houweling, T. A., & Kunst, A. E. (2010). Socio-economic
inequalities in childhood mortality in low- and
middle-income countries: A review of the international evidence. British Medical Bulletin, 93, 7–26.
doi:10.1093/bmb/ldp048.
Hrdy, S. B. (1999). Mother nature: Maternal instincts
and how they shape the human species. New York:
Ballantine Books.
Insel, T. R. (2010). The challenge of translation in social
neuroscience: A review of oxytocin, vasopressin,
and affiliative behavior. Neuron, 65(6), 768–779.
doi:10.1016/j.neuron.2010.03.005.
Early Childhood Health and the Life Course: The State of the Science and Proposed Research Priorities
Isles, A. R., & Wilkinson, L. S. (2008). Epigenetics: What
is it and why is it important to mental disease? British
Medical Bulletin, 85, 35–45. doi:10.1093/bmb/ldn004.
Janus, M., & Offord, D. (2000). Reporting on readiness to
learn at school in Canada. Canadian Journal of Policy
Research, 1, 71–75.
Javidi, H., & Yadollahie, M. (2012). Post-traumatic stress
disorder. Journal of Occupational and Environmental
Medicine, 3(1), 2–9.
Jirtle, R. L., & Skinner, M. K. (2007). Environmental epigenomics and disease susceptibility. Nature Reviews.
Genetics, 8(4), 253–262. doi:10.1038/nrg2045.
Kaplan, J. R., Manuck, S. B., Clarkson, T. B., Lusso,
F. M., & Taub, D. M. (1982). Social status, environment, and athersclerosis in cynomolgus monkeys.
Arteriosclerosis, 2, 359–368.
Karmiloff-Smith, A. (2007). Atypical epigenesis. Developmental Science, 10(1), 84–88.
doi:10.1111/j.1467-7687.2007.00568.x.
Kauffman, S. A. (1993). The origins of order: Selforganization and selection in evolution. New York:
Oxford University Press.
Kawachi, I., Adler, N. E., & Dow, W. H. (2010). Money,
schooling, and health: Mechanisms and causal evidence. Annals of the New York Academy of Sciences,
1186, 56–68. doi:10.1111/j.1749-6632.2009.05340.x.
Keating, D. P., & Hertzman, C. (1999). Developmental
health and the wealth of nations: Social, biological,
and educational dynamics. New York: The Guilford
Press.
Keltner, D., Gruenfeld, D., & Anderson, C. (2003). Power,
approach, and inhibition. Psychological Review,
110(2), 265–284.
Kershaw, P., Forer, B., Lloyd, J. E. V., Hertzman, C., Boyce,
W. T., Zumbo, B. D., et al. (2009). The use of population-level data to advance interdisciplinary methodology: A cell-through-society sampling framework for
child development research. International Journal of
Social Research Methodology, 12(5), 387–403.
Kessler, R. C., Duncan, G. J., Gennetian, L. A., Katz,
L. F., Kling, J. R., Sampson, N. A., et al. (2014).
Associations of housing mobility interventions for
children in high-poverty neighborhoods with subsequent mental disorders during adolescence. JAMA,
311(9), 937–948. doi:10.1001/jama.2014.607.
Keverne, E. B., Martensz, N. D., & Tuite, B. (1989).
Beta-endorphin concentrations in cerebrospinal fluid
of monkeys are influenced by grooming relationships.
Psychoneuroendocrinology, 14(1–2), 155–161.
Khan, Y., & Bhutta, Z. A. (2010). Nutritional deficiencies in the developing world: Current status and
opportunities for intervention. Pediatric Clinics of
North America, 57(6), 1409–1441. doi:10.1016/j.
pcl.2010.09.016.
Kim, Y. S., Koh, Y. J., & Leventhal, B. L. (2004).
Prevalence of school bullying in Korean middle
school students. Archives of Pediatrics & Adolescent
Medicine, 158(8), 737–741.
Kim, Y. S., Leventhal, B. L., Koh, Y. J., & Boyce,
W. T. (2009). Bullying increased suicide risk:
Prospective
study
of
Korean
adolescents.
89
Archives of Suicide Research, 13(1), 15–30.
doi:10.1080/13811110802572098.
King, M. L. (2007). Concepts of childhood: What we
know and where we might go. Renaissance Quarterly,
60(2), 371–407.
Kinnally, E. L., Capitanio, J. P., Leibel, R., Deng, L.,
LeDuc, C., Haghighi, F., & Mann, J. J. (2010a).
Epigenetic regulation of serotonin transporter
expression and behavior in infant rhesus macaques.
Genes, Brain, and Behavior, 9(6), 575–582.
doi:10.1111/j.1601-183X.2010.00588.x.
Kinnally, E. L., Karere, G. M., Lyons, L. A., Mendoza,
S. P., Mason, W. A., & Capitanio, J. P. (2010b).
Serotonin pathway gene-gene and gene-environment interactions influence behavioral stress
response in infant rhesus macaques. Development
and Psychopathology, 22(1), 35–44. doi:10.1017/
S0954579409990241.
Kinnally, E. L., Feinberg, C., Kim, D., Ferguson, K.,
Leibel, R., Coplan, J. D., & John Mann, J. (2011).
DNA methylation as a risk factor in the effects of
early life stress. Brain Behavior and Immunity.
doi:10.1016/j.bbi.2011.05.001.
Kittleson, M. M., Meoni, L. A., Wang, N. Y., Chu, A. Y.,
Ford, D. E., & Klag, M. J. (2006). Association of
childhood socioeconomic status with subsequent coronary heart disease in physicians. Archives of Internal
Medicine, 166(21), 2356–2361.
Knafo, A., Israel, S., & Ebstein, R. P. (2011). Heritability
of children's prosocial behavior and differential susceptibility to parenting by variation in the dopamine
receptor D4 gene. Development and Psychopathology,
23(1), 53–67. doi:10.1017/S0954579410000647.
Koopman, J. S., & Lynch, J. W. (1999). Individual causal
models and population system models in epidemiology. American Journal of Public Health, 89(8),
1170–1174.
Kraemer, H. C., Stice, E., Kazdin, A., Offord, D., &
Kupfer, D. (2001). How do risk factors work together?
Mediators, moderators, independent, overlapping
and proxy-risk factors. The American Journal of
Psychiatry, 158, 848–856.
Kraemer, H. C., Kiernan, M., Essex, M., & Kupfer,
D. J. (2008). How and why criteria defining moderators and mediators differ between
the Baron & Kenny and MacArthur approaches.
Health Psychology, 27(2 Suppl), S101–S108.
doi:10.1037/0278-6133.27.2(Suppl.).S101.
Krieger, N. (2001). Theories for social epidemiology in the
21st century: An ecosocial perspective. International
Journal of Epidemiology, 30, 668–677.
Kuh, D., & Ben-Shlomo, Y. (2004). A life course approach
to chronic disease epidemiology (2nd ed.). Oxford:
Oxford University Press.
Kuzawa, C. W., & Thayer, Z. M. (2011). Timescales of
human adaptation: The role of epigenetic processes.
Epigenomics, 3(2), 221–234. doi:10.2217/epi.11.11.
Ladd-Acosta, C., Pevsner, J., Sabunciyan, S., Yolken,
R. H., Webster, M. J., Dinkins, T., et al. (2007).
DNA methylation signatures within the human brain.
90
American Journal of Human Genetics, 81(6), 1304–
1315. doi:10.1086/524110.
Lam, L. L., Emberly, E., Fraser, H. B., Neumann, S. M.,
Chen, E., Miller, G. E., & Kobor, M. S. (2012).
Biological and environmental predictors of variable
DNA methylation in a human community cohort. PNAS.
Lawlor, D. A., Sterne, J. A., Tynelius, P., Davey Smith,
G., & Rasmussen, F. (2006). Association of childhood
socioeconomic position with cause-specific mortality in a prospective record linkage study of 1,839,384
individuals. American Journal of Epidemiology,
164(9), 907–915. doi:10.1093/aje/kwj319.
Lesch, K. P. (2007). Linking emotion to the social brain.
The role of the serotonin transporter in human social
behaviour. EMBO Reports, 8, S24–S29. doi:10.1038/
sj.embor.7401008.
Liew, C. C., Ma, J., Tang, H. C., Zheng, R., & Dempsey,
A. A. (2006). The peripheral blood transcriptome
dynamically reflects system wide biology: A potential diagnostic tool. The Journal of Laboratory and
Clinical Medicine, 147(3), 126–132. doi:10.1016/j.
lab.2005.10.005.
Liu, L., Li, Y., & Tollefsbol, T. O. (2008). Geneenvironment interactions and epigenetic basis of
human diseases. Current Issues in Molecular Biology,
10(1–2), 25–36.
Liu, L., Johnson, H. L., Cousens, S., Perin, J., Scott,
S., Lawn, J. E., et al. (2012). Global, regional, and
national causes of child mortality: An updated systematic analysis for 2010 with time trends since
2000. Lancet, 379(9832), 2151–2161. doi:10.1016/
S0140-6736(12)60560-1.
Lloyd, L. J., Langley-Evans, S. C., & McMullen, S. (2012).
Childhood obesity and risk of the adult metabolic syndrome: A systematic review. International Journal of
Obesity, 36(1), 1–11. doi:10.1038/ijo.2011.186.
Lumey, L. H., Stein, A. D., & Susser, E. (2011).
Prenatal famine and adult health. Annual Review
of Public Health, 32, 237–262. doi:10.1146/
annurev-publhealth-031210-101230.
Lupien, S. J., King, S., Meaney, M. J., & McEwen, B. S.
(2001). Can poverty get under your skin? Basal cortisol levels and cognitive function in children from
low and high socioeconomic status. Development and
Psychopathology, 13, 653–676.
MacDonald, J. L., & Roskams, A. J. (2009). Epigenetic
regulation of nervous system development by DNA
methylation and histone deacetylation. Progress in
Neurobiology, 88(3), 170–183.
Manuck, S. B., Craig, A. E., Flory, J. D., Halder, I., &
Ferrell, R. E. (2011). Reported early family environment covaries with menarcheal age as a function of
polymorphic variation in estrogen receptor-alpha.
Development and Psychopathology, 23(1), 69–83.
doi:10.1017/S0954579410000659.
Marmot, M. (2010). Fair society, healthy lives. Firenze:
L.S. Olschki.
Mazumdar, S., King, M., Liu, K. Y., Zerubavel, N., &
Bearman, P. (2010). The spatial structure of autism in
California, 1993-2001. Health & Place, 16(3), 539–
546. doi:10.1016/j.healthplace.2009.12.014.
W. Thomas Boyce and C. Hertzman
McDade, T. (2012). Early environments and the ecologics of inflammation. Proceedings of the National
Academy of Sciences of the United States of America,
109(Suppl 2), 17281–17288.
McEwen, B. S., & Gianaros, P. J. (2010). Central role
of the brain in stress and adaptation: Links to socioeconomic status, health, and disease. Annals of the
New York Academy of Sciences, 1186, 190–222.
doi:10.1111/j.1749-6632.2009.05331.x.
McGowan, P. O., & Szyf, M. (2010). The epigenetics of
social adversity in early life: Implications for mental
health outcomes. Neurobiology of Disease, 39(1),
66–72. doi:10.1016/j.nbd.2009.12.026.
McGowan, P. O., Sasaki, A., Huang, T. C., Unterberger,
A., Suderman, M., Ernst, C., et al. (2008). Promoterwide hypermethylation of the ribosomal RNA gene
promoter in the suicide brain. PloS One, 3(5), e2085.
McGowan, P. O., Sasaki, A., D'Alessio, A. C., Dymov, S.,
Labonte, B., Szyf, M., et al. (2009). Epigenetic regulation of the glucocorticoid receptor in human brain
associates with childhood abuse. Nature Neuroscience,
12(3), 342–348. doi:10.1038/nn.2270.
Meaney, M. J. (2010). Epigenetics and the biological definition of gene x environment interactions. Child Development, 81(1), 41–79.
doi:10.1111/j.1467-8624.2009.01381.x.
Meaney, M. J., & Ferguson-Smith, A. C. (2010).
Epigenetic regulation of the neural transcriptome: The
meaning of the marks. Nature Neuroscience, 13(11),
1313–1318. doi:10.1038/nn1110-1313.
Mehler, M. F. (2008). Epigenetic principles and mechanisms underlying nervous system functions in health
and disease. Progress in Neurobiology, 86(4),
305–341.
Miller, G. E., Chen, E., Fok, A. K., Walker, H., Lim,
A., Nicholls, E. F., et al. (2009a). Low early-life
social class leaves a biological residue manifested by
decreased glucocorticoid and increased proinflammatory signaling. Proceedings of the National Academy
of Sciences of the United States of America, 106(34),
14716–14721. doi:10.1073/pnas.0902971106.
Miller, A. H., Maletic, V., & Raison, C. L. (2009b).
Inflammation and its discontents: The role of cytokines in the pathophysiology of major depression.
Biological Psychiatry, 65(9), 732–741. doi:10.1016/j.
biopsych.2008.11.029.
Moffitt, T. E., Caspi, A., & Rutter, M. (2005). Strategy
for investigating interactions between measured genes
and measured environments. Archives of General
Psychiatry, 62(5), 473–481.
Mohr, S., & Liew, C. C. (2007). The peripheral-blood
transcriptome: New insights into disease and risk
assessment. Trends in Molecular Medicine, 13(10),
422–432. doi:10.1016/j.molmed.2007.08.003.
Morita, K., Saito, T., Ohta, M., Ohmori, T., Kawai, K.,
Teshima-Kondo, S., & Rokutan, K. (2005). Expression
analysis of psychological stress-associated genes in
peripheral blood leukocytes. Neuroscience Letters,
381(1–2), 57–62.
Msall, M. E., Bier, J. A., LaGasse, L., Tremont, M., &
Lester, B. (1998). The vulnerable preschool child: The
Early Childhood Health and the Life Course: The State of the Science and Proposed Research Priorities
impact of biomedical and social risks on neurodevelopmental function. Seminars in Pediatric Neurology,
5(1), 52–61.
Nesse, R. M., & Young, E. A. (2000). Evolutionary origins
and functions of the stress response. In G. Fink (Ed.),
Encyclopedia of stress (Vol. 2, pp. 79–84). San Diego:
Academic Press.
Norman, G. J., Hawkley, L. C., Cole, S. W., Berntson,
G. G., & Cacioppo, J. T. (2012). Social neuroscience: The social brain, oxytocin, and health. Social
Neuroscience, 7(1), 18–29. doi:10.1080/17470919.2
011.568702.
Oberg, C. (2008). Children of genocide: A legacy of lost
dreams. Pediatrics, 121(3), 611–615. doi:10.1542/
peds.2007-2208.
Oberlander, T. F., Weinberg, J., Papsdorf, M., Grunau, R.,
Misri, S., & Devlin, A. M. (2008). Prenatal exposure to
maternal depression, neonatal methylation of human
glucocorticoid receptor gene (NR3C1) and infant cortisol stress response. Epigenetics, 3(2), 97–106.
Obradovic, J., Bush, N. R., Stamperdahl, J., Adler, N. E.,
& Boyce, W. T. (2010). Biological sensitivity to context: The interactive effects of stress reactivity and
family adversity on socio-emotional behavior and
school readiness. Child Development, 81(1), 270–289.
Offord, D. R. (1995). Child psychiatric epidemiology:
Current status and future prospects. Canadian Journal
of Psychiatry. Revue Canadienne de Psychiatrie,
40(6), 284–288.
Olds, D. L., Kitzman, H. J., Cole, R. E., Hanks, C. A.,
Arcoleo, K. J., Anson, E. A., et al. (2010). Enduring
effects of prenatal and infancy home visiting by
nurses on maternal life course and government
spending: Follow-up of a randomized trial among
children at age 12 years. Archives of Pediatrics and
Adolescent Medicine, 164(5), 419–424. doi:10.1001/
archpediatrics.2010.49.
Ollikainen, M., Smith, K. R., Joo, E. J., Ng, H. K.,
Andronikos, R., Novakovic, B., et al. (2010). DNA
methylation analysis of multiple tissues from newborn
twins reveals both genetic and intrauterine components to variation in the human neonatal epigenome.
Human Molecular Genetics, 19(21), 4176–4188.
doi:10.1093/hmg/ddq336.
Ostrove, J. M., Adler, N. E., Kuppermann, M., &
Washington, A. E. (2000). Objective and subjective
assessments of socioeconomic status and their relationship to self-rated health in an ethnically diverse
sample of pregnant women. Health Psychology, 19(6),
613–618.
Overton, W. F., & Lerner, R. M. (2014). Fundamental concepts and methods in developmental science: A relational perspective. Research in Human Development,
11, 63–73.
Painter, R. C., Roseboom, T. J., & Bleker, O. P. (2005).
Prenatal exposure to the Dutch famine and disease
in later life: An overview. Reproductive Toxicology,
20(3), 345–352.
Painter, R., Osmond, C., Gluckman, P., Hanson, M.,
Phillips, D., & Roseboom, T. (2008). Transgenerational
effects of prenatal exposure to the Dutch famine on
91
neonatal adiposity and health in later life. BJOG: An
International Journal of Obstetrics & Gynaecology,
115(10), 1243–1249.
Panter-Brick, C., Lende, D., & Kohrt, B. (2012).
Children in global adversity: Physical health, mental health, behavioral health, and symbolic health. In
V. Maholmes & R. B. King (Eds.), The Oxford handbook of poverty and child development (pp. 603–621).
Oxford: Oxford University Press.
Pellegrini, A. D., Roseth, C. J., Mliner, S., Bohn,
C. M., Van Ryzin, M., Vance, N., et al. (2007).
Social dominance in preschool classrooms. Journal
of Comparative Psychology, 121(1), 54–64.
doi:10.1037/0735-7036.121.1.54.
Pellegrini, A. D., Van Ryzin, M. J., Roseth, C., BohnGettler, C., Dupuis, D., Hickey, M., & Peshkam, A.
(2010). Behavioral and social cognitive processes in
preschool children's social dominance. Aggressive
Behavior, 37(3), 248–257. doi:10.1002/ab.20385.
Pezawas, L., Meyer-Lindenberg, A., Drabant, E. M.,
Verchinski, B. A., Munoz, K. E., Kolachana, B. S.,
et al. (2005). 5-HTTLPR polymorphism impacts
human cingulate-amygdala interactions: A genetic
susceptibility mechanism for depression. Nature
Neuroscience, 8(6), 828–834.
Pluess, M., & Belsky, J. (2013). Vantage sensitivity:
Individual differences in response to positive experiences. Psychological Bulletin, 139(4), 901–916.
doi:10.1037/a0030196.
Quas, J. A., Bauer, A., & Boyce, W. T. (2004). Physiological
reactivity, social support, and memory in early childhood. Child Development, 75(3), 797–814.
Rifkin, J. (2009). The empathic civilization: The race to
global consciousness in a world in crisis. New York:
Jeremy P. Tarcher/Penguin.
Robert Wood Johnson Foundation Commission to Build a
Healthier America. (2009). Beyond Health Care: New
Directions to a Healthier America. Retrieved from
Princeton, NJ:
Robinson, G. E., Grozinger, C. M., & Whitfield, C. W.
(2005). Sociogenomics: Social life in molecular terms.
Nature Reviews Genetics, 6, 257–270.
Robinson, G. E., Fernald, R. D., & Clayton, D. F. (2008).
Genes and social behavior. Science, 322(5903), 896–
900. doi:10.1126/science.1159277.
Robison, A. J., & Nestler, E. J. (2011). Transcriptional and
epigenetic mechanisms of addiction. Nature Reviews.
Neuroscience, 12(11), 623–637. doi:10.1038/nrn3111.
Roseboom, T. J., van der Meulen, J. H., Osmond, C.,
Barker, D. J., Ravelli, A. C., Schroeder-Tanka, J. M.,
et al. (2000). Coronary heart disease after prenatal
exposure to the Dutch famine, 1944-45. Heart, 84(6),
595–598.
Roth, T. L., Lubin, F. D., Funk, A. J., & Sweatt, J. D.
(2009). Lasting epigenetic influence of early-life
adversity on the BDNF gene. Biological Psychiatry,
65(9), 760–769. doi:10.1016/j.biopsych.2008.11.028.
Russo, V. E. A., Cove, D. J., Edgar, L. G., Jaenisch, R.,
& Salamini, F. (Eds.). (1999). Development: Genetics,
epigenetics and environmental regulation. Berlin:
Springer-Verlag.
92
Rutter, M. (2006). Genes and behaviour: Nature/nurture interplay explained. Oxford, UK: Blackwell
Publishing.
Rutter, M. (2012). Achievements and challenges in the
biology of environmental effects. PNAS.
Sapolsky, R. M. (2003). Stress and plasticity in the limbic
system. Neurochemical Research, 28(11), 1735–1742.
Sapolsky, R. M. (2005). The influence of social hierarchy
on primate health. Science, 308(5722), 648–652.
Schneider, J., Dickinson, M., & Levine, J. (2012). Social
structures depend on innate determinants and chemosensory processing in Drosophila. PNAS.
Sentenac, M., Arnaud, C., Gavin, A., Molcho, M.,
Gabhainn, S. N., & Godeau, E. (2012). Peer victimization among school-aged children with chronic
conditions. Epidemiologic Reviews, 34(1), 120–128.
doi:10.1093/epirev/mxr024.
Shannon, K. E., Beauchaine, T. P., Brenner, S. L.,
Neuhaus, E., & Gatzke-Kopp, L. (2007). Familial and
temperamental predictors of resilience in children at
risk for conduct disorder and depression. Development
and Psychopathology, 19(3), 701–727. doi:10.1017/
S0954579407000351.
Shonkoff, J. (2012). Leveraging the biology of adversity and resilience to address the roots of disparities in health and development. Proceedings of the
National Academy of Sciences of the United States of
America,109(Suppl 2), 17302–17307.
Shonkoff, J. P., & Garner, A. S. (2012). The lifelong effects of early childhood adversity and toxic
stress. Pediatrics, 129(1), e232–e246. doi:10.1542/
peds.2011-2663.
Shonkoff, J. P., & Phillips, D. A. (Eds.). (2000). From
neurons to neighborhoods: The science of early child
development. Washington, DC: National Academy
Press.
Shorter, E. (1975). The making of the modern family.
New York: Basic Books.
Slavich, G. M., & Cole, S. W. (2013). The emerging field
of human social genomics. Clinical Psychological
Science: A Journal of the Association for Psychological
Science, 1(3), 331–348.
Sourander, A., Jensen, P., Ronning, J. A., Elonheimo, H.,
Niemela, S., Helenius, H., et al. (2007). Childhood
bullies and victims and their risk of criminality in
late adolescence: The Finnish From a Boy to a Man
study. Archives of Pediatrics & Adolescent Medicine,
161(6), 546–552. doi:10.1001/archpedi.161.6.546.
Stearns, P. N. (2011). Childhood in world history (2nd
ed.). New York, NY: Routledge.
Steptoe, A., Feldman, P. J., Kunz, S., Owen, N., Willemsen,
G., & Marmot, M. (2002). Stress responsivity and
socioeconomic status: A mechanism for increased cardiovascular disease risk? European Heart Journal, 23,
1757–1763.
Stevens, H. E., Leckman, J. F., Coplan, J. D., & Suomi,
S. J. (2009). Risk and resilience: Early manipulation of macaque social experience and persistent behavioral and neurophysiological outcomes.
W. Thomas Boyce and C. Hertzman
Journal of the American Academy of Child and
Adolescent Psychiatry, 48(2), 114–127. doi:10.1097/
CHI.0b013e318193064c.
Suomi, S. J. (2006). Risk, resilience, and gene x environment interactions in rhesus monkeys. Annals of
the New York Academy of Sciences, 1094, 52–62.
doi:10.1196/annals.1376.006.
Syme, S. L. (2008). Reducing racial and social-class
inequalities in health: The need for a new approach.
Health Affairs (Millwood), 27(2), 456–459.
doi:10.1377/hlthaff.27.2.456.
Szyf, M., McGowan, P., & Meaney, M. J. (2008). The social
environment and the epigenome. Environmental and
Molecular Mutagenesis, 49(1), 46–60. doi:10.1002/
em.20357.
Thompson, P. M., Martin, N. G., & Wright, M. J.
(2010). Imaging genomics. Current Opinion
in Neurology, 23(4), 368–373. doi:10.1097/
WCO.0b013e32833b764c.
Tsankova, N., Renthal, W., Kumar, A., & Nestler, E. J.
(2007). Epigenetic regulation in psychiatric disorders. Nature Reviews. Neuroscience, 8(5), 355–367.
doi:10.1038/nrn2132.
Turecki, G., Ernst, C., Jollant, F., Labonte, B., &
Mechawar, N. (2012). The neurodevelopmental origins of suicidal behavior. Trends in Neurosciences,
35(1), 14–23. doi:10.1016/j.tins.2011.11.008.
Turner, J. D., Pelascini, L. P., Macedo, J. A., & Muller,
C. P. (2008). Highly individual methylation patterns
of alternative glucocorticoid receptor promoters suggest individualized epigenetic regulatory mechanisms. Nucleic Acids Research, 36(22), 7207–7218.
doi:10.1093/nar/gkn897.
Turner, H. A., Finkelhor, D., Shattuck, A., & Hamby,
S. (2012). Recent victimization exposure and suicidal ideation in adolescents. Archives of Pediatrics
& Adolescent Medicine, 1–6. doi:10.1001/
archpediatrics.2012.1549.
U.S. Department of Health and Human Services. (2011).
CDC Health Disparities and Inequalities Report—
United States, 2011. Atlanta: U.S. Department of
Health and Human Services, Centers for Disease
Control and Prevention.
Uebelhack, R., Franke, L., Herold, N., Plotkin, M.,
Amthauer, H., & Felix, R. (2006). Brain and platelet
serotonin transporter in humans-correlation between
[123I]-ADAM SPECT and serotonergic measurements in platelets. Neuroscience Letters, 406(3), 153–
158. doi:10.1016/j.neulet.2006.06.004.
van IJzendoorn, M. H., Caspers, K., BakermansKranenburg, M. J., Beach, S. R., & Philibert, R. (2010).
Methylation matters: Interaction between methylation
density and serotonin transporter genotype predicts
unresolved loss or trauma. Biological Psychiatry,
68(5), 405–407. doi:10.1016/j.biopsych.2010.05.008.
Vogel, E. F., & Bell, N. W. (1961). The emotionally disturbed child as the family scapegoat. In N. W. Bell &
E. F. Vogel (Eds.), A Modern introduction to the family. London: Routledge & Kegan Paul.
Early Childhood Health and the Life Course: The State of the Science and Proposed Research Priorities
Waddington, C. H. (1959). Canalization of development and genetic assimilation of acquired characters.
Nature, 183(4676), 1654–1655.
Waddington, C. H. (2012). The epigenotype. 1942.
International Journal of Epidemiology, 41(1), 10–13.
doi:10.1093/ije/dyr184.
Wadsworth, M. E., Raviv, T., Reinhard, C., Wolff, B.,
Santiago, C. D. C., & Einhorn, L. (2008). An indirect
effects model of the association between poverty and
child functioning: The role of children. Journal of
Loss and Trauma, 30.
Weaver, I. C. (2009). Epigenetic effects of glucocorticoids. Seminars in Fetal and Neonatal Medicine,
14(3), 143–150. doi:10.1016/j.siny.2008.12.002.
Weaver, I. C., Cervoni, N., Champagne, F. A., D’Alessio,
A. C., Sharma, S., Seckl, J. R., et al. (2004).
Epigenetic programming by maternal behavior.
Nature Neuroscience, 7(8), 847–854.
Weikum, W. M., Oberlander, T. F., Hensch, T. K., &
Werker, J. F. (2012). Prenatal exposure to antidepressants and depressed maternal mood alter trajectory of infant speech perception. Proceedings of the
National Academy of Sciences, 109(2), 17221–17227.
doi:10.1073/pnas.1121263109.
West-Eberhard, M. J. (2003). Developmental plasticity and evolution. New York: Oxford University
Press.
93
Whittle, S., Yap, M. B., Sheeber, L., Dudgeon, P., Yucel,
M., Pantelis, C., et al. (2010). Hippocampal volume
and sensitivity to maternal aggressive behavior: A
prospective study of adolescent depressive symptoms.
Development and Psychopathology, 23(1), 115–129.
doi:10.1017/S0954579410000684.
Wolf, D. M., Vazirani, V. V., & Arkin, A. P. (2005).
Diversity in times of adversity: Probabilistic strategies in microbial survival games. Journal of
Theoretical Biology, 234(2), 227–253. doi:10.1016/j.
jtbi.2004.11.020.
Wright, R. O., & Christiani, D. (2010). Gene-environment
interaction and children's health and development.
Current Opinion in Pediatrics, 22(2), 197–201.
doi:10.1097/MOP.0b013e328336ebf9.
Wu, H., & Sun, Y. E. (2006). Epigenetic regulation of stem
cell differentiation. Pediatric Research, 59(4 Pt 2),
21R–25R. doi:10.1203/01.pdr.0000203565.76028.2a.
Yang, E.-J., Lin, E. W., & Hensch, T. K. (2012). A critical
period for acoustic preference in mice. Proceedings
of the National Academy of Sciences, 109(Suppl 2),
17213–17220.
Zhang, T. Y., & Meaney, M. J. (2010). Epigenetics
and the environmental regulation of the genome
and its function. Annual Review of Psychology,
61, 439–466, C431–433.. doi:10.1146/annurev.
psych.60.110707.163625.
Open Access This chapter is licensed under the terms of the Creative Commons Attribution 4.0 International License
(http://creativecommons.org/licenses/by/4.0/), which permits use, sharing, adaptation, distribution and reproduction in
any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license and indicate if changes were made.
The images or other third party material in this chapter are included in the chapter’s Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the chapter’s Creative Commons license
and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain
permission directly from the copyright holder.
Middle Childhood:
An Evolutionary-Developmental
Synthesis
Marco DelGiudice
Middle childhood—conventionally going from
about 6–11 years of age—is a crucial yet underappreciated phase of human development. On the
surface, middle childhood may appear like a
slow-motion interlude between the spectacular
transformations of infancy and early childhood
and those of adolescence. In reality, this life stage
is anything but static: the transition from early to
middle childhood heralds a global shift in cognition, motivation, and social behavior, with profound and wide-ranging implications for the
development of personality, sex differences, and
even psychopathology (Table 1).
In the last two decades, converging theories and
findings from anthropology, primatology, evolutionary psychology, endocrinology, and behavior
genetics have revolutionized our understanding of
middle childhood. In this chapter, I show how
these diverse contributions can be synthesized into
an integrated evolutionary-developmental model
of middle childhood. I begin by reviewing the
main evolved functions of middle childhood and
the cognitive, behavioral, and hormonal processes
that characterize this life stage. Then, I introduce
the idea that the transition to middle childhood
works as a switch point in the development of life
history strategies (Del Giudice et al. 2009, 2012;
Del Giudice and Belsky 2011) and discuss three
insights in the nature of middle childhood that
arise from an integrated approach. This chapter
was originally published as a short paper in the
journal Child Development Perspectives (Del
Giudice 2014a). It is reprinted here with updated
references and a new section on the model’s implications for health development in a life course perspective (LCHD).
1
This chapter contains a modified version of a previously
published review and analysis of existing research, written by Marco Del Giudice, that appeared in Child
Development Perspectives.
Reprinted with permission from:
Del Giudice, M. (2014), Middle Childhood: An
Evolutionary-Developmental Synthesis. Child Dev
Perspect, 8: 193–200. doi:10.1111/cdep.12084.
M. DelGiudice (*)
Department of Psychology, University of New
Mexico, Logan Hall, 2001 Redondo Dr. NE,
Albuquerque, NM 87131, USA
e-mail: marcodg@unm.edu
What Is Middle Childhood?
Middle childhood is one of the main stages of
human development, marked by the eruption of the
first permanent molars around age 6 and the onset
of androgen secretion by the adrenal glands at
about 6–8 years (Bogin 1997). In middle childhood, body growth slows considerably, usually following a small mid-growth spurt. At the same time,
muscularity increases and the body starts accumulating fat (the adiposity rebound; Hochberg 2008),
while sex differences in body composition become
more pronounced (Del Giudice et al. 2009; Wells
© The Author(s) 2018
N. Halfon et al. (eds.), Handbook of Life Course Health Development,
DOI 10.1007/978-3-319-47143-3_5
95
M. DelGiudice
96
Table 1 Development in middle childhood
Body growth
Brain growth
Motor and perceptual
skills
Cognitive skills
Motivation and social
behavior
Psychopathology
Social context
Behavior genetics
Eruption of permanent molars
Mid-growth spurt, followed by decelerating skeletal growth
Increased muscle mass
Increased adiposity and BMI (adiposity rebound)
Initial development of axillary hair and body odor
Increased sex differences in adiposity (F > M), bone strength, and muscularity (M > F)
Emergence of sex differences in vocal characteristics
Approaching peak of overall brain volume
Peak of gray matter volume
Continuing increase in white matter volume/integrity
Increased gross motor skills (e.g., walking)
Increased fine motor skills
Local-global shift in visual processing preferences
Increased reasoning and problem-solving skills (e.g., concrete operations)
Increased self-regulation and executive functions (inhibition, attention, planning, etc.)
Increased mentalizing skills (multiple perspectives, conflicting goals)
Increased navigational skills (working memory, ability to understand maps)
Acquisition of cultural norms (e.g., prosociality)
Complex moral reasoning (conflicting points of view)
Increased pragmatic abilities (gossiping, storytelling, verbal competition, etc.)
Consolidation of status/dominance hierarchies
Changes in aggression levels (individual trajectories)
Development of disgust
Changes in food preferences (e.g., spicy foods)
Onset of sexual/romantic attraction
Increased frequency of sexual play
Increased sense of gender identity
Peak of sex segregation
Peak of sex differences in social play (including play fighting vs. play parenting)
Increased sex differences in physical aggression (M > F)
Emergence of sex differences in attachment styles
Early peak of psychopathology onset (externalizing, anxiety, phobias, ADHD)
Peak onset of fetishistic attractions
Emergence of sex differences in conduct disorders (M > F)
Active involvement in caretaking, foraging, domestic tasks, helping
Expectations of responsible behavior
Attribution of individuality and personhood (“getting noticed”)
Increased heritability of general intelligence and language skills
New genetic influences on general intelligence, language, aggression, and prosociality
See the main text for supporting references
BMI body mass index, M male, F female, ADHD attention deficit hyperactivity disorder
2007). Figure 1 places middle childhood in the
broader context of human growth from conception
to adolescence.
In biological terms, middle childhood corresponds to human juvenility—a stage in which the
individual is still sexually immature, but no lon-
ger dependent on parents for survival. In social
mammals and primates, juvenility is a phase of
intense learning—often accomplished through
play—in which youngsters practice adult behavioral patterns and acquire essential social and foraging skills. Indeed, the duration of juvenility in
Middle Childhood: An Evolutionary-Developmental Synthesis
97
Fig. 1 Developmental trajectories of human growth and sex hormones production, from conception to adolescence.
Arrows show the landmark events that characterize middle childhood (Reproduced from Del Giudice 2014a)
primates correlates strongly with the size and
complexity of social groups, as well as with cortical brain volume (Joffe 1997). Social learning
in juvenility can be understood as investment in
embodied capital—skills and knowledge that
cost time and effort to acquire but increase an
individual’s performance and reproductive success (Kaplan et al. 2000).
Human children are no exception to this pattern. Social learning is universally recognized as
a key evolved function of middle childhood and
is enabled by a global reorganization of cognitive
functioning known as the five-to-seven shift
(Weisner 1996). By age 6, the brain has almost
reached its maximum size and receives a decreasing share of the body’s glucose after the consumption peak of early childhood (Fig. 1; Giedd
and Rapoport 2010; Kuzawa et al. 2014).
However, brain development proceeds at a sustained pace, with intensive synaptogenesis in cortical areas (gray matter) and rapid maturation of
axonal connections (white matter; Lebel et al.
2008). The transition to middle childhood is
marked by a simultaneous increase in perceptual
abilities (including a transition from local to
global visual processing), motor control (including the emergence of adult-like walking), and
complex reasoning skills (Bjorklund 2011; Poirel
et al. 2011; Weisner 1996). The most dramatic
changes probably occur in the domain of selfregulation and executive functions: children
become much more capable of inhibiting
unwanted behavior, maintaining sustained attention, making and following plans, and so forth
(Best et al. 2009; Weisner 1996; see also
McClelland et al. 2017). Parallel improvements
M. DelGiudice
98
take place in mentalizing (the ability to understand and represent mental states) and moral reasoning, as children become able to consider
multiple perspectives and conflicting goals
(Jambon and Smetana 2014; Lagattuta et al.
2009).
In traditional societies, older relatives—especially parents and grandparents—are the main
sources of knowledge for juveniles, supplemented
by peers and—where available—professional
teachers. Storytelling—both fictional and based in
real events—is a powerful technology for transmitting knowledge about foraging and social
skills, avoidance of dangers, topography, wayfinding, and social roles and norms (Scalise Sugiyama
2011). Storytelling mimics the format of episodic
memories, providing the child with a rich source
of indirect experience (Scalise Sugiyama 2011).
Intriguingly, episodic memory shows dramatic
and sustained improvements across middle childhood (Ghetti and Bunge 2012).
However, children at this age are not just
learning and playing. Cross-culturally, middle
childhood is the time when children are expected
to start helping with domestic tasks—such as caring for younger siblings, collecting food and
water, tending animals, and helping adults prepare food (Bogin 1997; Lancy and Grove 2011;
Scalise Sugiyama 2011; Weisner 1996). In favorable ecologies, juveniles can contribute substantially to family subsistence (Kramer 2011).
Thanks to marked increases in spatial cognition—reflected in the emerging ability to understand maps—and navigational skills, children
become able to memorize complex routes and
find their way without adult supervision
(Bjorklund 2011; Piccardi et al. 2014). The
important role of juveniles in collecting and preparing food may explain why the emotion of disgust does not fully develop until middle childhood
(Rozin 1990a).
The transition to middle childhood is typically
associated with a strong separation in gender
roles, even in societies where tasks are not rigidly
assigned by sex. Spontaneous sex segregation of
boys and girls peaks during these years, as does
the frequency of sexually differentiated play (Del
Giudice et al. 2009). On a broader social level,
cross-cultural evidence shows that juveniles start
“getting noticed” by adults—that is, they begin to
be viewed fully as people with their own individuality, personality, and social responsibility
(Lancy and Grove 2011).
In summary, the life stage of juvenility/middle
childhood has two major interlocking functions:
social learning and social integration in a system
of roles, norms, activities, and shared knowledge.
While children are still receiving sustained
investment from parents and other relatives—in
the form of food, protection, knowledge, and so
forth—they also start to actively contribute to
their family economy. By providing resources
and sharing the burden of child care, juveniles
can boost their parents’ reproductive potential.
The dual nature of juveniles as both receivers and
providers explains many psychological features
of middle childhood and has likely played a
major role in the evolution of human life history
(Kramer 2011).
1.1
Adrenarche
The transition to middle childhood is coordinated
by a remarkable endocrinological event: the
awakening of the adrenal glands, or adrenarche
(Auchus and Rainey 2004; Hochberg 2008).
Starting at about 6–8 years—with much individual variation and only minor differences between
males and females—adrenal glands begin to
secrete increasing amounts of androgens (see
Fig. 1), mainly dehydroepiandrosterone (DHEA)
and its sulfate (DHEAS). Adrenal androgens
have only minor effects on physical development,
but they have powerful effects on brain functioning. DHEA and DHEAS promote neurogenesis
and modulate gamma-aminobutyric acid (GABA)
and glutamate receptors; moreover, DHEA can
act directly on androgen and estrogen receptors.
Even more important, adrenal androgens can be
converted to estrogen or testosterone in the brain
(Campbell 2006; Del Giudice et al. 2009). As sex
hormones, adrenal androgens play a twofold role:
They activate sexually differentiated brain pathways that had been previously organized by the
hormonal surges of prenatal development and
Middle Childhood: An Evolutionary-Developmental Synthesis
infancy (Fig. 1), and they further organize brain
development along sexually differentiated trajectories (Del Giudice et al. 2009).
Adrenal androgens likely provide a major
impulse for many of the psychological changes
of middle childhood (Campbell 2006, 2011; Del
Giudice et al. 2009), including the emergence
and intensification of sex differences across
domains (see Table 1). Since the age of adrenarche
correlates strongly with that of gonadarche (the
awakening of the testes/ovaries that marks the
beginning of puberty; Hochberg 2008), human
development shows a peculiar pattern in which
sexually differentiated brain pathways are activated several years before the development of
secondary sexual characteristics. This developmental pattern (shared by chimpanzees and, to a
lesser extent, gorillas; Bernstein et al. 2012)
results in a temporary decoupling between physical and behavioral development, consistent with
the idea of middle childhood as a sexually differentiated phase of social learning and experimentation (Geary 2010). Moreover, adrenal
androgens promote extended brain plasticity
through synaptogenesis and may play an important role in shifting the allocation of the body’s
energetic resources away from brain development and toward the accumulation of muscle and
fat in preparation for puberty (Campbell 2006,
2011; see also Kuzawa et al. 2014).
2
The Transition to Middle
Childhood
as a Developmental Switch
Point
The evolutionary model of middle childhood
sketched in the previous section can be enriched
and extended by considering the role of adrenarche
as a developmental switch (Del Giudice et al.
2009). The concept of a developmental switch was
introduced by West-Eberhard (2003); a switch is a
regulatory mechanism that activates at a specific
point in development, collects input from the
external environment or the state of the organism,
and shifts the individual along alternative pathways—ultimately resulting in the development of
99
alternative phenotypes (morphological, physiological, or behavioral traits of an organism). For
example, a switch may regulate the development
of aggressive behavior so that safe conditions
entrain the development of low levels of aggression, whereas threatening environments trigger
high levels of aggression. Developmental switches
enable adaptive plasticity—the ability of an organism to adjust its phenotype to match the local environment in a way that promotes biological fitness
(West-Eberhard 2003). In other words, plastic
organisms track the state of the environment—
usually through indirect cues—and use this information to develop alternative phenotypes that tend
to promote survival or reproduction under different conditions.
Developmental switches work in a modular
fashion (see Fig. 2). Activation of a switch leads
to the coordinated expression of different genes—
both those involved in the regulatory mechanism
itself and those involved in the production of the
new phenotype. Moreover, alternative phenotypes (A and B in Fig. 2) involve the expression
of modular packages of genes specific to each
phenotype. Another key aspect of developmental
switches is that they integrate variation in the
environment with individual differences in the
genes that regulate the switch. For example, different individuals may have genetically different
thresholds for switching between aggressive and
nonaggressive phenotypes. Finally, the embodied
effects of past experiences and conditions (e.g.,
an individual’s previous exposure to stress or
nutritional conditions early in life) may also
modulate how the switch functions, allowing the
organism to integrate information over time and
across different life stages (Del Giudice 2014b;
Ellis 2013). In many instances, the effects of past
experience on developmental switches may be
mediated by epigenetic mechanisms (see Meaney
2010).
The concept of a developmental switch point
resembles that of a sensitive period, in that the
organism is maximally responsive to some environmental input. The crucial difference is that,
because genetic and environmental inputs converge in the regulatory mechanism, a developmental switch amplifies both environmental and
M. DelGiudice
100
Fig. 2 The concept of a developmental switch. A regulatory mechanism, which may operate through hormonal
signals, integrates current and past information from the
environment with the individual’s genotype. As a result,
the individual’s developmental trajectory is shifted along
alternative pathways—here, A and B—depending on
whether a threshold is reached within the mechanism. The
location of the threshold, the intensity of the signal, and
the timing of the switch point all depend on the joint
action of the current state of the environment, the embodied effect of past environmental conditions, and individual
variation in the genes involved in the regulatory mechanism. Each alternative pathway involves the modular
expression of a set of specific genes, in addition to the
shared genes expressed in the new developmental stage. A
developmental switch may integrate many sources of
input from the environment or produce graded phenotypes
rather than discrete alternatives such as A and B
genetic effects on the phenotype (West-Eberhard
2003). Indeed, the activation of a developmental
switch exposes many new potential sources of
genetic variation, including the genes involved in
the regulatory mechanism and in the expression
of the new phenotypes (Fig. 2).
Del Giudice et al. 2009) encompasses all the
major domains of behavior—from learning and
self-regulation to attachment and sexuality (see
Table 1). My colleagues and I (Del Giudice and
Belsky 2011; Del Giudice et al. 2009, 2012) have
argued that the transition to middle childhood is a
switch point in the development of life history
strategies, which are coordinate suites of morphological, physiological, and behavioral traits
that determine how organisms allocate their
resources to key biological activities such as
growth, reproduction, mating, and parenting (for
a non-technical overview of life history theory,
see Del Giudice et al. 2015). At the level of
2.1
A Switch Point in Life History
Development
The role of adrenarche as a developmental switch
is not limited to a single trait; in fact, the transition to middle childhood (or juvenile transition;
Middle Childhood: An Evolutionary-Developmental Synthesis
101
behavior, individual differences in life history
strategy are reflected in patterns of self-regulation,
aggression, cooperation and prosociality, attachment, sexuality, and so forth (Del Giudice and
Belsky 2011; Del Giudice et al. 2009, 2011; Ellis
et al. 2009). Although life history strategies are
partly heritable, they also show a degree of plasticity in response to the quality of the environment, including the level of danger and
unpredictability –embodied in the experience of
early stress—and the availability of adequate
nutritional resources. In a nutshell, dangerous
and unpredictable environments tend to favor fast
strategies characterized by early reproduction,
sexual promiscuity, unstable relationships,
impulsivity, risk taking, aggression, and exploitative tendencies, whereas safe and predictable
environments tend to entrain slow strategies characterized by late reproduction, stable relationships, high self-control, aversion to risk, and
prosociality. Slow strategies are also favored by
nutritional scarcity when danger is low (see Del
Giudice et al. 2016; Ellis et al. 2009).
Our argument is that adrenarche coordinates
the expression of individual differences in life
history strategy by integrating individual genetic
variation with information about the child’s
social and physical environment collected
throughout infancy and early childhood (Belsky
et al. 1991). The stress response system plays a
major role in gathering and storing information
about environmental safety, predictability, and
availability of resources; adrenarche contributes
by translating that information into adaptive, sexually differentiated patterns of behavior (Del
Giudice et al. 2011; Ellis and Del Giudice 2014).
Consistent with this view, both early relational
stress and early nutrition have been found to
modulate the timing of adrenarche (Ellis and
Essex 2007; Hochberg 2008). It is no coincidence
that the first sexual and romantic attractions typically develop in middle childhood, in tandem
with the intensification of sexual play (Bancroft
2003; Herdt and McClintock 2000). By interacting with peers and adults, juveniles receive feedback about the effectiveness of their nascent
behavioral strategies. The information collected
during middle childhood feeds into the next
developmental switch point, that of gonadarche
(Ellis 2013); the transition to adolescence offers
an opportunity for youth to adjust or revise their
initial strategy before attaining sexual and reproductive maturity (Del Giudice and Belsky 2011).
The role of adrenarche as a switch point in life
history development adds another level of complexity to the biological profile of juvenility.
Figure 3 outlines an integrated evolutionarydevelopmental model that brings together the
various strands of theory and evidence reviewed
in this chapter.
Fig. 3 An integrated evolutionary-developmental model
of middle childhood. Adrenarche is shown as a switch in
the development of life history strategies, as well as a key
mechanism underlying the normative changes of middle
childhood and the emergence and intensification of sex
differences. At a broader level, development in middle
childhood serves two complementary functions, social
integration and social competition
M. DelGiudice
102
3
Three Insights in the Nature
of Middle Childhood
3.1
Insight 1: Social Integration
and Social Competition Are
Complementary Functions
of Middle Childhood
Evolutionary accounts of middle childhood typically focus on learning, helping, and other forms
of social integration. A life history approach
emphasizes the need to consider social competition as a crucial, complementary function of
human juvenility. In the peer group, children compete for vital social resources—status, reputation,
allies, and friends. While learning and play are
relatively risk-free, they are not without consequences. The social position achieved in middle
childhood is a springboard for adolescence and
adulthood; popularity and centrality within the
peer network put a child at a considerable advantage, with potentially long-term effects on mating
and reproductive success (Del Giudice et al. 2009).
Physical and relational aggression are obvious
tactics for gaining influence, but social competition
also occurs through prosocial behaviors such as
forming alliances, doing favors, and displaying
valuable skills. Indeed, managing the balance
between prosocial and coercive tactics is an important part of developing social skills (Hawley 2014).
More broadly, competition shapes many aspects of
cognitive and behavioral development in middle
childhood; for example, increased pragmatic abilities allow children to gossip, joke, tease, and
engage in verbal duels—all forms of social competition mediated by language (Locke and Bogin
2006). Intensifying social competition also contributes to explain the early peak of psychopathology
onset observed in middle childhood, characterized
by increasing rates of externalizing disorders (e.g.,
conduct disorder), anxiety disorders (including
social phobia), and attention deficit hyperactivity
disorder (ADHD; Del Giudice et al. 2009).
3.2
Insight 2: Sexual Selection
Contributes to the Emergence
and Intensification of Sex
Differences in Middle
Childhood
By determining children’s initial place in
social networks and hierarchies, competition
in middle childhood indirectly affects their
ability to attract sexual and romantic partners
later. In other words, middle childhood is a
likely target for sexual selection—that is, natural selection arising from the processes of
choosing mates (mate choice) and competing
for mates (mating competition). My colleagues
and I (Del Giudice et al. 2009) argued that
sexual selection is one reason why sex differences emerge and intensify in middle childhood. In particular, sex differences in physical
aggression increase markedly, in tandem with
sex differences in muscularity and play fighting. At the same time, attachment styles begin
to diverge between males and females, with
insecurely attached boys becoming more
avoidant and insecure girls becoming more
preoccupied/ambivalent (Del Giudice 2009;
Del Giudice and Belsky 2010). Different
attachment styles are conducive to different
social strategies and may be adaptive in regulating children’s nascent relationships with
peers. There is initial evidence that attachment
styles in middle childhood reflect the effects
of prenatal sex hormones, which according to
our model are activated by adrenal androgens
(Del Giudice and Angeleri 2016). Sexual
selection also has indirect implications for the
development of psychopathology; for example, marked sex differences in the prevalence
of conduct disorders become apparent at the
beginning of middle childhood, likely reflecting the stronger role of aggression in boys’
social competition (see Del Giudice et al.
2009; Martel 2013).
Middle Childhood: An Evolutionary-Developmental Synthesis
3.3
Insight 3: In Middle Childhood,
Heightened Sensitivity
to the Environment Goes
Hand in Hand
with the Expression of New
Genetic Factors
When an organism goes through a developmental
switch point, inputs from the environment combine with the individual’s genotype to determine
the resulting phenotype. For example, when adrenal androgens begin to increase during the transition to middle childhood, they activate many
hormone-sensitive brain pathways that have been
dormant since infancy. In doing so, they release
previously hidden genetic variation (Del Giudice
et al. 2009). Thus, middle childhood should be
characterized by a mixture of heightened sensitivity to the environment—possibly mediated by
newly activated epigenetic mechanisms (Meaney
2010) and expression of new genetic factors.
Evidence of increased sensitivity to the environment in middle childhood is not hard to find. Two
intriguing and little-known examples concern the
development of food preferences and erotic
fetishes. In cultures where chili pepper is an essential part of the diet, children tend to dislike spicy
food until middle childhood and then increase rapidly their preference for the flavor of chili as a
result of social learning (Rozin 1990b). Fetishistic
attractions also tend to form in middle childhood,
with the onset of pleasurable sensations toward the
object of the fetish (e.g., rubber, shoes) that later
become fully eroticized (Lawrence 2009). The
onset of fetishistic attractions is part of a generalized awakening of sexuality in middle childhood
(Table 1) and illustrates the potential for rapid plasticity with long-lasting outcomes. Enhanced sensitivity to the environment extends beyond individual
learning to acquiring social norms: for example,
cross-cultural differences in prosocial behavior are
absent in young children but emerge clearly during
middle childhood (House et al. 2013).
On the genetic side of the equation, general
intelligence and language skills increase markedly in heritability from early to middle childhood. In both cases, new genetic factors come
into play around age 7 (Davis et al. 2009; Hayiou-
103
Thomas et al. 2012). Studies of prosociality and
aggression find the same pattern, with new
genetic influences on behavior emerging during
the transition to middle childhood (Knafo and
Plomin 2006; van Beijsterveldt et al. 2003).
These genetic findings dovetail with converging
evidence that individual changes in levels of
aggression are especially frequent during the
transition to juvenility (Del Giudice et al. 2009).
4
Implications for Health
Development
The main focus of this chapter has been on psychological development, but the implications of
the evolutionary-developmental synthesis extend
to both mental and physical health. The transition
to middle childhood seems to be a switch point for
a number of growth and metabolic processes that
have long-term impact on health, including the
risk for obesity and type 2 diabetes (Hochberg
2008, 2010). These processes become apparent in
middle childhood (e.g., anticipated onset of the
adiposity rebound, rapid weight gain, onset of
insulin resistance), but respond to the accumulated
effects of early nutrition and other sources of
stress, starting from prenatal life (e.g., intrauterine
growth restriction; see Salsberry et al. 2017). From
the standpoint of the model presented here, one
can predict that metabolic changes in middle
childhood will reflect both the “programming”
effects of the early environment (Gluckman et al.
2005) and the activation of new genetic factors.
Consistent with this view, a recent study has documented significant genetic correlations between
puberty timing, insulin levels, type 2 diabetes, and
cardiovascular disease (Day et al. 2015). Moreover,
some of those factors are likely to be expressed in
sexually differentiated ways, and the different patterns of health risk associated with early adrenarche and puberty in boys and girls may be
usefully interpreted in light of different constraints
on life history trade-offs in the two sexes (see
Hochberg 2010). These predictions are consistent
with the nonlinear and multilevel nature of developmental processes—one of the guiding principles
of LCHD emphasized in this volume.
104
Another intriguing implication of this perspective is that the juvenile transition may be a promising—and still virtually unexplored—developmental window for intervention. While intervening to
change early life conditions may be desirable in
view of their long-term effects, this approach is not
always possible or realistic. In addition, prenatal
factors such as fetal nutrition and gestational stress
may be especially difficult to target, as they do not
simply mirror the mother’s conditions but reflect a
complex—and partially conflictual—interplay
between fetal and maternal factors (e.g., Del
Giudice 2012; Gangestad et al. 2012; Haig 1993).
However, the logic of developmental switches
(Fig. 2) suggests that the activation of the mechanisms that initiate the switch (e.g., adrenarche)
may correspond to a transient phase of instability
and openness in the system. If so, it should be possible to exploit that phase to maximize the efficacy
of focused interventions—including pharmacological ones. Of course, this would require a better
understanding of how different hormonal and neurobiological systems interact during the transition
to middle childhood; the existing evidence points
to a central role of the hypothalamic-pituitaryadrenal (HPA) axis, the hypothalamic-pituitaryadrenal-thyroid (HPT) axis, and the insulin/
insulin-like growth factor 1 (IGF-1) signaling systems as mediators of life history allocations, not
just in humans but in other vertebrates as well (see
Del Giudice et al. 2015; Ellis and Del Giudice
2014). Those systems might be used as direct targets for intervention, but also as “endophenotypes”
or early indicators of the efficacy of interventions.
Importantly, neurobiological systems such as the
HPA axis regulate both metabolism and behavior—
as components of coordinated life history allocations—so that many of the same processes may be
relevant to both physical and mental health. The
idea that intervening during a biological transition
may afford more leverage to alter developmental
trajectories resonates with two key principles of
LCHD, that is, the nonlinearity of developmental
processes and their time sensitivity.
Probably the most important take-home point
of an evolutionary-developmental approach is
that researchers should be more cautious in
M. DelGiudice
assuming that undesirable developmental outcomes reflect dysregulation of a biological system
(e.g., see Kim et al. 2017) and more open to the
possibility that those outcomes may be part of
adaptive—or formerly adaptive—strategies for
survival and reproduction. As I have argued in
detail elsewhere (Del Giudice 2014b, c), a life history framework is especially useful in teasing out
the logic of potentially adaptive combinations of
traits, highlighting critical factors in the environment, and bridging behavioral development with
physical growth trajectories. As an example, consider the association between intrauterine growth
restriction and early maturation in children
(Hochberg 2008, 2010). This can be interpreted as
a manifestation of physiological dysregulation
due to prenatal adversity or as an adaptive programming effect on children’s metabolic processes and life history trajectories. This alternative
interpretation is supported by the association
between low birth weight, anticipated puberty,
and early childbearing in women (e.g., Nettle
et al. 2013). A third possibility is that low birth
weight partly reflects reduced energetic and metabolic investment by the mother during pregnancy,
which may be adaptive as a component of a fast
life history strategy. If so, the association between
reduced fetal growth and earlier sexual maturation may not be fully causal, but rather result—at
least in part—from shared genetic, or epigenetic,
factors that influence life history strategy in both
the mother and the offspring. Clearly, the implications for intervention are going to be quite different depending on which of these scenarios apply.
Another recent example is the finding that early
adrenarche is associated with reduced white matter volume in the frontal lobe of children (Klauser
et al. 2015). Again, the standard interpretation is
that early DHEA exposure has a disruptive effect
on neurodevelopmental processes; however, it is
also possible that different trajectories of brain
development—and even associated “symptoms”
such as anxiety and aggressive behaviors—may
instead reflect alternative strategies on a fast-slow
continuum of life history variation. Ellis et al.
(2012) present an extended analysis of adolescent
risk-taking from this perspective and discuss sev-
Middle Childhood: An Evolutionary-Developmental Synthesis
eral implications for the design of interventions.
The LCHD principle that evolution both enables
and constrains plasticity is especially relevant in
this regard; evolutionary scenarios are not just
interesting explanatory “stories,” but can illuminate limits as well as opportunities for intervention. For example, when considering mother-fetus
interactions, the existence of partial conflicts of
interest on nutrition, cortisol production, and so on
suggests that interventions designed to favor the
fetus may sometimes have detrimental side effects
for the mother and vice versa (see Del Giudice
2014b; Haig 1993).
In considering potential adaptive explanations
for apparently pathological outcomes, it is
important to remember that biologically adaptive
traits may carry substantial costs. Fitness is ultimately about reproductive success; natural selection does not necessarily promote psychological
well-being or physical health and may sacrifice
survival in exchange for enhanced reproduction.
Moreover, even adaptive developmental processes may result in genuinely maladaptive outcomes for some individuals (Frankenhuis and
Del Giudice 2012). It follows that the existence
of substantial psychological, social, or health
costs does not automatically qualify a trait or
behavior as biologically maladaptive (see Del
Giudice 2014b; Ellis et al. 2012; Ellis and Del
Giudice 2014).
5
Conclusions
We cannot make sense of human development
without understanding middle childhood and its
many apparent paradoxes. An evolutionarydevelopmental approach illuminates the complexity of this life stage and shows how different levels
of analysis—from genes to society—can be tied
together in a coherent synthesis. This emerging
view of middle childhood can help developmental
scientists appreciate its centrality in the human
life history and stimulate ideas for research and
intervention. The study of middle childhood may
finally be ready to come of age, opening up promising avenues for a better understanding of health
development across the life course.
105
References
Auchus, R. J., & Rainey, W. E. (2004). Adrenarche–
physiology, biochemistry and human disease. Clinical
Endocrinology, 60, 288–296.
Bancroft, J. (Ed.). (2003). Sexual development in childhood. Bloomington: Indiana University Press.
Belsky, J., Steinberg, L., & Draper, P. (1991). Childhood
experience, interpersonal development, and reproductive strategy: An evolutionary theory of socialization.
Child Development, 62, 647–670.
Bernstein, R. M., Sterner, K. N., & Wildman, D. E. (2012).
Adrenal androgen production in catarrhine primates
and the evolution of adrenarche. American Journal of
Physical Anthropology, 147, 389–400.
Best, J. R., Miller, P. H., & Jones, L. L. (2009). Executive
functions after age 5: Changes and correlates.
Developmental Review, 29, 180–200.
Bjorklund, D. F. (2011). Children’s thinking: Cognitive
development and individual differences (5th ed.).
Belmont: Wadsworth.
Bogin, B. (1997). Evolutionary hypotheses for human
childhood. Yearbook of Physical Anthropology, 40,
63–89.
Campbell, B. C. (2006). Adrenarche and the evolution
of human life history. American Journal of Human
Biology, 18, 569–589.
Campbell, B. C. (2011). Adrenarche and middle childhood. Human Nature, 22, 327–349.
Davis, O. S. P., Haworth, C. M. A., & Plomin, R. (2009).
Dramatic increase in heritability of cognitive development from early to middle childhood: An 8-year longitudinal study of 8,700 pairs of twins. Psychological
Science, 20, 1301–1308.
Day, F. R., Bulik-Sullivan, B., Hinds, D. A., Finucane, H. K.,
Murabito, J. M., Tung, J. Y., et al. (2015). Genetic determinants of puberty timing in men and women: Shared
genetic aetiology between sexes and with health-related
outcomes. Nature Communications, 6, 8842.
Del Giudice, M. (2009). Sex, attachment, and the development of reproductive strategies. Behavioral and
Brain Sciences, 32, 1–21.
Del Giudice, M. (2012). Fetal programming by maternal stress: Insights from a conflict perspective.
Psychoneuroendocrinology, 37, 1614–1629.
Del Giudice, M. (2014a). Middle childhood: An
evolutionary-developmental
synthesis.
Child
Development Perspectives, 8, 193–200.
Del Giudice, M. (2014b). Early stress and human behavioral development: Emerging evolutionary perspectives. Journal of Developmental Origins of Health and
Disease, 5, 270–280.
Del Giudice, M. (2014c). An evolutionary life history
framework for psychopathology. Psychological
Inquiry, 25, 261–300.
Del Giudice, M., & Angeleri, R. (2016). Digit ratio (2D:4D)
and attachment styles in middle childhood: Indirect
evidence for an organizational effect of sex hormones.
Adaptive Human Behavior and Physiology, 2, 1–10.
106
Del Giudice, M., & Belsky, J. (2010). Sex differences in
attachment emerge in middle childhood: An evolutionary hypothesis. Child Development Perspectives,
4, 97–105.
Del Giudice, M., & Belsky, J. (2011). The development
of life history strategies: Toward a multi-stage theory.
In D. M. Buss & P. H. Hawley (Eds.), The evolution of
personality and individual differences, (pp. 154–176).
New York: Oxford University Press.
Del Giudice, M., Angeleri, R., & Manera, V. (2009).
The juvenile transition: A developmental switch
point in human life history. Developmental Review,
29, 1–31.
Del Giudice, M., Ellis, B. J., & Shirtcliff, E. A. (2011).
The adaptive calibration model of stress responsivity. Neuroscience & Biobehavioral Reviews, 35,
1562–1592.
Del Giudice, M., Angeleri, R., & Manera, V. (2012).
Juvenility and the juvenile transition. In R. J. R.
Levesque (Ed.), Encyclopedia of adolescence, (pp.
1534–1537). New York: Springer.
Del Giudice, M., Gangestad, S. W., & Kaplan, H. S.
(2015). Life history theory and evolutionary psychology. In D. M. Buss (Ed.), The handbook of evolutionary psychology – Vol 1: Foundations (2nd ed.,
pp. 88–114). New York: Wiley.
Del Giudice, M., & Angeleri, R. (2016). Digit ratio (2D:4D)
and attachment styles in middle childhood: Indirect
evidence for an organizational effect of sex hormones.
Adaptive Human Behavior and Physiology, 2, 1–10.
Ellis, B. J. (2013). The hypothalamic-pituitary-gonadal
axis: A switch-controlled, condition-sensitive system
in the regulation of life history strategies. Hormones
and Behavior, 64, 215–225.
Ellis, B. J., & Del Giudice, M. (2014). Beyond allostatic
load: Rethinking the role of stress in regulating human
development. Development and Psychopathology, 26,
1–20.
Ellis, B. J., & Essex, M. J. (2007). Family environments,
adrenarche and sexual maturation: A longitudinal
test of a life history model. Child Development, 78,
1799–1817.
Ellis, B. J., Figueredo, A. J., Brumbach, B. H., & Schlomer,
G. L. (2009). The impact of harsh versus unpredictable
environments on the evolution and development of life
history strategies. Human Nature, 20, 204–268.
Ellis, B. J., Del Giudice, M., Dishion, T. J., Figueredo,
A. J., Gray, P., Griskevicius, V., et al. (2012). The
evolutionary basis of risky adolescent behavior: Implications for science, policy, and practice.
Developmental Psychology, 48, 598–623.
Frankenhuis, W. E., & Del Giudice, M. (2012). When
do adaptive developmental mechanisms yield maladaptive outcomes? Developmental Psychology, 48,
628–642.
Gangestad, S. W., Caldwell Hooper, A. E., & Eaton, M. A.
(2012). On the function of placental corticotropinreleasing hormone: A role in maternal-fetal conflicts over blood glucose concentrations. Biological
Reviews, 87, 856–873.
M. DelGiudice
Geary, D. C. (2010). Male, female: The evolution of
human sex differences. Washington, DC: American
Psychological Association.
Ghetti, S., & Bunge, S. A. (2012). Neural changes
underlying the development of episodic memory
during middle childhood. Developmental Cognitive
Neuroscience, 2, 381–395.
Giedd, J. N., & Rapoport, J. L. (2010). Structural MRI
of pediatric brain development: What have we learned
and where are we going? Neuron, 67, 728–734.
Gluckman, P. D., Hanson, M. A., & Spencer, H. G. (2005).
Predictive adaptive responses and human evolution.
Trends in Ecology & Evolution, 20, 527–533.
Haig, D. (1993). Genetic conflicts in human pregnancy.
QUARTERLY REVIEW OF BIOLOGY, 68, 495–532.
Hawley, P. H. (2014). Ontogeny and social dominance:
A developmental view of human power patterns.
Evolutionary Psychology, 12, 318–342.
Hayiou-Thomas, M. E., Dale, P. S., & Plomin, R. (2012).
The etiology of variation in language skills changes
with development: A longitudinal twin study of language from 2 to 12 years. Developmental Science, 15,
233–249.
Herdt, G., & McClintock, M. (2000). The magical age of
10. Archives of Sexual Behavior, 29, 587–606.
Hochberg, Z. (2008). Juvenility in the context of life history theory. Archives of Disease in Childhood, 93,
534–539.
Hochberg, Z. (2010). Evo-devo of child growth III:
Premature juvenility as an evolutionary trade-off.
Hormone Research in Pædiatrics, 73, 430–437.
House, B. R., Silk, J. B., Henrich, J., Barrett, H. C., Scelza,
B. A., Boyette, A. H., et al. (2013). Ontogeny of prosocial behavior across diverse societies. Proceedings
of the National Academy of Sciences USA, 110,
14586–14591.
Jambon, M., & Smetana, J. G. (2014). Moral complexity
in middle childhood: Children’s evaluations of necessary harm. Developmental Psychology, 50, 22–33.
Joffe, T. H. (1997). Social pressures have selected for
an extended juvenile period in primates. Journal of
Human Evolution, 32, 593–605.
Kaplan, H., Hill, K., Lancaster, J., & Hurtado, A. M. (2000).
A theory of human life history evolution: Diet, intelligence, and longevity. Evolutionary Anthropology, 9,
156–185.
Kim, P., Evans, G. W., Chen, E., Miller, G., & Seeman, T.
(2017). How socioeconomic disadvantages get under
the skin and into the brain to influence health development across the lifespan. In N. Halfon, C. B. Forrest,
R. M. Lerner, & E. Faustman (Eds.), Handbook of life
course health-development science. Cham: Springer.
Klauser, P., Whittle, S., Simmons, J. G., Byrne, M. L.,
Mundy, L. K., Patton, G. C., et al. (2015). Reduced
frontal white matter volume in children with early
onset of adrenarche. Psychoneuroendocrinology, 52,
111–118.
Knafo, A., & Plomin, R. (2006). Prosocial behavior from
early to middle childhood: Genetic and environmental influences on stability and change. Developmental
Psychology, 42, 771–786.
Middle Childhood: An Evolutionary-Developmental Synthesis
Kramer, K. L. (2011). The evolution of human parental care and recruitment of juvenile help. Trends in
Ecology and Evolution, 26, 533–540.
Kuzawa, C. W., Chugani, H. T., Grossman, L. I., Lipovich, L.,
Muzik, O., Hof, P. R., et al. (2014). Metabolic costs and
evolutionary implications of human brain development.
Proceedings of the National Academy of Sciences of
the United States of America, 111, 13010–13015.
Lagattuta, K. H., Sayfan, L., & Blattman, A. J. (2009).
Forgetting common ground: Six- to seven-yearolds have an overinterpretive theory of mind.
Developmental Psychology, 46, 1417–1432.
Lancy, D. F., & Grove, M. A. (2011). Getting noticed:
Middle childhood in cross-cultural perspective.
Human Nature, 22, 281–302.
Lawrence, A. A. (2009). Erotic target location errors: An
underappreciated paraphilic dimension. Journal of Sex
Research, 46, 194–215.
Lebel, C., Walker, L., Leemans, A., Phillips, L., &
Beaulieu, C. (2008). Microstructural maturation
of the human brain from childhood to adulthood.
NeuroImage, 40, 1044–1055.
Locke, J. L., & Bogin, B. (2006). Language and life
history: A new perspective on the development and
evolution of human language. Behavioral and Brain
Sciences, 29, 259–280.
Martel, M. M. (2013). Sexual selection and sex differences in the prevalence of childhood externalizing
and adolescent internalizing disorders. Psychological
Bulletin, 139, 1221–1259.
McClelland, M., Morrison, F., Gestsdóttir, S., Cameron, C.,
Bowers, E. D., Duckworth, A., Little, T., & Grammer.
J. (2017). Self-regulation. In N. Halfon, C. B. Forrest,
R. M. Lerner, & E. Faustman (Eds.), Handbook of life
course health-development science. Cham: Springer.
Meaney, M. J. (2010). Epigenetics and the biological
definition of gene x environment interactions. Child
Development, 81, 41–79.
Nettle, D., Dickins, T. E., Coall, D. A., & de Mornay
Davies, P. (2013). Patterns of physical and psychological development in future teenage mothers. Evolution,
Medicine, and Public Health, 2013, 187–196.
107
Piccardi, L., Leonzi, M., D’Amico, S., Marano, A., &
Guariglia, C. (2014). Development of navigational
working memory: Evidence from 6- to 10-yearold children. British Journal of Developmental
Psychology, 32, 205–217.
Poirel, N., Simon, G., Cassotti, M., Leroux, G., Perchey,
G., Lanoë, C., Lubin, A., Turbelin, M. R., Rossi, S.,
Pineau, A., & Houdé, O. (2011). The shift from local
to global visual processing in 6-year-old children is
associated with grey matter loss. PloS One, 6, e20879.
Rozin, P. (1990a). Development in the food domain.
Developmental Psychology, 26, 555–562.
Rozin, P. (1990b). Getting to like the burn of chili pepper:
Biological, psychological, and cultural perspectives.
In B. G. Green, J. R. Mason, & M. R. Kare (Eds.),
Chemical senses – Volume 2: Irritation. New York:
Dekker.
Salsberry, P., Tanda, R., Anderson, S. E., & Kamboj, M.
K. (2017). Pediatric type 2 diabetes: Prevention and
treatment through a life course health development
framework. In N. Halfon, C. B. Forrest, R. M. Lerner,
& E. Faustman (Eds.), Handbook of life course healthdevelopment science. Cham: Springer.
Scalise Sugiyama, M. (2011). The forager oral tradition
and the evolution of prolonged juvenility. Frontiers in
Psychology, 2, 133.
van Beijsterveldt, T. C. E. M., Bartels, M., Hudziak,
J. J., & Boomsma, D. I. (2003). Causes of stability
of aggression from early childhood to adolescence: A
longitudinal genetic analysis in Dutch twins. Behavior
Genetics, 33, 591–605.
Weisner, T. S. (1996). The 5–7 transition as an ecocultural project. In A. Sameroff & M. Haith (Eds.),
The five to seven year shift: The age of reason and
responsibility (pp. 295–326). Chicago: University of
Chicago Press.
Wells, J. C. K. (2007). Sexual dimorphism of body
composition. Best Practice & Research Clinical
Endocrinology & Metabolism, 21, 415–430.
West-Eberhard, M. J. (2003). Developmental plasticity
and evolution. New York: Oxford University Press.
Open Access This chapter is licensed under the terms of the Creative Commons Attribution 4.0 International License
(http://creativecommons.org/licenses/by/4.0/), which permits use, sharing, adaptation, distribution and reproduction in
any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license and indicate if changes were made.
The images or other third party material in this chapter are included in the chapter’s Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the chapter’s Creative Commons license
and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
Adolescent Health Development:
A Relational Developmental
Systems Perspective
Richard M. Lerner, Claire D. Brindis,
Milena Batanova, and Robert Wm. Blum
Consistent with the United Nations Conventions,
adolescence encompasses the second decade of
life (Lerner and Steinberg 2009). Since the emergence of the study of this portion of the life
course (Hall 1904), adolescence has been
regarded as a period characterized by purportedly
troublesome transformations (e.g., in bodily
attributes associated with puberty; Susman and
Dorn 2009, 2013) and allegedly problematic
transitions (e.g., in regard to socioemotional
functions linked to self-definition or identity,
Erikson 1959, or to changes in the focus of social
The original version of this chapter was revised.
An erratum to this chapter can be found at
https://doi.org/10.1007/978-3-319-47143-3_27
The writing of this chapter was supported in part by grants
from the John Templeton Foundation. We are grateful to
Elizabeth Goodman for her comments
R.M. Lerner, PhD (*)
Tufts University, Medford, MA, USA
e-mail: Richard.lerner@tufts.edu
M. Batanova, PhD
Institute for Applied Research in Youth Development,
Tufts University, Medford, MA, USA
C.D. Brindis, DrPH
University of California-San Francisco (UCSF),
Philip R. Lee Institute for Health Policy Studies and
the Adolescent and Young Adult Health National
Resource Center, San Francisco, CA, USA
R.Wm. Blum, MD, MPH, PhD
Johns Hopkins Bloomberg School of Public Health,
Baltimore, MD, USA
relationships, from parents to peers, Freud 1969).
When viewed from this “deficit” model, adolescents were seen by both scholars and the general
public as both dangerous to themselves and to
society (Anthony 1969). They were regarded as
“problems to be managed” (Roth and BrooksGunn 2003a, b).
Certainly, it may be argued that adolescence is
the most profound period of change within the
life span. As in infancy and early to middle
childhood, the individual’s physiological, psychological, behavioral, and social relationship
characteristics undergo both quantitative and
qualitative changes, that is, transitions and
transformations. For instance, changes in the
prefrontal cortex, increases in the interconnectivity
among brain regions, and increases in dopamine
levels provide both vulnerabilities to risk and
opportunities for growth in cognitive control
(Steinberg 2010). At the same time, most youth
in Western societies are experiencing great
contextual changes, such as changing schools
(e.g., Eccles 2004) and the increased relevance of
peer influences on behavior (e.g., Gardner and
Steinberg 2005). Moreover, in adolescence, the
individual has the cognitive, behavioral, and
social relational skills to contribute actively and
often effectively to his or her own developmental
changes (Lerner 1982; Lerner and BuschRossnagel 1981; Lerner and Walls 1999; Ricco and
Overton 2011). In contrast to earlier developmental periods, adolescents have a burgeoning capacity
© The Author(s) 2018
N. Halfon et al. (eds.), Handbook of Life Course Health Development,
DOI 10.1007/978-3-319-47143-3_6
109
110
for self-governance, for formulating and taking
actions that exert at least some control over their
own development. That is, there is uniquely
marked development of intentional agency in
adolescence. Agency involves self-regulatory
skills that include, for instance, the selection
of goals or the formulation of purposes that are of
importance to adolescents’ developing senses of
self and growing attempts to find a means to
“matter” in their world (Eccles 2004; Freund and
Baltes 2002; Mascolo and Fischer 2015). There
is also growth in the use of executive functioning,
strategic thinking, and behavioral skills in recruiting goal-related resources that enable actions
optimizing the chances of fulfilling their purposes. Furthermore, there is also development of
the ability to compensate effectively when goals
are blocked and/or to select new goals when initial optimization attempts fail and the chances of
attaining an initial goal are lost (Gestsdottir and
Lerner 2008; McClelland et al. 2015).
In general, scholars who study the adolescent
period acknowledge that multiple dimensions of
change involving the individual and his or her
context characterize this portion of the life span
(Lerner et al. 2015; Lerner and Steinberg 2009).
However, substantial variation has emerged in
scholars’ approach to this individual-context
relationship. Analogous to interpreting only main
effects of statistical analyses framed by the
General Linear Model, some scholars have
focused on one domain of change (e.g., adolescent identity development) and have not emphasized other levels of organization within the
ecology of human development (e.g., see Marcia
1980). Other scholars have taken a reductionist
orientation to the phenomena of adolescence and
have sought to use biogenic ideas (e.g., genetic or
evolutionary biological ideas; see Bjorkland and
Ellis, 2005) to interpret all psychological and
social relationship phenomena of the period (see
Elder 1980, 1998; Elder et al. 2015, for critiques
of the main effect or reductionist approaches).
In turn, however, in 2003, Roth and BrooksGunn (2003a, b) reviewed the literature on youth
development programs aimed at enhancing health
and thriving among adolescents. They concluded
that adolescents should be regarded not as problems to be managed but, instead, as resources to
R.M. Lerner et al.
be developed. Their insight built on at least three
sources. First, Larson (2000) provided a compelling vision for research aimed at emphasizing the
strengths of adolescence (e.g., involving their
burgeoning capacities for intentional selfregulation) and for using these strengths to
promote positive youth development (PYD).
Instead of health or positive development being
conceptualized as the diminution or the
absence of disease or of problem behaviors,
respectively, the PYD perspective suggested that
there were facets of thriving that, when promoted,
resulted in healthy and positive development.
Second, Eccles and Gootman (2002) edited a
field-defining report issued by the National
Academy of Sciences about the ways in which
community programs for youth development
could promote several attributes of psychological
and behavioral functioning believed to be indicative of such development. The report included a
summary of the attributes of positive development as involving constructs that could be summarized by “Five Cs:” competence, confidence,
character, connection, and caring (see too, Lerner
et al. 2005). Third, Hamilton (1999) explained
that the idea of PYD was being used in the developmental science literature in three ways: (a) as a
label for a model of the processes through which
health and thriving developed in adolescence, (b)
as a philosophy for or an approach to designing
community-based programs aimed at promoting
thriving, and (c) as instances of such programs.
The emphasis on strengths as compared to
deficits and on links between the positive development of youth and their engagement with contextual resources or ecological “developmental
assets” (Benson et al. 2006), represented by
community-based youth development programs,
reflected the spirit of the times (the zeitgeist) of
developmental science during the latter years of
the twentieth century and the first decade of the
twenty-first century (Lerner 2012; Lerner et al.
2015). Conceptions of human development,
which in earlier periods stressed genetic or maturational determinants of development or
approaches to development that reduced human
behavior to stimulus-response relations, were
being replaced by ideas emphasizing that
development involved mutually influential rela-
Adolescent Health Development: A Relational Developmental Systems Perspective
tions among a person’s biological and psychological characteristics, social relationships, and
influences from families, schools, community
institutions, out-of-school-time (OST) programs,
and both the designed and natural environment.
Developmental scientists argued that all of
these influences, although always present across
life, change in their significance across both life
and across historical periods (e.g., Bronfenbrenner
and Morris 2006; Lerner 2012; Overton 2013,
2015). Indeed, we explain below that, today,
these ideas have a new manifestation quite compatible with the PYD perspective: They are found
in the seven principles of life course health development (see Halfon and Forrest 2017) that
address the conceptualization of the nature and
bases of health and positive behavior development across the life span.
Therefore, in the contemporary study of adolescent development, the main effect and reductionist approaches of the past are regarded as
anachronistic ideas incapable of producing models
that adequately account for the integration of multiple levels of organization within the ecology of
human development (Lerner 2015; Lerner and
Benson 2013a, b; Lerner et al. 2015). In particular,
such traditional approaches overlook the burgeoning information indicating that this integration
reflects systemic relations, that is, mutually influential patterns of covariation within and across
these levels (e.g., see von Bertalanffy 1933, 1968).
At this writing, then, models of adolescent development that are associated with what Overton has
explained as a relational developmental systems
(RDS) metatheory define the cutting edge of theory-predicated scholarship about adolescent
development. This metatheory is derived from a
process-relational paradigm (Overton 2015), and
the concepts associated with RDS thinking are
being used to describe, explain, and optimize the
course of development in the second decade of life
and, as such, to frame applied research aimed at
promoting health and positive development among
diverse adolescents (Lerner et al. 2015).
It is not coincidental that concepts associated
with the RDS metatheory are entirely consistent
with the seven life course health development
principles proposed by Halfon and Forrest
(2017). This congruence of ideas is based on a
111
few common scholarly roots of these two sets of
ideas for understanding human development.
For example, the life course conceptions of Elder
(1998; Elder et al. 2015), the life span developmental ideas of Baltes (e.g., 1997; Baltes et al.
2006), and the constructs associated with
Bronfenbrenner’s (1979, 2005; Bronfenbrenner
and Morris 2006) bioecological model of human
development have shaped both RDS thinking and
the life course health development principles.
It will be useful, then, to discuss the features of
the RDS metatheory and, in this context, point to
the compatibility between RDS concepts and
the life course health development principles.
This discussion will enable us to illustrate how
the ideas associated with both approaches to adolescent development can innovatively integrate
and extend scholarship about transitions and
transformations characterizing the adolescent
period. In addition, because of the emphasis in
both sets of ideas on the concept of relative plasticity in human development—that is, on the
potential for systematic changes in the structure
and function of health assets across the life
course—we will discuss how these two
approaches afford optimism that relational
changes linked to positive change in the health
developmental system can be identified and used
to promote thriving in adolescence.
1
Viewing Adolescent
Development
Through the Lens
of the Relational
Developmental Systems
Metatheory
The study of human development, in general,
and adolescent development, more specifically,
has evolved from a field dominated by split,
reductionist
(psychogenic
or
biogenic)
approaches to a multidisciplinary scholarly field
(Lerner and Steinberg 2009), one that seeks to
integrate variables from biological through cultural and historical levels of organization across
the life span into a synthetic, coactional system
(e.g., Elder 1998; Ford and Lerner 1992; Gottlieb
1997, 1998; Lerner 2012). Reductionist accounts
112
of development that adhere to a Cartesian dualism pull apart (split) facets of the integrated
developmental system (Overton 2015). For
instance, reductionist views typically elevate the
importance of such split formulations as nature
versus nurture, continuity versus discontinuity,
stability versus instability, or basic versus
applied science (Lerner 2002, 2006).
Split approaches are rejected by proponents of
theories derived from an RDS metatheory (e.g.,
Mistry and Wu 2010; Overton 2013; Overton and
Lerner 2014), which in turn are derived from a
process-relational paradigm (Overton 2015).
Across the past four plus decades, several scholars have provided ideas contributing to the evolution of this paradigm (e.g., Baltes 1997; Baltes,
et al., 2006; Brandtstädter 1998; Bronfenbrenner
1979, 2005; Bronfenbrenner and Morris 2006;
Elder 1998; Elder et al. 2015; Ford and Lerner
1992; Nesselroade 1988; Overton 1973; Overton
and Reese 1981; Riegel 1975, 1976; and, even
earlier, see von Bertalanffy 1933).
Overton (e.g., 2013, 2015) has integrated and
extended this scholarship. Overton explains that,
compared to a Cartesian worldview, the processrelational paradigm focuses on process (systematic changes in the developmental system),
becoming (moving from potential to actuality; a
developmental process as having a past, present,
and future; Whitehead 1929/1978), holism (the
meanings of entities and events derive from the
context in which they are embedded), relational
analysis (assessment of the mutually influential
relations within the developmental system), and
the use of multiple perspectives and explanatory
forms (employment of ideas from multiple
theory-based models of change within and of the
developmental system) in understanding human
development. Within the process-relational paradigm, the organism is seen as inherently active,
self-creating (autopoietic), self-organizing, selfregulating (agentic), nonlinear/complex, and
adaptive (Overton 2015; see too Sokol et al.
2015). Similarly, these ideas are echoed in the
life course health development principles (specifically, Nos. 1 and 3) that health development
emerges as a consequence of complex, nonlinear
process that results from person⇔environment
R.M. Lerner et al.
coactions that are multidimensional, multidirectional, and multilevel.
Accordingly, both within the RDS approach to
theory and the life course health development
principles, split conceptions are eschewed in
favor of a metatheory that emphasizes the study
and integration of different levels of organization, ranging from biology/physiology to culture
and history, as a means to understand life span
human development (Lerner 2006; Overton
2013, 2015), the production of health, and the
development of disease. Accordingly, conceptual
emphasis is placed on mutually influential relations between individuals and contexts, represented as individual ⇔ context relations.
This representation of the coactions between
person and setting within RDS-based models is
not meant to convey a person-context interaction—which is typically represented in the
developmental literature as person X context. An
interaction connotes that the entities involved in
the relation are separate and independent (as in a
statistical interaction) and that, as such, their
association involves a linear combination of discrete and separate variables. Both before and
after the interaction, these entities (variables) are
independent and unchanged by each other. The
bidirectional arrow used in the RDS illustration
of person ⇔ context relations is intended to
emphasize that the coaction of individual and
context involves the entire developmental system. As such, the relations among levels of the
autopoietic system, and not independent linear
combinatorial attributes, are the focus in such a
model. Indeed, the fusion of individual and context within the developmental system means that
across ontogeny (across the life span), any portion of the system is inextricably embedded
with—or embodied by, in Overton’s (2013,
2015) conceptualization—all other portions of
the developmental system. Embodiment refers
to the way individuals behave, experience, and
live in the world by being active agents with particular kinds of bodies; the body is integratively
understood as a form (a biological referent), as
lived experience (a psychological referent), and
as an entity in active engagement with the world
(a sociocultural referent) (Overton 2015).
Adolescent Health Development: A Relational Developmental Systems Perspective
113
Similarly, within the life course health development perspective, health development is also
embodied: It is an emergent property of living
systems (Principle 1), and it develops continuously across the life span (Principle 2).
Within the context of such a bidirectional relational system, the embeddedness within history
(temporality) is of fundamental significance
(Elder 1998; Elder et al. 2015). We may note that
the developmental system is embedded in history
(temporality). This embeddedness means that
change is constant in the developmental system
and that, as such, there may be either stochastic
or systematic changes in person⇔context relations across time and place (Elder 1998; Elder
et al. 2015; Misteli 2013). Sensitivity of change
to time and place is also a key idea within the life
course health development Principle 4, which
states that health development is highly sensitive
to the timing and social structure of environmental exposures. The presence of such temporality
in individual ⇔ context relations within the
developmental system means that there always
exists some potential for systematic change and,
thus, for (relative) plasticity in human development. In short, potential plasticity in individual
⇔ context relations derives from the “arrow of
time” (Lerner 1984; Lerner and Benson 2013a, b;
Overton 2013) running through the integrated
(relational) developmental system.
focus in developmental analysis. He notes that the
process-relational paradigm involves different
moments within a research program.
One moment involves the idea of the identity of
opposites, a second moment involves the opposites
of identity, and a third (relationally integrative)
moment involves the synthesis of wholes. In discussing these three moments of scientific analysis
within RDS approaches to developmental science,
we point to the predominant trait model of individuality, the Five-Factor Theory (FFT, involving the
purported Big Five “personality traits” of conscientiousness, agreeableness, neuroticism, openness to
experience, and extraversion; Costa and McCrae
1980, 2006; McCrae et al. 2000). The FFT example
is a means to explain these RDS-based moments
and to contrast their use with thinking associated
with Cartesian, split, reductionist approaches to the
study of the individual development.
The first moment recognizes that both individual and context define— and are mutually
constituted by—each other in one moment, or
point, in a program of developmental inquiry.
Overton (2010, p. 14) notes that:
2
The identity of opposites, therefore, emphasizes
the fused person ⇔ context relationship as the
primary unit of analysis for understanding development. As such, in this moment of research,
developmental scientists would reject the idea
that there are any aspects of human behavioral
development— for instance, entities such as
traits—that “are more or less immune to environmental influences” (McCrae et al. 2000, pp. 175);
the idea that such entities are indicators of split
notions reflecting “nature over nurture” (McCrae
et al. 2000, p. 173) would also be rejected by
developmental scientists working within this first
moment of analysis.
The second moment that Overton (2010,
2013, 2015; Overton and Müller 2013) discusses
is the opposites of identity. This moment allows
Three Moments of Analysis
in an RDS Approach
to Adolescent Health
Development
To elucidate the role of time and place in contributing to the bidirectional relations of focal concern
within RDS metatheory, developmental scientists
may focus on either the role of the individual or the
context,
in
particular
instantiations
of
individual⇔context exchanges. This focus may
seem contradictory to the fusion among levels of
organization emphasized in this approach. However,
as we noted earlier, Overton (2015) embeds the
RDS metatheory in the process-relational paradigm
(see, as well, Sokol et al. 2015). Overton uses this
paradigm to explain the possibility of this changing
The principle of the identity of opposites establishes the identity among parts of a whole by casting them not as exclusive contradictions as in the
split epistemology, but as differentiated polarities
(i.e., coequals) of a unified (i.e., indissociable)
inclusive matrix—as a relation. As differentiations, each pole defines and is defined by its
opposite.
R.M. Lerner et al.
114
one, in effect, to hold the other parts of the
integrated system in abeyance and focus on one
part of the system; however, the ultimate aim is
one of reintegrating the part into the whole at a
subsequent moment. Overton (2013, pp. 47–48)
explains that:
The limitation of the identity moment of analysis is
that, in establishing a flow of categories of one into
the other, a stable base for inquiry that was provided by bedrock material atoms of the split
metatheory is eliminated…Reestablishing a stable
base—not an absolute fixity, nor an absolute relativity, but a relative relativity (Latour 1993)—
within relational metatheory requires moving to a
second moment of analysis. In this moment of
opposition, the law of contradiction is reasserted
and categories again exclude each other. As a consequence of this exclusion, parts exhibit unique
identities that differentiate each from the other.
Therefore, when functioning within this second moment of analysis, developmental scientists could focus solely on attributes of
individuals, for instance, the purported traits of
conscientiousness, agreeableness, neuroticism,
openness to experience, and extraversion that
comprise the Big Five components of the FFT
(Costa and McCrae 1980, 2006; McCrae et al.
2000) and, for instance, study the psychometric
properties of these constructs to provide “objective,” or quantitative, indices of these attributes.
Indeed, such psychometric work has often been
a part of research programs framed by RDS
models (e.g., Damon 2008; Lerner et al. 2015).
Thus, working within this second moment of
analysis, developmental scientists following an
RDS-based model, and social or personality
researchers using a Cartesian split model, would
be engaging in commensurate work. However,
the difference between these two groups of
scholars is brought to the fore when the third
moment of analysis discussed by Overton
(2015) is considered.
The third moment, the synthesis of wholes,
occurs when the first two moments are embedded
in a multi-perspective process-relational paradigm and are recognized as mutually necessary
in a systematic, integrative program of research,
wherein one needs both of the first two moments.
That is, “A complete relational program requires
principles according to which the individual
identity of each concept of a formerly dichotomous pair is maintained while simultaneously it
is affirmed that each concept constitutes, and is
constituted by the other” (Overton and Müller
2013, p. 35).
Accordingly, the developmental scientist
working within an RDS model would use an
“objective” measure studied within the second
moment of analysis within an integrated, relational empirical approach that focused on the
individual⇔context relation. Clearly, the trait
theorist would not take such a step, given that the
work of such scholars is framed by the ideas,
noted above, that context is irrelevant to the
understanding (i.e., successful prediction) of the
life course of the manifestation of traits. Indeed,
in their belief “that personality traits are more or
less immune to environmental influences”
(McCrae et al. 2000, pp. 175), trait theorists
maintain that contextual conditions, whether
similar or not, are irrelevant to prediction; given
the purported biological base of traits, only nature
variables have predictive efficacy.
However, there is abundant evidence that purported traits are in fact not “trait-like” at all, that
is, these attributes reflect relations between individuals and contexts, as they occur at particular
times and places (Ardelt 2000; Block 1995,
2010; Elder 1998; Elder et al. 2015; Roberts
et al. 2006). Indeed, methodological work framed
by RDS concepts (e.g., Molenaar and Nesselroade
2014, 2015; Nesselroade 1988; Nesselroade and
Molenaar 2010) indicates that the purported life
span stability of traits, as well as the purported
immunity to contextual influences, is empirically
counterfactual. Moreover, underscoring the importance of this third moment of analysis discussed by
Overton (2015), these methodological innovations
demonstrate the ability to index with psychometric
precision integrative individual ⇔ context (including individual ⇔ individual) units of analysis (e.g.,
Molenaar 2014; Molenaar et al. 2014; Molenaar and
Nesselroade 2015). This innovation in developmental methodology is important for understanding
key features of the individual⇔context relational
process propelling developmental change in
adolescence.
Adolescent Health Development: A Relational Developmental Systems Perspective
3
Developmental Regulations,
Adaptive Developmental
Regulations, and Human
Agency in RDS Metatheory
Given the analytic moment of the identity of
opposites—that each component of the developmental system constitutes, and is constituted by,
the other components of the system—RDS
metatheory focuses on the “rules” or processes
which govern, or regulate, exchanges between
individuals and their contexts. Such processes are
the function of the developmental system. An
RDS program of research might seek to understand the nature of relations between individuals
and their contexts, including the dynamics of
those relations across the life course. For instance,
RDS-based research might ask how specific features of the individual and specific features of the
context coalesce to influence the substantive
course of individual⇔individual relations.
Brandtstädter (1998) termed these bidirectional
relations “developmental regulations” and noted
that, when developmental regulations involve
mutually beneficial individual⇔context relations,
then these developmental regulations are adaptive. Developmental regulations are the fundamental feature of human life, that is, all human
life exists in a context and involves bidirectional
exchanges with it (Darwin 1859; Forrest 2014;
Tobach and Schneirla 1968). These exchanges
involve physiological systems and functions
(e.g., respiration, circulation, digestion, reproduction) and behaviors (e.g., social affiliation
and cooperation, or aggression and competition,
as might be involved in protection, hunting, and
scavenging; Johanson and Edey 1981) and
involve both organismic self-regulation (e.g.,
hypothalamic functioning, circadian rhythms) and
intentional self-regulation (e.g., goal selection,
resource recruitment, and executive functioning;
Gestsdóttir and Lerner 2008).
Gestsdóttir et al. (2014) note that self-regulation
is a multidimensional construct, involving a range
of behaviors, from basic physiological functions
to complex intentional cognitive processes (e.g.,
Bandura 2001; Brandtstädter 1998; McClelland
et al. 2015). As such, self-regulation pertains to all
115
aspects of adaption, as people alter their thoughts,
attention, emotions, and behaviors to react to contextual events and, as well, to influence selected
features of their contexts. Here, culture plays a key
moderating role. Trommsdorff (2012) notes that
self-regulation “is assumed to develop by organizing inner mental processes and behavior in line
with cultural values, social expectations, internalized standards, and one’s self-construal” (p. 19).
The developmental course of self-regulation is,
in effect, the developmental course of human
agency in the context of individual desires, purposes, needs, goals and identity, perceptions of and
coactions with other people, the physical ecology,
and culture (e.g., Damon 2008; Geldhof et al. 2010;
Gestsdóttir and Lerner 2008; Lerner et al. 2001).
Agency is a defining feature of the active, self-creating (autopoietic, enactive), and nonlinear adaptive living system (Overton and Lerner 2014;
Narvaez 2008; Witherington 2014). Such agency is
the individual’s contribution to adaptive developmental regulations (Brandtstädter 1998, 1999).
The development of agency begins in early
life, primarily with organismic self-regulation
processes. However, as we have noted, by the
time of adolescence, self-regulation is increasingly intentional and purposeful (Damon 2008)
and involves the self-system and the phenomena
associated with identity development (Gestsdóttir
and Lerner 2008; Lerner et al. 2001). The adolescent instantiation of agency reflects ideas associated with the work of comparative psychologists
Tobach and Schneirla (1968), who distinguished
between the biosocial functioning of insects (e.g.,
ants) and the psychosocial functioning of organisms with higher psychological levels, levels that
are marked by greater plasticity, rather than stereotypy, in their eventual highest levels of
ontogenetic change. Such higher levels provide
the physiological base for symbolic functioning.
In turn, reflecting the life course health development principles that evolution enables as well
as constrains health development pathways and
plasticity (Principle No. 5) and that optimal
health development promotes survival, enhances
thriving, and protects against disease (Principle
No. 6), evolutionary biologists Jablonka and
Lamb (2005) note that both psychological
R.M. Lerner et al.
116
processes and cultural processes are integrated
with the genetic and epigenetic processes of evolution to make human adaptiveness and contributions qualitatively different than corresponding
instances of adaptiveness of social contributions
among other organisms. Together, these comparative psychology and evolutionary biology literatures suggest that, among humans, adaptiveness
and positive contributions reflect integrated cognitive, emotional, and behavioral processes that
involve abstract, symbolic constructs, such as
language or moral reasoning, or character virtues
(e.g., Lerner and Callina 2014).
of the developmental systems—that is, molecular,
physiological, behavioral, cultural, and evolutionary
processes (e.g., Cole 2014; Meaney 2014; Slavich
and Cole 2013)—and with the Bornstein (2017)
“specificity principle,” we suggest, therefore, that
addressing a multipart “what” question is the key to
conducting programmatic research about the function, structure, and content of health development
in adolescence. In other words, to test RDS-based
ideas about the ontogenetically changing structure
of adolescent development in general and health
development more specifically, researchers need to
ascertain answers to the following multipart “what”
questions:
4
1. What specific structure-content relations
emerge; at
2. What specific levels of organization within
the relational developmental system that are
linked to;
3. What specific antecedent and consequent
adaptive developmental regulations (to what
trajectory of individual⇔context relations); at
4. What specific points in adolescent development; for
5. What specific groups of youth; living in
6. What specific contexts; at
7. What specific points in time (history)?
Implications for Research
About Adolescent Health
Development
Empirically, indexing such facets of adolescent
health development, in general, or the complex
dimensions of evolutionary change linked to adolescent health development more specifically
(e.g., Gissis and Jablonka 2011; Lerner et al.
2015; Slavich and Cole 2013) may involve both
point-in-time (cross-sectional) assessments and
historical (longitudinal) assessments (Lerner
2004). Scientists must conduct such assessments
within the context of recognizing that contexts
are complex (e.g., they exist at multiple levels of
organization; Bronfenbrenner 1979). Individuals
cannot necessarily act in ways that benefit all levels and all components of the context at all
times and places (Elder 1998). Thus, adaption
is not treated as a categorical concept (as something that either exists or not) but, instead, as a
multivariate concept comprised of ordinal or
interval dimensions. Researchers studying
adaptation would not ask, then, whether it
exists or not; rather, the question would be how
beneficial is the developmental regulation (the
individual⇔context relation) for specific people
or specific social institutions of the context, at
specific times and in specific places (e.g., see
Bornstein 2006, 2017).
Consistent both with the seventh principle of
life course health development, that is, that the
cadence of human health development results from
the synchronized timing of variables from all levels
Such work may have several benefits.
Questions about coherence and reliability or consistency of indicators of health development with
respect to variation in context may be especially
useful for understanding the developmental
course of multidimensional, latent variable
conceptions of health among diverse adolescents.
For instance, are specific indicators of, say, cardiovascular, respiratory, nutritional, or mental health
manifested consistently across time and place?
To what extent and for whom and under which
conditions can biologic propensities to such
health developmental challenges be moderated?
Consider nutrition more closely. How do adolescents’ diets influence their cardio-metabolic
risk as adolescents and adults? Do adolescents
maintain a healthy diet across different settings
involving peers and family members? Is there
age-associated variation in answers to this
question, for instance, variation associated with
Adolescent Health Development: A Relational Developmental Systems Perspective
pubertal development or pubertal status/stage
(e.g., early, on time, or late)? Do answers to these
questions vary in relation to gender? As well, do
they vary in relation to normative social transitions (e.g., moving from elementary school to
middle school)? How do peer social networks
influence these behaviors or family mealtime
behaviors? Are answers moderated or changed
by the socioeconomic, cultural, religious, or
national contexts of youth? In turn, do answers
here vary in relation to media exposure about eating and about desirable body types that may be
prevalent for youth living in particular settings at
particular times in history? Given the synchronization issues raised by the seventh life course
health development principle, how are all these
answers moderated or changed by what may be
nonnormative life or historical events in the lives
of adolescents (Baltes et al. 2006)? Examples
here may be the death of a parent or deployment
in the military (Cozza and Lerner 2013), family
disruption due to divorce or separation, or family
challenges in the face of environmental tragedies
such as weather-related calamities or living in
settings beset by violence.
The life course health development principles
remind us that health development is highly sensitive to the timing and social structure of environmental events (Principle 4), and these
examples of questions reflecting the specificity of
health-related individual⇔context relations that
may impinge on the thriving of adolescents
underscore the subtle and nuanced nature of the
developmental system within which adolescent
health development is embodied.
The idea of embodiment and of adolescent
health development being an emergent property
of the living system (Principle 1) underscores
that it is insufficient to take a “main effect”
approach to the study of even a facet of health
development as complex as the effects of nutrition.
Scientists and practitioners framing their research
and applications, respectively, within RDS-based
models and/or life course health development
principles must also ask how do answers to all the
questions we have posed regarding nutrition
covary with comparable questions pertaining to
adolescent lifestyle changes (e.g., involving
117
sleep, exercise, or sexual debut), healthy status of
bodily systems, and the presence of chronic and
acute health challenges, of both more biological
(e.g., hormonal changes) and more social origins
(e.g., school bullying).
5
Conclusions
The interrelated “what” questions we have presented, illustrated by the example of nutrition’s
effects on health development, will, then, help
developmental and medical science collaborate to
understand the assuredly complex questions that
need to be addressed in a scholarly agenda that
comprehensively describes, explains, and optimizes the course of individual health development
across the adolescent period. Developmental scientists know that in some senses each adolescent
is like every other adolescent (e.g., there are
nomothetic principles that apply to functioning of
the physiological and psychological changes prototypic of the period), that each adolescent is like
only some other adolescent (e.g., group differential variation exists as, for instance, when both the
presence of good health status and healthy disparities vary across gender, race, socioeconomic level,
culture, and nation of residence), and that each
adolescent is like no other adolescent (e.g., there
are idiographic characteristics pertinent to every
young person, for instance, resulting from the
relations among his/her timing of molecular,
physiological, behavioral, cultural, and evolutionary processes brought into high relief by life
course health development Principle 7). Therefore,
the scholarly agenda brought to the fore by RDS
metatheory and by the principles of life course
health development will require scholars seeking
to understand and to promote adolescent health to
accept that their work will be exceedingly more
complex than work predicated on main effect or
reductionist approaches.
However, the relative plasticity in the
development of human health and positive development that is found in studies that have been
derived from or associated with RDS-based or life
course health development-related scholarship,
respectively (e.g., Lerner et al. 2015), is a basis
118
for optimism that embracing such complexity is
not only the intellectually correct path to take. It is
also a feasible approach to adopt. The advances in
relational developmental systems-predicated
methods that we have discussed (e.g., Molenaar
et al. 2014; Molenaar and Nesselroade 2014,
2015; Nesselroade and Molenaar 2010) provide
powerful research and data analytic tools enabling
such complexity to be integratively assessed.
The adolescent decade is “privileged” because of
the generally disease-free status of individuals
within it, as compared to subsequent decades of life
(e.g., Susman and Dorn 2009; Paus 2009).
Nevertheless, application of methods linked to the
ideas of RDS metatheory and the principles of life
course health development hold the promise of narrowing the disparities that exist across time and
place in adolescent health development. In addition,
therefore, these conceptions have one other vital
asset; their use in research and application will
enhance the probability that all youth will transition
from adolescence into the adult years in manners
that will enable their life course trajectories to more
prominently be marked by health and thriving. Such
a contribution by these conceptual frameworks will
make quite vivid the insight of Lewin (1954) that
there is nothing more practical than a good theory.
Acknowledgement Supported (in full or in part) by
Grant # U45MC27709 from the Department of Health and
Human Services, Health Resources and Services
Administration, Maternal and Child Health Bureau (Title
V, Social Security Act), Division of Child, Adolescent and
Family Health, Adolescent Health Branch.
References
Anthony, E. J. (1969). The reactions of adults to adolescents and their behavior. In G. Caplan & S. Lebovici
(Eds.), Adolescence: Psychosocial perspectives
(p. 77). New York: Basic Books.
Ardelt, M. (2000). Still stable after all these years?
Personality stability theory revisited. Social
Psychology Quarterly, 63(4), 392–405.
Baltes, P. B. (1997). On the incomplete architecture of
human ontogeny: Selection, optimization, and compensation as foundations of developmental theory.
American Psychologist, 52, 366–380.
Baltes, P. B., Lindenberger, U., & Staudinger, U. M.
(2006). Lifespan theory in developmental psychology.
In W. Damon & R. M. Lerner (Eds.), Theoretical mod-
R.M. Lerner et al.
els of human development. Vol. 1: Handbook of child
psychology (6th ed., pp. 569–664). Hoboken: Wiley.
Bandura, A. (2001). Social cognitive theory: An agentic
perspective. Annual Review of Psychology, 52 1–26.
Palo Alto: Annual Reviews, Inc.
Benson, P. L., Scales, P. C., Hamilton, S. F., & Semsa,
A., Jr. (2006). Positive youth development: Theory,
research, and applications. In R. M. Lerner (Ed.),
Theoretical models of human development, Volume
1 of handbook of child psychology (6th ed.,
pp. 894–941). Editors-in-chief: W. Damon & R. M.
Lerner. Hoboken: Wiley.
Bjorklund, D. F., & Ellis, B. J. (2005). Evolutionary psychology and child development: An emerging synthesis. In B. J. Ellis & D. F. Bjorklund (Eds.), Origins
of the social mind: Evolutionary psychology and child
development (pp. 3–18). New York: Guilford.
Block, J. (1995). A contrarian view of the five-factor
approach to personality description. Psychological
Bulletin, 117, 187–215.
Block, J. (2010). The five-factor framing of personality and beyond: Some ruminations. Psychological
Inquiry, 21(1), 2–25.
Bornstein, M. H. (2006). Parenting science and practice. In K. A. Renninger, & I. E. Sigel (Vol. Eds.),
Handbook of child psychology, Vol. 4: Child psychology in practice (6th ed., pp. 893–949). Editors-inChief: W. Damon, & R. M. Lerner. Hoboken: Wiley.
Bornstein, M. H. (2017). The specificity principle in
acculturation science. Perspectives on Psychological
Science, 12(1), 3–45.
Brandtstädter, J. (1998). Action perspectives on human
development. In R. M. Lerner (Ed.), Handbook
of child psychology, Vol. 1 (5th ed., pp. 807–866).
New York: Wiley.
Brandtstädter, J. (1999). The self in action and development: Cultural, biosocial, and ontogenetic bases of
intentional self-development. In J. Brandtstädter &
R. M. Lerner (Eds.), Action and self-development:
Theory and research through the life-span (pp. 37–65).
Thousand Oaks: Sage.
Bronfenbrenner, U. (1979). The ecology of human development. Cambridge, MA: Harvard University Press.
Bronfenbrenner, U. (2005). Making human beings human.
Thousand Oaks: Sage Publications.
Bronfenbrenner, U., & Morris, P. A. (2006). The bioecological model of human development. In R. M. Lerner
(Ed.), Theoretical models of human development.
Volume 1 of Handbook of child psychology (6th ed.,
pp. 795–828). Editors-in-chief: W. Damon & R. M.
Lerner. Hoboken: Wiley.
Cole, S. W. (2014). Human social genomics. PLoS
Genetics, 10(8), 1–7.
Costa, P. T., Jr., & McCrae, R. R. (1980). Still stable after
all these years: Personality as a key to some issues in
adulthood and old age. In P. B. Baltes & O. G. Brim
Jr. (Eds.), Life span development and behavior (Vol. 3,
pp. 65–102). New York: Academic Press.
Costa, P. T., Jr., & McCrae, R. R. (2006). Age changes in
personality and their origins: Comment on Roberts,
Adolescent Health Development: A Relational Developmental Systems Perspective
Walton, and Viechtbauer (2006). Psychological
Bulletin, 132, 28–30.
Cozza, S. J., & Lerner, R. M. (Eds.). (2013). Military children and families. The Future of Children, 23(2), 3–11.
Damon, W. (2008). The path to purpose: Helping our
children find their calling in life. New York: Simon
and Schuster.
Darwin, C. (1859). The origin of species by means of natural selection or the preservation of favoured races in
the struggle for life. London: J. Murray.
Eccles, J. S. (2004). Schools, academic motivation, and
stage-environment fit. In R. M. Lerner & L. Steinberg
(Eds.), Handbook of adolescent psychology (2nd ed.,
pp. 125–153). Hoboken: Wiley.
Eccles, J. S., & Gootman, J. A. (Eds.). (2002). Community
programs to promote youth development/committee
on community-level programs for youth. Washington,
DC: National Academy Press.
Elder, G. H., Jr. (1980). Adolescence in historical perspective. In J. Adelson (Ed.), Handbook of adolescent psychology (pp. 3–46). New York: Wiley.
Elder, G. H., Jr. (1998). The life course and human development. In R. M. Lerner (Ed.), Handbook of child
psychology, Volume 1: Theoretical models of human
development (5th ed., pp. 939–991). Editor-in-chief:
W. Damon. New York: Wiley.
Elder, G. H., Jr., Shanahan, M. J., & Jennings, J. A.
(2015). Human development in time and place. In
M. H. Bornstein and T. Leventhal (Eds.), Handbook of
child psychology and developmental science, Volume
4: Ecological settings and processes in developmental systems (7th ed., pp. 6–54). Editor-in-chief: R. M.
Lerner. Hoboken: Wiley.
Ford, D. L., & Lerner, R. M. (1992). Developmental systems
theory: An integrative approach. Newbury Park: Sage.
Forrest, C. B. (2014). A living systems perspective on
health. Medical Hypotheses, 82, 209–2143.
Freud, A. (1969). Adolescence as a developmental disturbance. In G. Caplan & S. Lebovici (Eds.), Adolescence
(pp. 5–10). New York: Basic Books.
Freund, A. M., & Baltes, P. B. (2002). Life-management
strategies of selection, optimization and compensation: Measurement by self-report and construct validity. Journal of Personality and Social Psychology,
82(4), 642–662.
Gardner, M., & Steinberg, L. (2005). Peer influence on
risk taking, risk preference, and risky decision making
in adolescence and adulthood: An experimental study.
Developmental Psychology, 41, 625–635.
Geldhof, G. J., Little, T. D., & Colombo, J. (2010).
Self-regulation across the lifespan. In M. E. Lamb
& A. M. Freund (Vol. Eds.), Social and emotional
development. Volume 2 of the Handbook of lifespan development. Editor-in-Chief: R. M. Lerner.
Hoboken: Wiley.
Gestsdóttir, G., & Lerner, R. M. (2008). Positive development in adolescence: The development and role of
intentional self regulation. Human Development, 51,
202–224.
Gestsdottir, S., Geldhof, G. J., Paus, T., Freund, A. M.,
Aðalbjarnardóttir, S., Lerner, J. V., & Lerner, R. M.
119
(2014). Self-regulation among youth in four Western
cultures: Is there an adolescence-specific structure of the Selection-Optimization-Compensation
(SOC) model? International Journal of Behavioral
Development, 39(4), 346–358.
Gissis, S. B., & Jablonka, E. (Eds.). (2011).
Transformations of Lamarchism: From subtle fluids to
molecular biology. Cambridge, MA: The MIT Press.
Gottlieb, G. (1997). Synthesizing nature-nurture: Prenatal
roots of instinctive behavior. Mahwah: Erlbaum.
Gottlieb, G. (1998). Normally occurring environmental
and behavioral influences on gene activity: From central dogma to probabilistic epigenesis. Psychological
Review, 105(4), 792–802.
Halfon, N., & Forrest, C. B. (2017). The emerging theoretical framework of life course health development. In
N. Halfon, C. B. Forrest, R. M. Lerner, & E. Faustman
(Eds.), Handbook of life course health-development
science. Cham: Springer.
Hall, G. S. (1904). Adolescence: Its psychology and its
relations to psychology. anthropology, sociology, sex,
crime, religion, and education. New York: Appleton.
Hamilton, S. F. (1999). A three-part definition of youth
development. Unpublished manuscript. Ithaca: Cornell
University College of Human Ecology.
Jablonka, E., & Lamb, M. J. (2005). Evolution in four
dimensions: Genetic, epigenetic, behavioral, and symbolic variation in the history of life. Cambridge, MA:
MIT Press.
Johanson, D. C., & Edey, M. A. (1981). Lucy: The beginnings of humankind. New York: Simon & Schuster.
Larson, R. W. (2000). Toward a psychology of positive
youth development. American Psychologist, 55(1),
170–183.
Latour, B. (1993). We have never been modern (trans.
Catherine Porter), Cambridge, MA: Harvard
University Press.
Lerner, R. M., & Busch-Rossnagel, N. A. (Eds.). (1981).
Individuals as producers of their development: A lifespan perspective. New York: Academic Press.
Lerner, R. M. (1982). Children and adolescents as producers of their own development. Developmental Review,
2, 342–370.
Lerner, R. M. (1984). On the nature of human plasticity.
New York: Cambridge University Press.
Lerner, R. M., & Walls, T. (1999). Revisiting individuals as producers of their development: From dynamic
interactionism to developmental systems. In J.
Brandtstädter & R. M. Lerner (Eds.), Action and selfdevelopment: Theory and research through the lifespan (pp. 3–36). Thousand Oaks: Sage.
Lerner, R. M. (2002). Concepts and theories of human
development (3rd ed.). Mahwah: Lawrence Erlbaum
Associates.
Lerner, R. M. (2004). Liberty: Thriving and civic engagement among America’s youth. Thousand Oaks: Sage
Publications.
Lerner, R. M., Lerner, J. V., Almerigi, J. B., Theokas, C.,
Phelps, E., Gestsdottir, S., et al. (2005). Positive Youth
Development, Participation in community youth
development programs, and community contribu-
120
tions of fifthgrade adolescents: Findings from the first
wave of the 4-H study of Positive Youth Development.
Journal of Early Adolescence, 25(1), 17–71.
Lerner, R. M. (Ed.). (2006). Theoretical models of human
development. Volume 1 of Handbook of child psychology
(6th ed.). Editors-in-chief: W. Damon & R. M. Lerner.
Hoboken: Wiley.
Lerner, R. M. (2012). Essay review: Developmental science: Past, present, and future. International Journal
of Developmental Science, 6(1–2), 29–36.
Lerner, R. M., & Callina, K. S. (2014). The study of
character development: Towards tests of a relational
developmental systems model. Human Development,
57(6), 322–346.
Lerner, R. M. (Editor-in-Chief). (2015). Handbook of
child psychology and developmental science (7th ed.).
Hoboken: Wiley.
Lerner, R. M., & Benson, J. B. (Eds.). (2013a). Embodiment
and epigenesis: Theoretical and methodological issues
in understanding the role of biology within the relational
developmental system. Volume 1: Philosophical, theoretical, and biological dimensions. advances in child
development and behavior (Vol. 44). London: Elsevier.
Lerner, R. M., & Benson, J. B. (Eds.). (2013b).
Embodiment and epigenesis: Theoretical and methodological issues in understanding the role of biology
within the relational developmental system. Volume 2:
Ontogenetic dimensions. Advances in child development and behavior (Vol. 45). London: Elsevier.
Lerner, R. M., & Callina, K. S. (2015). The study of character development: Towards tests of a relational developmental systems model. Human Development, 57(6),
322–346.
Lerner, R. M., & Steinberg, L. (2009). The scientific
study of adolescent development. In R. M. Lerner &
L. Steinberg (Eds.), Handbook of adolescent psychology (3rd ed., pp. 3–14). Hoboken: Wiley.
Lerner, R. M., Freund, A. M., De Stefanis, I., & Habermas,
T. (2001). Understanding developmental regulation in
adolescence: The use of the selection, optimization, and
compensation model. Human Development, 44, 29–50.
Lerner, R. M., Lerner, J. V., Bowers, E., & Geldhof, G. J.
(2015). Positive youth development: A relational
developmental systems model. In W. F. Overton &
P. C. Molenaar (Eds.), Theory and method. Volume
1 of the Handbook of child psychology and developmental science (7th ed., pp. 607–651). Editor-in-chief:
R. M. Lerner. Hoboken: Wiley.
Lewin, K. (1954). Behavior and development as a function
of the total situation. In L. Carmichael (Ed.), Manual
of child psychology (2nd ed.). New York: Wiley.
Marcia, J. E. (1980). Identity in adolescence. In J. Adelson
(Ed.), Handbook of adolescent psychology (pp. 159–
187). New York: Wiley.
Mascalo, M. F., & Fischer, K. W. (2015). The dynamic
development of thinking, feeling, and acting:
Infancy through adulthood In W. F. Overton & P. C.
M. Molenaar (Eds.), Handbook of child psychology and developmental science (7th ed.), Volume 1:
Theory and method. Editor-in-chief: R. M. Lerner.
Hoboken: Wiley.
R.M. Lerner et al.
McClelland, M. M., Geldhof, G. J., Cameron, C. E.,
& Wanless, S. B. (2015). Development and selfregulation. In W. F. Overton & P. C. Molenaar (Eds.),
Theory and method. Volume 1 of the Handbook of
child psychology and developmental science (7th ed.).
Editor-in-chief: R. M. Lerner. Hoboken: Wiley.
McCrae, R. R., Costa, P. T., Hrebickova, M., Ostendord,
F., Angleitner, A., & Avia, M. D. (2000). Nature
over nurture: Temperament, personality, and life
span development. Journal of Personality and Social
Psychology, 78(1), 173–186.
Meaney, M. (2014, October 10). Epigenetics offer hope
for disadvantaged children. [Children and family:
Blog]. Retrieved from http://childandfamilyblog.com/
epigenetics-offer-hope-disadvantaged-children/
Misteli, T. (2013). The cell biology of genomes: Bringing
the double helix to life. Cell, 152(6), 1209–1212.
Mistry, J., & Wu, J. (2010). Navigating cultural worlds
and negotiating identities: A conceptual model.
Human Development, 53(1), 5–25.
Molenaar, P. C. (2014). Dynamic models of biological pattern formation have surprising implications for understanding the epigenetics of development. Research in
Human Development, 11(1), 50–62.
Molenaar, P. C., & Nesselroade, J. R. (2014). New trends
in the inductive use of relation developmental systems
theory: Ergodicity, nonstationarity, and heterogeneity. In P. C. Molenaar, R. M. Lerner, & K. M. Newell
(Eds.), Handbook of developmental systems and methodology (pp. 442–462). New York: Guilford Press.
Molenaar, P. C. M., & Nesselroade, J. R. (2015). Systems
methods for developmental research. In W. F. Overton
& P. C. M. Molenaar (Eds.), Handbook of child psychology and developmental science (7th ed.), Volume
1: Theory and method. Editor-in-chief: R. M. Lerner.
Hoboken: Wiley.
Molenaar, P. C., Lerner, R. M., & Newell, K. M. (2014).
Developmental systems theory and methodology: A
review of the issues. In P. C. Molenaar, R. M. Lerner,
& K. M. Newell (Eds.), Handbook of developmental systems and methodology (pp. 3–18). New York:
Guilford Press.
Narvaez, D. (2008). Human flourishing and moral development: Cognitive and neurobiological perspectives
of virtue development. In L. Nucci & D. Narvaez
(Eds.), Handbook of moral and character education
(pp. 310–327). Oxford: Routledge.
Nesselroade, J. R. (1988). Some implications of the traitstate distinction for the study of development over the
life-span: The case of personality. In P. B. Baltes, D. L.
Featherman, & R. M. Lerner (Eds.), Life-span development and behavior (Vol. 8, pp. 163–189). Hillsdale:
Erlbaum.
Nesselroade, J. R., & Molenaar, P. C. M. (2010).
Emphasizing intraindividual variability in the study
of development over the life span. In W. F. Overton
(Ed.), The handbook of life-span development. Vol. 1:
Cognition, biology, methods (pp. 30–54). Editor-inchief: R. M. Lerner. Hoboken: Wiley.
Overton, W. F. (1973). On the assumptive base of the
nature-nurture controversy: Additive versus interactive
conceptions. Human Development, 16, 74–89.
Adolescent Health Development: A Relational Developmental Systems Perspective
Overton, W. F. (2010). Life-span development: Concepts
and issues. In W. F. Overton (Ed), Cognition, biology,
and methods across the lifespan. Handbook of life-span
development. (Vol. 1, pp. 1–29). Editor-in-chief: R. M.
Lerner. Hoboken: Wiley.
Overton, W. F. (2013). Relationism and relational developmental systems; A paradigm for developmental science
in the post-Cartesian era. In R. M. Lerner and J. B.
Benson (Eds.), Advances in child development and
behavior, volume 44: Embodiment and epigenesis:
Theoretical and methodological issues in understanding
the role of biology within the relational developmental system, Part A: Philosophical, theoretical, and
biological dimensions (pp. 24–64). London: Elsevier.
Development of Character 49.
Overton, W. F. (2015). Process and relational developmental systems. In W. F. Overton & P. C. Molenaar (Eds.),
Theory and method. Volume 1 of the Handbook of
child psychology and developmental science (7th ed.,
pp. 9–62). Editor-in-chief: R. M. Lerner. Hoboken: Wiley.
Overton, W. F., & Lerner, R. M. (2014). Fundamental concepts and methods in developmental science: A relational perspective. Research in Human Development,
11(1), 63–73.
Overton, W. F., & Mueller, U. (2013). Meta-theories,
theories, and concepts in the study of development.
In R. M. Lerner, M A. Easterbrooks, & J. Mistry
(Eds.), Comprehensive handbook of psychology:
developmental psychology: Vol. 6. (pp. 19–58). Editorin-Chief: Irving B. Weiner. New York: Wiley.
Overton, W. F., & Reese, H. W. (1981). Conceptual prerequisites for an understanding of stability–change
and continuity–discontinuity. International Journal of
Behavioral Development, 4, 99–123.
Paus, T. (2009). Brain development. In R. M. Lerner &
L. Steinberg (Eds.), Handbook of adolescent psychology (3rd ed., pp. 95–115).
Ricco, R., & Overton, W. F. (2011). Dual systems competence ß-à procedural processing: A relational developmental systems approach to reasoning. Developmental
Review, 31, 119–150. http://dx.doi.org/10.1016/j.
dr.2011.07.005.
Riegel, K. F. (1975). Toward a dialectical theory of human
development. Human Development, 18, 50–64.
Riegel, K. F. (1976). The dialectics of human development. American Psychologist, 31, 689–700.
Roberts, B. W., Walton, K. E., & Viechtbauer, W. (2006).
Patterns of mean-level change in personality traits
across the life course: A meta-analysis of longitudinal
studies. Psychological Bulletin, 132(1), 1–25.
121
Roth, J. L., & Brooks-Gunn, J. (2003a). What is a youth
development program? Identification and defining
principles. In F. Jacobs, D. Wertlieb, & R. M. Lerner
(Eds.), Enhancing the life chances of youth and families:
Public service systems and public policy perspectives:
Vol. 2 handbook of applied developmental science:
Promoting positive child, adolescent, and family
development through research, policies, and programs
(pp. 197–223). Thousand Oaks: Sage.
Roth, J. L., & Brooks-Gunn, J. (2003b). What exactly is a
youth development program? Answers from research and
practice. Applied Developmental Science, 7, 94–111.
Slavich, G. M., & Cole, S. W. (2013). The emerging field
of human social genomics. Clinical Psychological
Science, 1, 331–348.
Sokol, B. W., Hammond, S., Kuebli, J., & Sweetman, L.
(2015). The development of agency. In W. F. Overton
& P. C. Molenaar (Eds.,) Theory and method. Volume
1 of development of the Handbook of child psychology
and developmental science (7th ed.). Editor-in-chief:
R. M. Lerner. Hoboken: Wiley.
Steinberg, L. (2010). A dual systems models of adolescent risk-taking. Developmental Psychobiology, 52,
216–224.
Susman, E., & Dorn, L. D. (2009). Puberty: Its role in
development. In R. M. Lerner & L. Steinberg (Eds.),
Handbook of adolescent psychology (3rd ed., pp. 116–
151). Hoboken: Wiley.
Susman, E. J., & Dorn, L. D. (2013). Puberty: Its role in
development. In R. M. Lerner, M. A., Easterbrooks,
& J. Mistry (Eds.), Handbook of psychology, Volume
6: Developmental psychology (2nd ed., pp. 289–320).
Editor-inchief: I. B. Weiner. Hoboken: Wiley.
Tobach, E., & Schneirla, T. C. (1968). The biopsychology
of social behavior of animals. In R. E. Cooke & S. Levin
(Eds.), Biologic basis of pediatric practice (pp. 68–82).
New York: McGraw-Hill.
Trommsdorff, G. (2012). Development of “agentic”
regulation in cultural context: The role of self and world
views. Child Development Perspectives, 6(1), 19–26.
von Bertalanffy, L. (1933). Modern theories of development. London: Oxford University Press.
von Bertalanffy, L. (1968). General systems theory.
New York: Braziller.
Whitehead, A. N. (1929/1978). Process and reality.
Corrected edn. New York: The Free
Witherington, D. C. (2014). Self-organization and explanatory pluralism: Avoiding the snares of reductionism in developmental science. Research In Human
Development, 11, 22–36.
Open Access This chapter is licensed under the terms of the Creative Commons Attribution 4.0 International License
(http://creativecommons.org/licenses/by/4.0/), which permits use, sharing, adaptation, distribution and reproduction in
any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license and indicate if changes were made.
The images or other third party material in this chapter are included in the chapter’s Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the chapter’s Creative Commons license
and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain
permission directly from the copyright holder.
Emerging Adulthood as a Critical
Stage in the Life Course
David Wood, Tara Crapnell, Lynette Lau,
Ashley Bennett, Debra Lotstein, Maria Ferris,
and Alice Kuo
1
Introduction to Emerging
Adulthood
The path that individuals take from dependency in
childhood to independence in adulthood is now a
longer and more complicated one than at any
other point in history (Arnett 2014). From the
mid- to late twentieth century and extending into
the twenty-first century, industrial societies have
experienced a surge in the concept of individualism and increased salience of self-realization and
personal expression (Arnett 1998). Moreover, we
have seen a dramatic increase in the number of
youth seeking post-high school education, which
is required for success as the economy transitions
from an industrial to an information-based econThe original version of this chapter was revised.
An erratum to this chapter can be found at
https://doi.org/10.1007/978-3-319-47143-3_27
D. Wood, MD, MPH (*)
Department of Pediatrics, ETSU College
of Medicine, PO Box 70578, Johnson City,
TN 37614, USA
e-mail: wooddl@etsu.edu
T. Crapnell, OTD, OTR/L • L. Lau, PhD
D. Lotstein, MD, MPH • M. Ferris, MD, PhD, MPH
A. Kuo, MD, PhD
UCLA, Department of Pediatrics, Los Angeles,
CA, USA
A. Bennett, MD
Department of Pediatrics, USC, Los Angeles, CA, USA
omy (Rifkin 2011). However, stagnation of wages
for low-skilled workers and the lack of work
opportunities for youth and young adults, combined with the increased costs of education and
independent living, have made the pathway to
independence and adulthood prolonged, complex,
and varied, creating a new stage in the life course
that has been labeled emerging adulthood (EA).
While not all life course or developmental scientists agree that emerging adulthood constitutes a
new developmental stage, there is agreement that
social and economic forces have prolonged entry
into adulthood and with significant role and developmental challenges beyond the traditional adolescent years (Cote 2014).
At the beginning of this stage, 17–18 years of
age, emerging adults are generally dependent, living with their parents or caretakers, beginning to
engage in romantic relationships, and attending
high school. At the end of this stage, mid- to late
20s, most emerging adults live independently, are
in long-term relationships, and have clear career
paths ahead of them. How they traverse this life
stage is dependent upon the personal, family, and
social resources they possess as they enter this
stage of life, dynamic and reciprocal interaction
between the emerging adult and their environment,
and the supports they receive during this stage. The
result is that there are many pathways that youth
and young adults pursue through this stage to
achieve stable adulthood. For example, 40% do not
© The Author(s) 2018
N. Halfon et al. (eds.), Handbook of Life Course Health Development,
DOI 10.1007/978-3-319-47143-3_7
123
D. Wood et al.
124
pursue post-high school education. While 60%
entered college immediately after graduating high
school, many drop out or interrupt their college
education with periods of work. Some 33% in this
stage remain unmarried; however, 67% of them
achieve stable domestic partners. Importantly, only
a minority of emerging adults are employed in fulltime jobs, limiting the economic opportunities they
experience (U.S. Census Bureau. American
Community Survey 2006).
Emerging adulthood is considered to be the
volitional years, as it offers the most opportunity
for identity exploration in the areas of love, work,
and worldviews (Arnett 2000). During this time,
individuals begin to develop the characteristic
qualities necessary for becoming self-sufficient,
engage in mature, committed relationships, assume
more adult roles and responsibilities, and obtain a
level of education and training that sets the foundation for work during the adult years. Characterizing
emerging adulthood as a stage in the life course has
proven to be beneficial to explain the social, cognitive, and psychological development that occurs
during this stage. Research demonstrating continued brain development into the late 20s provides
further justification for viewing emerging adulthood as a stage in the life course (Spear 2000).
2
Conceptual Framework
The Life Course Health Development
(LCHD) model posits that myriad factors (e.g.,
biological, psychological, cultural) on multiple
levels (e.g., micro, meso, macro) interact simultaneously in a transactional fashion to influence
an individual’s LCHD during each stage to
determine a “health developmental” trajectory
(see also Halfon and Forrest 2017). The Life
Course Health Development (LCHD) model further posits that transitions and pivotal points in
an individual’s life have the potential to influence and alter an individual’s developmental
pathways. EA is a life stage characterized by
changes in person-context cognitive, emotional,
physical, and social domains, and the ultimate
pathway achieved by the emerging adults during
this stage is determined by the ongoing, dynamic,
and reciprocal interactions between the individ-
ual and their environment. The degree of agency
and role exploration that characterizes EA results
in the potential for growth in intellectual and
emotional functioning (Arnett 2000). EA represents a broad and diverse but fundamentally
important area of consideration by virtue of the
multiple avenues through which an individual’s
developmental trajectories and outcomes may be
influenced. Important developmental challenges
during EA include the continued formation of
identity and values, which occur in the midst of
frequent changes in personal relationships, living arrangements, vocational and educational
pursuits, and social roles (Shanahan 2000).
No stage in life, other than perhaps infancy,
experiences such dynamic and complex changes
on the personal, social, emotional, neuroanatomical, and developmental levels. For the 10 years
between 18 and 28, the vast majority of emerging
adults change living situation, change their primary relationships, complete education or vocational preparation, get married, have children,
and transition from adolescent/dependent roles to
adult/independent roles. This occurs during often
volatile emotional, neurodevelopmental, and
social development. Increasing agency occurs at
the same time as decreasing institutional and
family supports. The theoretical framework
developed by Learner and others to create the
positive youth development theory nicely
explains how the developmental trajectories that
emerge during the period of EA are dependent
upon multiple influential, bidirectional, personcontext coactions. Individuals during emerging
adulthood act as co-developers of their own
developmental pathways, adaptively responding
to different biological, social, cultural, and physical environmental contexts that they influence
and are also influenced by (Learner and Overton
2008). Successfully navigating the developmental challenges inherent in EA will likely, in large
part, influence the developmental trajectory of
adulthood because these challenges ultimately
influence the important adult outcomes of independent living, committed intimate relationships,
and vocational and educational achievement.
Youth and young adults with chronic disease or
disabilities face additional challenges (disease
management, disease complications, limitations
Emerging Adulthood as a Critical Stage in the Life Course
in opportunities, etc.) in the context of these multiple domains of growth and development, which
impact their pathway through this stage.
The continued positive trajectory of the emerging adult’s mental health, identity formation, education achievement, social relationships, and
other developmental areas is somewhat dependent on the degree to which there are matches or
mismatches between the individual and his/her
resources and the environmental challenges and
supports. If the transition, such as school to work,
provides a reasonable and developmentally
appropriate challenge and the emerging adult successfully navigates that challenge, then the developmental trajectory of the emerging adult will be
enhanced. In contrast, if the same transition lacks
supports or is an inappropriate match for the
emerging adult’s abilities (e.g., an inadequate
vocational program for an emerging adult with a
learning disorder or lack of support for chronic
disease management) and the emerging adult
experiences failure, then the developmental trajectory may be impaired, resulting in significantly
less achievement or developmental progression
across the lifespan. A major defining characteristic of the stage of EA is that contexts are changing
significantly (family to independence or romantic
relationships, school to work or disconnected
state, dependent living to independent living
arrangements). The changes are so significant that
emerging adults need substantial supports to navigate the transition successfully. Emerging adults
with disabilities or chronic health conditions
require more support to maximize their potential
development during EA (Table 1).
The above table outlines the seven principles
of the Life Course Health Development framework. These principles can be applied to the
stage of emerging adulthood in a limited fashion
due to the limited research literature focused on
this life stage. However, conceptually, using the
seven principles to view the stage of emerging
adulthood can be instructive and can lead to
additional research questions (see end of this
article). For example, Principle 5 states that
health development expressions are malleable
and enable and constrain health development
pathways and plasticity. According to Bogin’s
reserve capacity hypothesis, prolonged child-
125
Table 1 Principles of the Life Course Health Development
framework
Principle
1. Health
development
2. Unfolding
3. Complexity
4. Timing
5. Plasticity
6. Thriving
7. Harmony
Brief description
Health development integrates the
concepts of health and
developmental processes into a
unified whole
Health development unfolds
continuously over the lifespan,
from conception to death, and is
shaped by prior experiences and
environmental interactions
Health development results from
adaptive, multilevel, and
reciprocal interactions between
individuals and their physical,
natural, and social environments
Health development is sensitive to
the timing and social structuring
of environmental exposures and
experiences
Health development phenotypes
are malleable and enabled and
constrained by evolution to
enhance adaptability to diverse
environments
Optimal health development
promotes survival, enhances
well-being, and protects against
disease
Health development results from
the balanced interactions of
molecular, physiological,
behavioral, cultural, and
evolutionary processes
hood into adolescence and perhaps emerging
adulthood leads to greater biologic and social
resilience in adulthood, which leads to enhanced
fertility and greater longevity (Bogin 2013).
Principle 4 states that health development is sensitive to life course timing and social structuring
of the environment. As we described above, this
is particularly true of emerging adulthood, where
the interaction between age, personal development, and environment (peers, schools, social
institutions, etc.) leads to multiple pathways to
identify formation and academic and social
achievement during this stage (Benson and Elder
2011; Benson et al. 2012).
Multiple factors can influence the life course
during EA, including factors at the macro-level
(historical and societal influences), meso-level
(parent-child relationship, family environment,
D. Wood et al.
126
and socioeconomic status), and microlevel (individual cognitive, personality, and emotional
development). The timing of these exposures
during EA, which we consider a critical or sensitive period in the life course development, can
significantly impact adult outcomes. For example, an emerging adult that engages in criminal
activity and is convicted of a felony will suffer
repercussions that will greatly diminish their
chances of achieving success in relationships and
work. In the sections that follow, we review the
macro-, meso-, and microlevel influences occurring during EA for emerging adults generally.
Next we explore the additional challenges faced
by those with selected chronic health or developmental conditions including mental illness or
substance abuse, diabetes, chronic renal failure,
and autism, to serve as case studies of the
increased complexity faced by emerging adults
with chronic disease. Finally, we finish up with a
set of questions and issues that are research priorities for developing an LCHD research agenda
on the stage of emerging adulthood.
3
Macro-level Influences
on the Trajectory
of Emerging Adulthood
Emerging adulthood, like many other developmental stages, is a period in the life course that is
culturally constructed and not universal (Arnett
1998, 2000). Thus, the very existence and trajectory of EA are dependent on macro-level societal
expectations and influences. For many years, it
was theorized that an individual transitioned
from adolescence (which begins in puberty and
ends in the late teens) into young adulthood. Over
history and cross-culturally, the length of time
during which an individual spends in adolescence
has been determined by the age at which the person enters marriage or a committed relationship
(Schlegel and Barry III 1991, Gilmore 1990). In
fact, human life history posits that the life stage
of adolescence came into existence about
75,000 years ago due to changes in human societies and cultures associated with pair bonding and
living in large groups (Bogin 2013). So massive
culture and reproductive change led to a change
in life history with the addition of a new period of
development. It is theorized by those promoting
emerging adulthood as a new life stage that the
same kind of massive cultural and reproductive
change is happening once again and new social
and cultural demands of adult life necessitate a
longer and more complex prologue to adult life.
Although the functional outcome of EA, causing a delayed transition to adulthood, appears
relatively novel, this practice of prolonging the
transition to adulthood may be dated back to
early modern English society from the 1500s to
1700s (Ben-Amos 1994). Similar to what generally occurs during EA today, individuals during
that era took part in a “life cycle service” between
their late teens and 20s, wherein they would
engage in developing important vocational skills
(e.g., a trade or craft) and marriage was often
postponed until the late 20s for both men and
women; adult roles and responsibilities for individuals during that era were provided only gradually, as each individual acquired the desired
character qualities deemed appropriate by society
(Ben-Amos 1994). It was not until industrialization in America began to emerge in the nineteenth
century that the concept of individualism developed and strengthened (Rotundo 1994). The
twentieth century marked the first time that an
individual could obtain and gain control over the
resources that would allow them to choose the
timing of major life events and personal expression was valued in society (Modell 1991).
As we move forward into the twenty-first
century, individualism in contemporary postindustrial society continues to strengthen, while
the age at which individuals marry continues to
increase, and individuals increasingly seek to
pursue other life course events (Modell 1991;
Alwin 1988; Bellah et al. 2007). As a reflection
of these trends, recent evidence indicates that
contemporary postindustrial society considers
marriage status of low importance in the determination of whether or not an individual has
reached adulthood; rather, societies in developed countries appear to now consider the
acquisition of certain character qualities as the
most important indicator of having attained
Emerging Adulthood as a Critical Stage in the Life Course
adulthood, the top three character qualities of
which include (1) accepting responsibility for
oneself, (2) making independent decisions, and
(3) assuming financial independence – all of
which emphasize an individual’s ability to be
self-sufficient (Scheer et al. 1996; Greene et al.
1992; Arnett 1997, 1998).
4
Meso-level Influences of Life
Trajectories
During Emerging Adulthood
Earlier life experiences and family environment
can impact one’s life trajectory from childhood
through EA and into adulthood. Longitudinal
studies that span early childhood through EA
indicate that there is both continuity and discontinuity of healthy and unhealthy paths and outcomes. (Masten et al. 2006, 2005). In this section,
we will be discussing the impact of the parentchild relationship, family environment, and other
sociodemographic factors, such as socioeconomic status (SES) and its influence in EA.
4.1
Earlier Parent-Child
Relationships
The quality of parent-child relationships during
EA is, in large part, a function of the history of
early parent-child attachment experiences.
Attachment theory suggests that positive bonding
with an emotionally available adult during early
childhood facilitates the development of the
child’s capacity to bond with others and respond
to stressful events. A history of positive attachment experiences will ultimately provide a foundation for positive interactions with others and
foster secure, lasting relationships in EA which
subsequently influence the LCHD (Mikulincer M
and Shaver PR 2009), whereas insecure attachments can result in mistrust or lack of security
and reduced sense of self-efficacy and selfesteem. Simultaneously, social learning theory
suggests that styles of family interactions learned
in early and middle childhood are carried by
emerging adults into adulthood (Whitbeck et al.
127
1994). Familiar patterns of interaction help individuals deal with new situations and have a significant influence on the emerging adult’s
capacity to deal with the multiple changes that
occur during EA. Alternatively, inadequate parenting, disrupted family bonds, and poverty are
environmental risks for childhood-onset conduct,
behavioral and emotional problems, and educational underachievement that can persist into
adolescence and emerging adulthood (Moffit and
Caspi 2001).
Adverse events experienced in childhood such
as parental divorce or alcoholism or the experience of abuse are major risk factors for the leading causes of illness and death as well as poor
quality of life in adults in the USA. Disruptions in
the parent relationship (e.g., divorce) can have
strong, negative effects on the parent-child relationship (Aquilino 1994) causing significant emotional impact throughout the life course. Parental
divorce may influence the emerging adults’ ability
to form stable romantic relationships and their
decision to marry early or may undermine the
emerging adult’s financial ability to attend college
(Jacquet and Surra 2001, Cherlin et al. 1998).
Importantly, parental acceptance and support
for independence have been linked to higher
self-esteem, individualism, and feelings of worthiness among emerging adults (Ryan and Lynch
1989). Critical to the emerging adult’s achievement of independence are feeling connected,
secure, understood, and loved in their families
and having the willingness to call on parental
resources for help in making choices in educational,
relationship, living, and other changes that confront the emerging adult. Persistent connectedness to parents facilitates rather than undermines
ongoing identity development in emerging
adulthood (Grotevant and Cooper 1986; Ryan
and Lynch 1989).
4.2
Childhood Socioeconomic
Status
Socioeconomic status (SES), family supports, and
the neighborhood environment all can contribute
to the positive or negative life trajectory prior to
D. Wood et al.
128
and during EA (Galobardes et al. 2008; Gilman
2012; Gilman and Loucks 2012). Socioeconomic
status has been identified as one of the most
important health determinants throughout the life
course (Miller et al. 2015). Low financial
resources and all the other exposures that go along
with poverty – poor schools, chaotic families,
exposure to violence, and victimization – limit
opportunities for growth or acquisition of other
resources that enhance quality of life (Wadsworth
et al. 2016; Kim et al. 2015).
In addition to being an important predictor of
disease-specific morbidity and mortality in adulthood, early childhood poverty has been associated with lower adult educational attainment
(Duncan and Brooks-Gunn 2000). Educational
achievement has a major influence on the life trajectory, including financial stability and health in
EA and onward throughout adulthood. Seventy
percent of emerging adults who grow up in poverty delay enrollment in postsecondary education, in comparison to 40% who grow up in
household of higher SES. Furthermore, research
has indicated that individuals who delay
enrollment past the age of 22 are less likely to
ever enroll in postsecondary education and less
likely to complete a degree (Feliciano and
Ashtiani 2012). The result is that only 22% of
young adults from low-income families earn college degrees, while 48% of young adults from
higher-income families earn at least an associate’s degree. Earning a college education results
in not only higher lifetime earnings but enhancement of multiple aspects of psychosocial development (Evans and Cassells 2014).
However, while SES has been found to be predictive of educational attainment, the impact of
poverty can be mitigated by a number of factors
such as individual characteristics of self-efficacy
and hope. Students from poor families that have
high self-efficacy and a similar concept of hope
for educational attainment do equally as well in
school as their higher SES peers (Osgood et al.
2005). Recent research by Dweck and colleagues
indicates that students’ mindsets are critically
linked to resilience and achievement and that
they can be changed through brief interventions,
leading to substantial increases in measures of
resilience and achievement (Yeager and Dweck
2012). These studies suggest that in addition to
SES, individual characteristics also have a large
influence on outcomes of EA and that these characteristics can be influenced through individual
interventions. Individual characteristics and
development are discussed in the next section.
5
Microlevel Influences
on the Trajectories
During Emerging Adulthood
As discussed in previous sections, individualism
and the qualities of character have become
increasingly important in the transition to EA and
ultimately reaching full adulthood in postindustrial, developed societies. Studies have shown
that the ability to accept responsibility for one’s
self, make independent decisions, and assume
financial independence are the top three most
important factors in becoming an adult (Arnett
2014). In order to achieve individualism and
obtain these important qualities of character, one
needs to reach some degree of cognitive and psychological maturity, as well as possess some level
of resilience. These individual factors are discussed in this section.
5.1
Cognitive Development
One of the identified qualities of character that is
important in EA and reaching full adulthood is
independent decision-making. While the ability
to make independent decisions is a reflection of
one’s psychological and moral identity, it also
has to do with cognitive maturity and the ability
to weigh a variety of considerations before deciding on a choice. Recent neuroscience research
indicates that brain development (particularly in
the prefrontal cortex) continues well into the
third decade of life, ultimately resulting in the
integration and coordination of cognitions, emotion and action, and strategic executive control
(Luciana et al. 2005). The continuous unfolding
and acquisition of specific neurodevelopmental
capacities during adolescences and EA influence
Emerging Adulthood as a Critical Stage in the Life Course
the acquisition of goal directedness and future
orientation that have been observed behaviorally
during EA (Dahl 2004; Nelson et al. 2012; Nurmi
1999; Steinberg et al. 2006). As new capacities
emerge, they are available to solve problems,
delay gratification, and filter unnecessary input.
With maturation of these skills, emerging adults
are also more capable of reflecting on the influence of their environment and on their internal
state, regulate their emotions, and use problemsolving skills to effectively compromise, which is
important for the development of meaningful
social interactions and personal relationships as
well as in the work environment. These skills also
support an emerging adult’s capacity for optimally interacting with the health-care system,
managing their health-care needs, and making
decisions that will influence their long-term
health outcomes.
However, the preceding paragraph assumes
that the individual has experienced optimal neurodevelopment up to the point that they enter the
stage of EA. Studies have demonstrated that
exposure to chronic stress during childhood
(e.g., poverty) or experiencing adverse childhood events (e.g., child maltreatment, neglect,
parental divorce, parental substance abuse) may
cause detrimental impact to the developing
brain. For example, repeated exposure to stressful events has been associated with structural
differences in specific brain regions (i.e., amygdala, hippocampus, and prefrontal cortex), which
are in turn associated with functional differences
in learning, memory, and aspects of executive
functioning. Furthermore, preliminary studies
go on to suggest that there may be sensitive periods of brain development with increased susceptibility to the effects of stress and adverse events.
These environmentally induced modulations in
neurodevelopment can impact an individual’s
cognitive development and their capacity to
develop the necessary skills and relationships
that will enable them to thrive.
An emerging adult, who has experienced
chronic stress or adverse events earlier in childhood and adolescence and is cognitively immature, is more likely, through adverse interaction
with their environment, to suffer secondary
129
effects such as school failure, risky/impulsive
behaviors, accidental injury, criminal activity, or
substance misuse or overuse. These behaviors
may in turn interfere with the ongoing development of an optimal pathway to adulthood.
Feelings of isolation and rates of substance abuse
in individuals often peak during this period as
emerging adults are faced with the stresses of
having to navigate societal structures that are not
adequately informed or equipped to address the
needs of an emerging adult population – all while
their brains have yet to reach full maturation
(Spear 2000). An impulsive emerging adult
fueled by alcohol and despair is at high risk for
suicide. It is thus apparent that multiple factors
related to an individual’s cognitive and emotional
development may intersect and interact to multiply the likelihood of adverse or positive health
outcomes in EA. It is crucial for service providers
to recognize the level of cognitive maturation an
emerging adult possesses and tailor their interventions and supports based on this.
5.2
Identity Formation
Identity formation is a major developmental
activity during EA. Identity development, occurs
in a number of dimensions: (1) psychological
dimension, or ego identity via a sense of temporalspatial continuity and its concomitants; (2) the
personal dimension, or a behavioral and character repertoire that differentiates the individuals;
and (3) the social dimension, or recognized roles
within a community. These components come
together during the stage of identity formation
(adolescence and EA) and stabilization (EA and
young adulthood), and once the identity is considered stable, this is when a relatively firm sense
of ego identity is developed, behavior and character become stabilized, and communitysanctioned roles are acquired (2002).
Identity formation during adolescence was
thought to be a critical milestone in adolescence;
however, it has been recognized that in certain
societal contexts, identity formation continues
beyond adolescence. In the postindustrial society, with the prolongation of educational and
D. Wood et al.
130
vocational attainment, prolongation of identity
exploration in the three main areas of love, work,
and worldviews into the 20s has become the
norm. Identity formation is critical in EA, as it
has direct implications on psychological and
moral identity and in achieving the three qualities of character deemed as important in the transition to adulthood.
5.3
Resilience in Emerging
Adulthood
Resilience is an individual’s capacity to adapt to
change in healthy and flexible ways during stressful events (Catalano et al. 2004). Resilience can
be measured in various ways, including internal
adaption (e.g., well-being, happiness, or selfconcept) or external adaption (academic achievement, relationship development) to adverse
circumstances, such as how well a person navigates and achieves the developmental tasks presented by the external world (e.g., educational
achievement, stable work/career, marriage, etc.).
Resiliency in adolescence and young adulthood
is higher among youth with higher intellectual
resources, optimistic future orientation, the presence of caring relationships with positive adult
role models, and opportunities to succeed and
serve the community. Emerging adulthood is a
particularly important stage in the life course to
understand resilience because the important
changes in functional capacity, educational
achievement, and social roles are large and varied
and have a significant influence on life course
outcomes.
Key individual characteristics that predict
resilience during EA are goal-directed motivation
and planfulness about the future. In addition,
adult support and mentorship are important factors that help promote resilience (Miller GE 2015;
Masten et al. 2005; Arnett 2005). Furstenberg,
Brooks-Gunn, and others studied young women
who became pregnant during adolescence and
found that a small proportion with positive adult
relationships had dramatic change in trajectory
for the better (Furstenberg 2002). Masten, study-
ing a cohort of low-income youth (beginning at
ages 8–12) over a 20-year period, found that a
number of factors predict competence in EA
including higher intellectual capacity, higher parenting quality, and higher SES (Masten and
Coatsworth 1998). When they examined resilience between the 10- and 20-year follow-up, they
found that youth doing well in EA was predicted
from doing well 10 years earlier. In the context of
high adversity, childhood IQ was particularly an
important moderator and predictor of resilience.
During EA, individuals who were doing well
from high-adversity backgrounds had significant
personal resources in intelligence and personal
competence and a history of receiving high-quality parenting (Masten et al. 2004).
6
Trajectories
During Emerging Adulthood
for Emerging Adults
with Chronic Health
Condition
Chronic health conditions can significantly
impact the developmental trajectory of emerging adults during this life stage. Many types of
chronic conditions, including those that impact
physical, intellectual, or emotional functioning,
may undermine the assumption of adult roles by
the emerging adult, undermine success in
school, impair the transition to work, and make
living independently more of a challenge.
Eighteen percent of youth aged 12–17 in the
USA are defined as having a special health-care
need, meaning they have a chronic physical,
medical, emotional, or developmental condition
that requires a more intensive use of health-care
and related services. Emerging adults with
chronic conditions are at risk for a number of
problems during transition to adulthood, including experiencing gaps in needed medical and
related services and gaps in health insurance
(Lotstein et al. 2008, Reiss et al. 2005). Below
we review the impact of several specific chronic
conditions on the growth trajectory during
emerging adulthood.
Emerging Adulthood as a Critical Stage in the Life Course
6.1
Autism Spectrum Disorders
(ASDs)
For youth with autism spectrum disorders (ASDs)
and their families, the transition from adolescence to emerging adulthood is a time of uncertainty and loss of entitlement to many services
that were available while in the public school system under the Individuals with Disabilities
Education Act (IDEA). In comparison to childhood, young adults with autism often experience
a dearth of appropriate supports and opportunities. Leaving high school is associated with deterioration in ASD symptom presentation, increase
in maladaptive behaviors, and worsening family
functioning (Taylor and Seltzer 2010, 2011a, b).
Many persons with ASD are able to work successfully within the community (GarciaVillamisar et al. 2000; Mawhood and Howlin
1999); however, the majority experience difficulty
securing meaningful employment (Eaves and Ho
2008; Howlin et al. 2004; Newman et al. 2011).
A recent study indicated that only 58% have ever
worked for pay. In addition, it was found that
only one in five individuals ever lived independently (Roux et al. 2015). Even among those
employed, their jobs tended to be low level and
low income (Cimera and Cowan 2009; Eaves and
Ho 2008; Howlin et al. 2004). Jobs often ended
prematurely because of social or behavioral difficulties or other work-related difficulties
(Mawhood and Howlin 1999).
Young adults on the autism spectrum are
reported to experience difficulties in assuming
other important adult roles and responsibilities,
including attaining postsecondary education and
living independently (Roux et al. 2015). A number of studies (Billstedt et al. 2005; GillespieLynch et al. 2012; Howlin et al. 2004) have found
that having an IQ of around 70 seemed to be a
critical cutoff point for better outcome (e.g., level
of independence). A number of comorbid conditions can complicate the trajectory for youth with
intellectual disability through adolescence and
EA, such as seizure disorder, which substantially
increase in prevalence from childhood through
adolescence into adulthood. Adults with ASD
and related conditions are more vulnerable to
131
anxiety and depression which can require specific
treatments (Skokauskas and Gallagher 2010;
White et al. 2009). The majority of young adults
with ASD remain dependent on parents for support in living, recreation, and occupational situations. The availability of appropriate resources,
services, and supports for the individuals with
ASD and their families is key for successful transition into adulthood and better outcomes
throughout adult life. The following elements are
important for high-quality transition services and
associated with improved young adult outcomes:
(1) individualized services that reflect the
strengths of the individual, (2) positive career
development and early work experiences, (3) collaboration and interagency involvement, (4) family supports and expectations, (5) fostering
self-determination and independence, (6) social
and employment-related skill instruction, and (7)
establishment of job-related supports. Therefore,
outcomes for emerging adults with ASD could be
improved if social and institutional supports
available through adolescence continued into
adulthood.
6.2
Type 1 Diabetes Mellitus (DM)
The study of emerging adults with type 1 DM has
elucidated the potential challenges facing emerging adults with diabetes and other chronic health
conditions that have significant self-management
burden. Emerging adults with type 1 diabetes
face extensive behavioral demands in order to
maintain their health, including the necessity for
precisely scheduled daily insulin injections,
blood glucose monitoring, dietary monitoring,
regular physical exercise, and the management of
DM-related complications. In early adulthood,
frequent changes in roles, living situations, educational routines, jobs, friendships, and romantic
relationships are common and can undermine the
routines and the resolve needed to maintain metabolic control (Anderson and Wolpert 2004).
The developmental tasks of emerging adulthood may be at odds with maintaining the intensive self-care that is required of those with type 1
diabetes and vice versa. For example, the tasks of
D. Wood et al.
132
establishing autonomy and prevailing egocentrism may be associated with the emerging adult
not wanting to follow medical advice or advice of
his or her family (Masten et al. 1995). Heightened
concern for peer acceptance or the establishment
of intimate relationships is characteristic of this
period of development. Emerging adults may be
reluctant to admit to their significant others that
they have type 1 diabetes or any other chronic
condition that puts them at risk for being rejected
(Madsen et al. 2002). Moreover, the major cognitive developmental milestone during the stage of
emerging adulthood is the ability to think
abstractly (Erikson 1994). Those who have not
yet reached this developmental milestone may
struggle with taking responsibility to maintain
good disease management and understanding the
consequences of poor disease management.
Lastly, increased rates of drinking, illicit drug
use, or other high-risk behaviors may disrupt the
emerging adults’ lifestyle and negatively impact
self-care management of diabetes, resulting in
(Madsen et al. 2002) lack of control of diabetes
and the resulting physical and mental consequences of hypo- or hyperglycemia, which
undermine emerging adults’ abilities to perform
in school, maintain relationships, or keep
employment.
As has been demonstrated in other chronic
childhood conditions, the transition from pediatric- to adult-centered medical care for youth with
DM is associated with a decline in DM control
(Busse et al. 2007; Oeffinger et al. 2006). Adverse
health-related outcomes documented in the
young adult population with diabetes include a
decline in disease self-care behaviors and an
increased risk for diabetes-related complications
(Bryden et al. 2001; Fredericks et al. 2010; Yeung
et al. 2008). In addition, emerging adults with
diabetes are at higher than average risk for psychosocial morbidity including social delays and
isolation, impaired social competence, and emotional problems such as depression (Helgeson
et al. 2007). As in the case in ASD, additional
support for an emerging adult with DM during
the transition to adult medical care has been
shown to help decrease the adverse outcomes
seen with usual practice. For example, DM tran-
sition programs that include introductions to new
adult care providers or intensive care coordination have been found to be protective (HolmesWalker et al. 2007).
6.3
Chronic Kidney Disease
Chronic kidney disease (CKD) is a condition
characterized by disease progression, significant
cardiovascular morbidity, growth failure, neurocognitive impairment, and impaired quality of
life changes (Sarnak 2003; Copelovitch et al.
2011; Gerson et al. 2006; McKenna et al. 2006).
CKD is divided into five stages based on glomerular filtration rate, with stage 1 being the mildest
form and stage 5 the worst form, also called endstage kidney disease (ESKD ), requiring renal
replacement therapy (dialysis or transplant)
(Hogg et al. 2003). The prevalence of pediatric
CKD is unknown as it may be silent in early
stages. The prevalence of pediatric ESKD is 15
cases per million population (Saran et al. 2015).
African-Americans and Latinos are disproportionately affected by CKD in part due to a higher
incidence of glomerular conditions (Ferris et al.
2006). While the 10-year survival for adolescentonset ESKD is 80–85% (much higher than adultonset ESKD patients), this still represents a
30-fold increase in mortality compared to the
general US adolescent population. Survival is
better for younger adolescents, males, Caucasians,
Asians, and transplant recipients (2006).
However, emerging adults with CKD achieve
adult milestones (e.g., employment, marriage)
less frequently than their healthy peers (Bartosh
et al. 2003).
The burden of care directly correlates with the
stage of CKD. Based on the mean number of
unique medications, those with CKD stages 1–4
take 6.2 ± 4 of medications, those with peritoneal
dialysis (CKD 5) take 8.5 ± 2 medications, those
with kidney transplant (CKD 5) take 9.7 ± 4.2
medications, and those on hemodialysis (CKD 5)
take 11.3 ± 2.1 (So et al. 2011). The complexity
of care also include procedures such as selfcatheterization several times per day, fluid and
dietary restrictions, blood pressure measure-
Emerging Adulthood as a Critical Stage in the Life Course
ments daily, and injections (erythropoiesisstimulating agents once to thrice weekly, growth
hormone daily, or insulin several times per day).
The life course of pediatric-onset CKD-ESKD
varies by etiology and age at onset of this condition, but once they reach ESKD, most patients
share the same comorbidities (hypertension, anemia, acidosis, metabolic bone disease, and
growth delay). Patients with greater level of CKD
have a decreased sense of self-worth, perceive a
poor future, and feel limited in their physical and
psychosocial capacities to have the same potential and opportunity as their healthy peers.
While most children and adolescent patients
with ESKD will receive a kidney transplant,
they likely will experience dialysis prior to
receiving an organ. The most common cause of
kidney transplant rejection in adolescents and
emerging adults is treatment nonadherence
(Andreoni et al. 2013). Adherence among adolescents is compromised by poor understanding
and poor consequence recognition leading to an
inconsistent commitment to the treatment regimens. Once the kidney transplant is lost, patients
return to dialysis and likely will not receive
another kidney transplant for many years. As in
adult patients, cardiovascular disease (CVD)
accounts for a majority of deaths in patients
with pediatric-onset CKD, but unlike adults,
pediatric-onset CKD patients rarely demonstrate symptomatic atherosclerosis (Shroff et al.
2011). Lastly, survivors of pediatric-onset CKD
are at greater risk for malignancies and posttransplant diabetes mellitus (Koukourgianni
et al. 2010).
Patients who transfer to adult-focused services without transition preparation and support
appear to be at greater risk to lose their kidney
transplant (Watson 2000, 2005). The international societies of internal medicine and pediatric
nephrology have published position statements
and policies to promote health-care transition
preparation (Watson 2005; Watson et al. 2011).
Strategies to increase patient autonomy, healthcare transition, and self-management are needed
to achieve successful outcomes at the time of
transfer to adult-focused providers.
133
6.4
Mental Health
and Substance Use
Emerging adulthood is a time of increased experimentation with tobacco, drugs, and alcohol
(Schulenberg et al. 2004; Schulenberg and Maggs
2002), increasing from rates of 12.2% in adolescence to 40.2% in young adulthood for cigarette
smoking, from 10.7% to 41.9% for binge drinking, from 11.2% to 20.3% for illicit drug use, and
from 2.6% to 14.9% for heavy alcohol use. While
cigarette, alcohol, and drug use are normative
during adolescence and emerging adulthood, a
number of factors predict excessive use rather
than experimentation including genetic (e.g.,
family history of alcohol abuse), biological (e.g.,
early puberty timing), family (e.g., low parental
monitoring), school (e.g., low grade average,
school failure), peer (e.g., peer substance use),
and youth personality and psychopathology (e.g.,
depression) factors (Cicchetti 1999). However,
the pattern of use/abuse of these substances is
influenced by the experience during the stage of
EA. For example, increasing responsibility,
work, and marriage all are associated with
decreased use of these substances (Masten et al.
1999; Bachman et al. 2014). Again, there is great
heterogeneity in the trajectories followed by individuals with regard to substance use during this
period, all influenced by the multiplicity of factors listed above.
Among young adults, 13.7% experience serious mental illness such as major depressive disorders, schizophrenia, and bipolar disorder
(AmericanPsychiatricAssociation 2013). The
neuropsychological pathology that has onset during EA has many of the predisposing factors
related to family adversities, such as social disadvantage, divorce, dysfunctional parenting, multiple family changes/moves, and peer group
choices. The expanded independence and selfreliance during EA may be difficult to handle for
youth with preexisting emotional or social challenges or poor attachment. The decrease in supports from school or family during EA may be
even further destabilizing, leading to increased
emotional and interpersonal difficulties. Young
D. Wood et al.
134
people with serious mental health conditions have
difficulty with impulse control and self-regulation
(Walker and Gowen 2011). Their apparent immaturity reflects a delay in social and emotional
development and undermines progress toward
goals in education, relationships, independence in
health, and health-care management.
Three-quarters of lifetime cases of mental,
emotional, and behavioral disorders begin by age
24 (Kessler et al. 2005). In emerging adults with
social, emotional, and behavioral disorders,
much higher risk is associated with onset in early
adolescence versus onset in late adolescence
(Mofitt and Caspi 2001). Dysfunctional family
environments and participation in deviant peer
group increase the likelihood of early-onset
delinquency followed by a long trajectory of
social, emotional, and behavioral problems.
Youth with later onset of social, emotional, and
behavioral problems are more commonly from
relatively healthy family backgrounds and more
likely to desist from delinquency by age 23
(Roisman et al. 2004). Similar patterns can be
seen for major psychiatric disorders such as
depression, with early-onset mood disorders
being representative of more severe recurrent
forms of mood disorders arising from increased
severity and complexity of family and biologic
risk factors (Fergusson and Woodward 2002) and
later-onset disorders reflecting lower levels of
risk factors in childhood.
7
Protective and Risk Factors
That Impact Emerging
Adulthood
The supports, opportunities, and experiences that
occur during the stage of EA greatly influence the
ultimate outcome of this stage and the ensuing
life trajectory into young adulthood. As discussed
in this chapter, critical influences during EA
include mentoring relationships, socioeconomic
supports, and educational and vocational opportunities. Emerging adulthood typically is accompanied by an increase in agency/independence
and decrease in social and institutional support
(e.g., schools and family), such that most indi-
viduals have more choice in day-to-day activities
and life decisions. This increase in choice may
result in greater success which will likely contribute to well-being. However, increased freedom can result in poor choices (e.g., drug use,
criminal activity) that have a negative impact on
the developmental trajectory as well (Schulenberg
and Maggs 2002; Schulenberg et al. 2004). For
example, many relationships and other aspects of
life are so unstable during EA that important and
powerful negative experiences can occur such as
date rape, school failure, and substance addiction. These experiences can have a lasting, if not
lifetime, detrimental impact on the developmental and mental health trajectory of the emerging
adult. Similarly, many potential powerful positive influences can occur during this period in life
(e.g., marriage, educational achievement, mentoring) that can enhance the developmental and
mental health trajectory. Therefore, experiences
both distal (early childhood) and proximal (during emerging adulthood) can influence, either
positively or negatively, the developmental and
mental health trajectory of the emerging adult
(Miller et al. 2015).
Practical supports during EA provided by
families include financial support for education,
transportation, independent living, health insurance, and other necessities. The primary determinant of parental support are the level of
parental resources. Emerging adults with wealthier parents and those with fewer siblings receive
more financial support for education (Steelman
and Powell 1991). Divorced parents provide less
financial support than parents with intact marriages (Cooney and Uhlenberg 1992). Support
from families by parents and grandparents during EA is also provided in the form of guidance
or advice, information, and emotional support.
Parents of emerging adults may also help the
emerging adult negotiate the health-care system,
formulate and carry out educational and vocational goals, and acquire leases for apartments
and even provide advice for intimate relationships. In short, continued social and financial
connection to parents with adequate emotional
and financial resources can be a very significant
factor in the positive life trajectory during
Emerging Adulthood as a Critical Stage in the Life Course
emerging adulthood (Aquilino 2005). Previous
studies have found a high correlation between
improved EA outcomes and adolescent hope in
the future (Bennett 2014). Hope is a form of personal capital that protects youth from social,
emotional, and behavioral disorders and correlates with and predicts the rates of many outcomes, including academic achievement, delay
of initiation of early sexual activity, and lower
likelihood of violent behavior. Hope requires a
sense of self-efficacy and knowledge of alternate
outcomes. Hope is reflected in adolescent and
young adult goals as they reflect and make ongoing assessments of their current and future
selves. Individual goals and aspirations relate to
the sense of control an individual feels in relation to a particular domain.
Positive youth development (PYD) programs
are approaches that provide supports to
adolescents and emerging adults to achieve
social, emotional, behavioral, and moral competence, foster self-efficacy, a clear and positive
identity, and belief in the future; and reinforce
pro-social norms (Catalano et al. 2004). Positive
development approaches involve a paradigm shift
from targeting the risk factors to enhancing the
assets and protective factors. The emphasis on
hope, empowerment, and well-being resonates
with youth and mental health professionals. The
PYD approach outlined above suggests that
effective programs to support EA would include
an individualized approach supporting young
people to identify and move toward personally
meaningful goals. This begins with envisioning a
positive future identity (“Who do I want to
become?”). Pursuing goals promotes a sense of
purpose, and making progress toward those goals
contributes to building feelings of efficacy,
empowerment, and self-determination, enhancing the individual’s ability to act as the primary
causal agent in pursuing personally meaningful
goals (Silbereisen and Lerner 2007).
A PYD perspective further suggests that youth
and emerging adults will benefit from learning
specific strategies for increasing and maintaining
interpersonal support from positive peers, family,
providers, and people in the community (Guerra
and Bradshaw 2008). Young people can learn
135
specific steps and skills that can help them
increase the quality and the extent of their interpersonal networks, as well as the amount of emotional, instrumental, and informational support
available to them. Using a PYD perspective suggests that the development of assets is an important recovery-oriented outcome in and of
themselves, as well as a mediator of longer-term
outcomes related to education, employment,
mental health, and general quality of life. Indeed,
a review of the available research on communitybased programs and interventions for EA reveals
a common focus on personal asset building
(Lerner 2014). Also consistent with the PYD perspective is that many of the programs and interventions include a focus on changing the
meso-environment of youth so that it encourages
young people to develop or express strengths and
assets.
8
Services and Supports
As we better understand the limits of and issues
related to EA, it is imperative that we review and
revise policy and current social and medical services and supports so they optimally address the
current and future needs of individuals during
this life stage. Current medical, mental health,
and social service delivery models are geared
toward either child or adult populations. In other
words, individuals generally seek child-serving
services between the ages of 0 and 18, and they
are subsequently transferred directly to adultserving services. However, the various biobehavioral and sociocultural factors discussed above
make this direct transition from child to adultcentered care ill-suited for optimally addressing
the nuances and challenges that individuals may
face during EA. For example, an adult-centered
medical doctor may regularly treat patients with
fixed habits and lifestyles, who may already suffer from a variety of chronic health conditions.
However, many such chronic health conditions in
adults represent a culmination of experiences
from an individual’s earlier years, which may not
yet be apparent during EA. It is thus strongly recommended that EA service providers reorient
136
themselves using a prevention-based paradigm
and familiarize themselves with the possible
antecedents to social, emotional, and medical
conditions so that they may make health-care or
social service recommendations to minimize risk
factors and to maximize strengths and assets to
facilitate development along optimal health trajectories. EA presents a unique opportunity for
service providers to intervene and influence an
individual’s LCHD before significant social,
emotional, and medical pathology fully develops,
utilizing existing knowledge of the skills and
capacities associated with EA to inform services
and interventions.
In addition to training service providers to
reorient their approach to care and become more
sensitive to antecedents of chronic health conditions in adulthood, the multifactorial nature of
the LCHD model suggests that it would be
important for service providers to consider horizontal (i.e., cross sector) and longitudinal (i.e.,
across the lifespan) service integration.
Specifically, the current model posits that myriad
factors (e.g., biological, psychological, cultural)
on multiple levels (e.g., micro, meso, macro)
interact simultaneously in a transactional fashion
to influence an individual’s LCHD and overall
health trajectory. However, most current training
models which emphasize specialization in their
independent fields and opportunities for crossdisciplinary case conceptualization and collaboration with regard to service delivery – particularly
with respect to preventive care, which may be of
special importance when considering EA as a
sensitive period of development – tend to be rare.
Such specialization in service delivery may result
in service fragmentation. Additionally, adult service providers generally do not work in tandem
with child service providers as individuals transition from one care setting to the next, consequently resulting in potential gaps in service
delivery and care, which may be exacerbated as
individuals work to rebuild rapport with their
new service providers. Moreover, health, mental
health, social service, and education professionals rarely collaborate to create comprehensive
models across the EA stage. Clearly, the current
service delivery infrastructure does not ade-
D. Wood et al.
quately support the needs and challenges unique
to emerging adult populations. It is thus important to recognize emerging adults as a unique
population requiring additional and specialized
skills of service providers and the creation of specific pathways for transition which support continuity of care.
The larger sociopolitical and cultural contexts
may also facilitate or create challenges which
impact service and intervention efforts directed at
emerging adults. The majority of individuals in
developed nations transitioning through EA, for
instance, may be classified as “digital natives,”
individuals who have been brought up in environments where exposure to digital technology may
be normative and who are therefore familiar with
computers and the Internet. As emerging adults
increasingly turn to digital and mobile solutions
to support and enhance their daily routines, it
behooves service providers to creatively explore
ways of engaging emerging adult populations
using technology. Use of social networks, such as
Instagram, Twitter, and Facebook, to advance
primary prevention efforts, increase awareness of
public health issues, and provide basic health and
psychological education affords service providers the opportunity to outreach to unprecedented
numbers and populations. Use of text messaging
may also be effective as a way of engaging with
digital native emerging adult populations and
promoting engagement and adherence by emerging adults or to promote youth development programs. Also, as service providers keep pace with
technological advancements, they may even
begin to explore novel methods of service delivery and intervention using digital or Internetbased platforms, which may increase “buy-in”
and potentially preventive service utilization,
among digitally native emerging adult populations. For example, service providers might
potentially leverage the data that digitally native
emerging adults might collect as part of the
“quantified self” movement (e.g., pedometer
information, weight, blood pressure, etc.) in an
effort to inform their ongoing care efforts.
On the other hand, due to clinical billing practices and regulations, as well as patient privacy
concerns and regulations (e.g., Health Insurance
Emerging Adulthood as a Critical Stage in the Life Course
Portability and Accountability Act, HIPAA), current service delivery models often tend to operate
in a fragmented and siloed fashion, which in turn
may limit the ability of service providers to
develop a holistic treatment plan to address each
individual’s needs. Further complicating this
fragmentation is the restricted access to health
records once an individual turns 18 years of age,
when parents who may have been primarily
responsible for managing their child’s healthcare needs no longer are able to access their
child’s medical records (unless explicit permission is granted by the child, who is now an emerging adult). Depending on each family’s cultural
context and the relationship between the emerging adult and his or her parents, this transition
may be a cause for significant stress and/or strife
between family members as they work to renegotiate previously familiar boundaries. Mothers of
children with diabetes, for example, have indicated feeling increased stress as their children
transitioned from pediatric to adult care, particularly if they perceived that their children were not
managing their health-care condition as well as
they had been when it was previously under
greater parent control (Allen et al. 2011). Such
stress might result in familial conflict and dysfunction which could subsequently interfere with
the emerging adult’s willingness or ability to
adequately access services. One potential solution to overcoming the issue of patient privacy
may be to implement public health programs and
tools to educate and empower individuals with
respect to managing and taking an active role in
their own care. However, it is important to underscore the need for larger, overall systems to
change in order to support service providers in
working together with individuals and their families to ensure continuity and comprehensiveness
of services and health-related information during
the transition.
Finally, the implementation of health-care
policies may have important implications for
service and intervention delivery with an emerging adult population. With the introduction of the
Patient Protection and Affordable Care Act of
2010 (ACA), for example, individuals may now
remain on their parents’ health insurance plans
137
until they turn 26 years of age. While this theoretically affords emerging adults the opportunity
to develop the necessary skills for interacting
with the health-care system and managing their
own care, research indicates that emerging adults
frequently perceived themselves as being at
lower risk for health problems and the financial
burden of health insurance as lacking in value.
Consequently, emerging adults may opt for highdeductible catastrophic insurance which generally costs less compared to other health insurance
plans, but translates to fewer opportunities for
interaction with the health service sector and
fewer opportunities for service providers to
intervene early during an individual’s LCHD trajectory prior to the onset of chronic adult disease. Importantly, research indicates that despite
preventive maintenance health visits being mandatory under the ACA, emerging adults were
less likely to request health maintenance visits.
Thus, it may be important for public health
efforts to determine the underlying factors associated with this diminished utilization of health
services by emerging adults and subsequently
develop health literacy programs to promote prevention mindedness and service utilization in
emerging adults.
9
Recommendations
for Research Priorities
The following is a list of issues in life course
research that are critical for better understanding the positive supports and challenges that
influence the trajectory of development of
emerging adults organized along the macro-,
meso-, micro-framework.
Macro Issues
• What community supports in emerging adulthood enhance health development into adulthood and improve outcomes for an emerging
adult with specific health, mental health, and
developmental conditions? Is there an “early
intervention model” to optimize outcomes for
EA? What model should be used for an
D. Wood et al.
138
•
•
•
•
emerging adult with chronic health, mental
health, and developmental conditions?
What are the historical, economic, and cultural influences on the meaning of EA, adaption, and maladaptation to the stage and
expectations for normative transitions? How
have these changed over time? How are they
different for an emerging adult with social,
health, mental health, and developmental
problems?
How have the delays in marriage/relationship
commitment
impacted
adult
life
trajectories?
What are the racial and ethnic differences for
how these factors interact to support development during EA?
How has the information economy and the
increased demands for education impacted the
life course for youth in upper, middle, and
lower socioeconomic strata? Are there
regional variations in these outcomes and how
can they be improved?
•
•
•
•
•
•
•
Meso Issues
• Earlier childhood development and family environment are predictors of adult outcomes, but it
Fig. 1 Moving toward
resilience: a model of
positive change in
emerging adulthood
is unclear how optimizing health development
can be achieved with supports during EA.
What is the interaction between health and
social system supports, family supports,
youth abilities, cognitive and psychological
maturity, and EA outcomes for an emerging
adult with and without mental or physical
disabilities?
How can the different support systems be integrated to provide seamless services to youth
and emerging adults with significant physical,
mental health, or intellectual disabilities?
Using the model below from the work of
Masten et al. (2005), how do these different
resources, supports, and personal characteristics interact to promote optimal development
during EA? (Fig. 1)
What factors promote resilience for youth during EA who lack family supports?
What is the role of extended families in support for EA?
How can studies of conditions and processes
that influence development during EA inform
educational/vocational/EA support policies?
How can data be gathered across this stage in
the lifespan given that emerging adults leave
school and have many changes in living arrangements, education, relationships, and work?
RESILIENCE IN EMERGING ADULTHOOD
Love
Work
Opportunities
Transformational
contexts
School
Good fortune
Mentors
Timing
Adult support
Hope
Capacity
for
Change
Future orientation
Self-direction
Self-efficacy
Motivation
Aspiration
Agency
Strategic
Executive
Control
Planfulness
Emerging Adulthood as a Critical Stage in the Life Course
Micro Issues
• To what degree is cognitive, social, physical,
moral, and spiritual development mediated
during emerging adulthood? How are these
developmental pathways impacted by chronic
health, mental health, and developmental
conditions?
• How can we optimize the person-context
match to promote development during EA
with and without chronic health, mental
health, and developmental conditions?
• What is the impact of positive turning points
in EA compared to early life stages on life trajectories into adulthood?
• During EA, exploration of new experiences is
normative. Is more exploration better or worse
and for which activities is it better or worse?
How is this equation different for an emerging
adult with chronic health, mental health, and
developmental conditions?
• Do emerging adults with particular health
conditions, such as diabetes, spina bifida,
solid organ transplant, or cancer survivorship,
experience sharply downward trajectories during EA, and what supports are effective to prevent these sharply downward trajectories?
• How does brain development progress during
EA? What is the impact of brain development
on the factors that influence the developmental trajectory during EA such as executive
control (planfulness, future orientation), motivation, self-efficacy, and hope?
References
Allen, D., Channon, S., Lowes, L., Atwell, C., & Lane,
C. (2011). Behind the scenes: The changing roles of
parents in the transition from child to adult diabetes
service. Diabetic Medicine, 28(8), 994–1000.
Alwin, D. F. (1988). From obedience to autonomy
changes in traits desired in children, 1924–1978.
Public Opinion Quarterly, 52, 33–52.
American Psychiatric Association. (2013). Diagnostic
and statistical manual of mental disorders. Arlington:
American Psychiatric Publishing.
Anderson, B. J., & Wolpert, H. A. (2004). A developmental perspective on the challenges of diabetes educa-
139
tion and care during the young adult period. Patient
Education and Counseling, 53, 347–352.
Andreoni, K. A., Forbes, R., Andreoni, R. M., Phillips, G.,
Stewart, H., & Ferris, M. (2013). Age-related kidney
transplant outcomes: Health disparities amplified in adolescence. JAMA Internal Medicine, 173, 1524–1532.
Aquilino, W. S. (1994). Impact of childhood family disruption on young Adults’ relationships with parents.
Journal of Marriage and the Family, 56, 295–313.
Aquilino, W. S. (2005). Impact of family structure on
parental attitudes toward the economic support of
adult children over the transition to adulthood. Journal
of Family Issues, 26, 143–167.
Arnett, J. J. (1997). Young People’s conceptions of the
transition to adulthood. Youth & Society, 29, 3–23.
Arnett, J. J. (1998). Learning to stand alone: The contemporary American transition to adulthood in cultural and
historical context. Human Development, 41, 295–315.
Arnett, J. J. (2000). Emerging adulthood: A theory of
development from the late teens through the twenties.
American Psychologist, 55, 469.
Arnett, J. J. (2005). The developmental context of substance use in emerging adulthood. Journal of Drug
Issues, 35, 235–254.
Arnett, J. J. (2014). Emerging adulthood: The winding road from the late teens through the twenties.
New York: Oxford University Press.
Bachman, J. G., O’malley, P. M., Schulenberg, J. E.,
Johnston, L. D., Bryant, A. L., & Merline, A. C.
(2014). The decline of substance use in young adulthood: Changes in social activities, roles, and beliefs.
New York: Psychology Press.
Bartosh, S. M., Leverson, G., Robillard, D., & Sollinger,
H. W. (2003). Long-term outcomes in pediatric renal
transplant recipients who survive into adulthood.
Transplantation, 76, 1195–1200.
Bellah, E. R. N., Bellah, R. N., Tipton, S. M., Sullivan,
W. M., Madsen, R., Swidler, A., Sullivan, W. M.,
& Tipton, S. M. (2007). Habits of the heart:
Individualism and commitment in American life.
Berkeley: University of California Press.
Ben-Amos, I. K. (1994). Adolescence and youth in early
modern England. New Haven: Yale University Press.
Bennett, A. (2014). Finding hope in hopeless environments. International Journal of Child Health And
Human Development, 7, 313.
Benson, J. E., & Elder, G. H. (2011). Young adult identities
and their pathways: A developmental and life course
model. Developmental Psychology, 47, 1646–1657.
Benson, J. E., Johnson, M. K., & Elder, G. H. (2012).
The implications of adult identity for educational and
work attainment in young adulthood. Developmental
Psychology, 48(6), 1752.
Billstedt, E., Gillberg, C., & Gillberg, C. (2005). Autism
after adolescence: Population-based 13-to 22-year follow-up study of 120 individuals with autism diagnosed
in childhood. Journal of Autism and Developmental
Disorders, 35, 351–360.
Bogin, B. (2013). Chapter 2, Childhood, adolescence
and longevity: A chapter on human Evolutionary
140
life history. In B. Hewlett (Ed.), Adolescent identify:
Evolutionary, cultural and developmental perspectives. New York: Routledge.
Brody, G. H., et al. (2013). Is resilience only skin
deep? Rural African Americans’ socioeconomic status–related risk and competence in preadolescence and psychological adjustment and
Allostatic load at age 19. Psychological Science.
doi:10.1177/0956797612471954.
Bryden, K. S., Peveler, R. C., Stein, A., Neil, A., Mayou,
R. A., & Dunger, D. B. (2001). Clinical and psychological course of diabetes from adolescence to young
adulthood a longitudinal cohort study. Diabetes Care,
24, 1536–1540.
Busse, F., Hiermann, P., Galler, A., Stumvoll, M.,
Wiessner, T., Kiess, W., & Kapellen, T. (2007).
Evaluation of patients’ opinion and metabolic control
after transfer of young adults with type 1 diabetes
from a pediatric diabetes clinic to adult care. Hormone
Research in Pædiatrics, 67, 132–138.
Catalano, R. F., Berglund, M. L., Ryan, J. A., Lonczak,
H. S., & Hawkins, J. D. (2004). Positive youth development in the United States: Research findings on
evaluations of positive youth development programs.
The Annals of the American Academy of Political and
Social Science, 591, 98–124.
Cherlin, A. J., Chase-Lansdale, P. L., & Mcrae, C. (1998).
Effects of parental divorce on mental health throughout the life course. American Sociological Review,
63(2), 239–249.
Cicchetti, D., et al. (1999). A developmental psychopathology perspective on drug abuse. Drug abuse:
Origins & interventions (pp. 97–117). Washington,
DC: American Psychological Association, xxiii,
492 pp.
Cimera, R. E., & Cowan, R. J. (2009). The costs of services and employment outcomes achieved by adults
with autism in the us. Autism, 13, 285–302.
Cooney, T. M., & Uhlenberg, P. (1992). Support from parents over the life course: The adult Child's perspective.
Social Forces, 71, 63–84.
Copelovitch, L., Warady, B. A., & Furth, S. L. (2011).
Insights from the chronic kidney disease in children
(Ckid) study. Clinical Journal of the American Society
of Nephrology, 6, 2047–2053.
Cote, J. E. (2014). The dangerous myth of emerging adulthood: An evidence-based critique of a flawed developmental theory. Applied Developmental Science, 18(4),
177–188.
Dahl, R. E. (2004). Adolescent brain development: A
period of vulnerabilities and opportunities. Keynote
address. Annals of the New York Academy of Sciences,
1021, 1–22.
Duncan, G. J., & Brooks-Gunn, J. (2000). Family poverty, welfare reform, and child development. Child
Development, 71, 188–196.
Eaves, L. C., & Ho, H. H. (2008). Young adult outcome
of autism Spectrum disorders. Journal of Autism and
Developmental Disorders, 38, 739–747.
D. Wood et al.
Erikson, E. H. (1994). Identity: Youth and crisis.
New York: Ww Norton & Company.
Evans, G. W., & Cassells, R. C. (2014). Childhood poverty, cumulative risk exposure, and mental health in
emerging adults. Clinical Psychological Science, 2(3),
287–296.
Feliciano, C., & Ashtiani, M. (2012). Postsecondary educational pathways of low-income youth: An analysis
of add health data. Irvine: Uc/Accord.
Fergusson, D. M., & Woodward, L. J. (2002). Mental
health, educational, and social role outcomes of
adolescents with depression. Archives of General
Psychiatry, 59, 225–231.
Ferris, M. E., Gipson, D. S., Kimmel, P. L., & Eggers,
P. W. (2006). Trends in treatment and outcomes of
survival of adolescents initiating end-stage renal disease care in the United States of America. Pediatric
Nephrology, 21, 1020–1026.
Fredericks, E. M., Dore-Stites, D., Well, A., Magee, J. C.,
Freed, G. L., Shieck, V., & James Lopez, M. (2010).
Assessment of transition readiness skills and adherence in pediatric liver transplant recipients. Pediatric
Transplantation, 14, 944–953.
Furstenberg, F. F. (2002). How it takes thirty years to do a
study. In Looking at lives: American longitudinal studies of the twentieth century (pp. 37–57).
Galobardes, B., Lynch, J. W., & Smith, G. D. (2008). Is
the Association between childhood socioeconomic
circumstances and cause-specific mortality established? Update of a systematic review. Journal of
Epidemiology and Community Health, 62, 387–390.
Garcia-Villamisar, D., Ross, D., & Wehman, P. (2000).
Clinical differential analysis of persons with autism
in a work setting: A follow-up study. Journal Of
Vocational Rehabilitation, 14, 183–185.
Gerson, A. C., Butler, R., Moxey-Mims, M., Wentz,
A., Shinnar, S., Lande, M. B., Mendley, S. R.,
Warady, B. A., Furth, S. L., & Hooper, S. R. (2006).
Neurocognitive outcomes in children with chronic
kidney disease: Current findings and contemporary
endeavors. Mental Retardation and Developmental
Disabilities Research Reviews, 12(3), 208–215.
Gillespie-Lynch, K., Sepeta, L., Wang, Y., Marshall, S.,
Gomez, L., Sigman, M., & Hutman, T. (2012). Early
childhood predictors of the social competence of adults
with autism. Journal of Autism and Developmental
Disorders, 42, 161–174.
Gilman, S. E. (2012). The successes and challenges of
life course Epidemiology: A commentary on Gibb,
Fergusson and Horwood (2012). Social Science &
Medicine, 75, 2124–2128.
Gilman, S. E., & Loucks, E. B. (2012). Invited commentary: Does the childhood environment influence
the Association between every X and every Y in
adulthood? American Journal of Epidemiology, 176,
684–688.
Gilmore, D. D. (1990). Manhood in the making: Cultural
concepts of masculinity. New Haven: Yale University
Press.
Emerging Adulthood as a Critical Stage in the Life Course
Greene, A., Wheatley, S. M., & Aldava, J. F. (1992).
Stages on Life’s way Adolescents’ implicit theories
of the life course. Journal of Adolescent Research, 7,
364–381.
Grotevant, H. D., & Cooper, C. R. (1986). Individuation in
family relationships. Human Development, 29, 82–100.
Guerra, N. G., & Bradshaw, C. P. (2008). Linking the
prevention of problem behaviors and positive youth
development: Core competencies for positive youth
development and risk prevention. New Directions for
Child and Adolescent Development, 2008, 1–17.
Halfon, N., & Forrest, C. B. (2017). The emerging theoretical framework of life course health development. In
N. Halfon, C. B. Forrest, R. M. Lerner, & E. Faustman
(Eds.), Handbook of life course health-development
science. Cham: Springer.
Helgeson, V. S., Snyder, P. R., Escobar, O., Siminerio,
L., & Becker, D. (2007). Comparison of adolescents
with and without diabetes on indices of psychosocial functioning for three years. Journal of Pediatric
Psychology, 32, 794–806.
Hogg, R. J., Furth, S., Lemley, K. V., Portman, R.,
Schwartz, G. J., Coresh, J., Balk, E., Lau, J., Levin, A.,
& Kausz, A. T. (2003). National Kidney Foundation’s
kidney disease outcomes quality initiative clinical
practice guidelines for chronic kidney disease in children and adolescents: Evaluation, classification, and
stratification. Pediatrics, 111, 1416–1421.
Holmes-Walker, D., Llewellyn, A., & Farrell, K. (2007).
A transition care Programme which improves diabetes
control and reduces hospital admission rates in young
adults with type 1 diabetes aged 15–25 years. Diabetic
Medicine, 24, 764–769.
Howlin, P., Goode, S., Hutton, J., & Rutter, M. (2004).
Adult outcome for children with autism. Journal of
Child Psychology and Psychiatry, 45, 212–229.
Jacquet, S. E., & Surra, C. A. (2001). Parental divorce and
premarital couples: Commitment and other relationship characteristics. Journal of Marriage and Family,
63, 627–638.
Kessler, R. C., Berglund, P., Demler, O., Jin, R.,
Merikangas, K. R., & Walters, E. E. (2005). Lifetime
prevalence and age-of-onset distributions of Dsm-iv
disorders in the National Comorbidity Survey
Replication. Archives of General Psychiatry, 62,
593–602.
Kim, P., Neuendorf, C., Bianco, H., & Evans, G. W.
(2015). Exposure to childhood poverty and mental
health symptomatology in adolescence: A role of coping strategies. Stress and Health, 32, 494–502.
Koukourgianni, F., Harambat, J., Ranchin, B., Euvrard,
S., Bouvier, R., Liutkus, A., & Cochat, P. (2010).
Malignancy incidence after renal transplantation
in children: A 20-year single-Centre experience.
Nephrology Dialysis Transplantation, 25, 611–616.
Lerner, R. M. (2014). Developmental science, developmental systems, and contemporary theories of human
development. In Handbook of child psychology.
New York: Wiley and Sons.
141
Lotstein, D. S., Inkelas, M., Hays, R. D., Halfon, N., &
Brook, R. (2008). Access to care for youth with special health care needs in the transition to adulthood.
Journal of Adolescent Health, 43, 23–29.
Luciana, M., Conklin, H. M., Hooper, C. J., & Yarger,
R. S. (2005). The development of nonverbal working
memory and executive control processes in adolescents. Child Development, 76, 697–712.
Madsen, S. D., Roisman, G. I., & Collins, W. A. (2002).
The intersection of adolescent development and intensive intervention: Age-related psychosocial correlates
of treatment regimens in the diabetes control and complication trial. Journal of Pediatric Psychology, 27,
451–459.
Masten, A. S., & Coatsworth, J. D. (1998). The development of competence in favorable and unfavorable
environments: Lessons from research on successful
children. American Psychologist, 53, 205.
Masten, A. S., Coatsworth, J. D., Neemann, J., Gest, S. D.,
Tellegen, A., & Garmezy, N. (1995). The structure and
coherence of competence from childhood through
adolescence. Child Development, 66, 1635–1659.
Masten, A. S., Hubbard, J. J., Gest, S. D., Tellegen, A.,
Garmezy, N., & Ramirez, M. (1999). Competence in
the context of adversity: Pathways to resilience and
maladaptation from childhood to late adolescence.
Development and Psychopathology, 11, 143–169.
Masten, A. S., Burt, K. B., Roisman, G. I., Obradovic,
J., Long, J. D., & Tellegen, A. (2004). Resources and
resilience in the transition to adulthood: Continuity
and change. Development and Psychopathology, 16,
1071–1094.
Masten, A. S., Roisman, G. I., Long, J. D., Burt, K. B.,
Obradović, J., Riley, J. R., Boelcke-Stennes, K., &
Tellegen, A. (2005). Developmental cascades: Linking
academic achievement and externalizing and internalizing symptoms over 20 years. Developmental
Psychology, 41, 733.
Masten, A. S., Burt, K. B., & Coatsworth, J. D. (2006).
Competence and psychopathology in development. D.
Cicchetti, C. Dante, & J. Donald (Eds). Developmental
psychopathology: Risk, disorder, and adaptation (Vol.
3, 2nd ed., pp. 696–738). Hoboken: John Wiley &
Sons Inc, xvi 944 pp.
Mawhood, L., & Howlin, P. (1999). The outcome of a
supported employment scheme for high-functioning
adults with autism or Asperger syndrome. Autism, 3,
229–254.
Mckenna, A. M., Keating, L. E., Vigneux, A., Stevens,
S., Williams, A., & Geary, D. F. (2006). Quality of
life in children with chronic kidney disease—Patient
and caregiver assessments. Nephrology Dialysis
Transplantation, 21, 1899–1905.
Mikulincer, M., & Shaver, P. R. (2009). The attachment
and behavioral systems perspective on social support.
Journal of Social and Personal Relationships, 26(1),
7–19.
Miller, G. E., Yu, T., Chen, E., & Brody, G. H. (2015 Aug
18). Self-control forecasts better psychosocial outcomes
142
but faster epigenetic aging in low-Ses youth. Proceedings
Of The National Academy Of Sciences., 112(33),
10325–10330.
Modell, J. (1991). Into One’s own: From youth to adulthood in the United States, 1920–1975. Berkeley:
University of California Press.
Moffitt, T. E., & Caspi, A. (2001). Childhood predictors
differentiate life-course persistent and adolescencelimited antisocial pathways among males and females.
Devcelopment and Psychopathology, 13, 355–375.
Nelson, C. A., Thomas, K. M., & De Haan, M. (2012).
Neuroscience of cognitive development: The role of
experience and the developing brain. Hoboken: John
Wiley & Sons.
Newman, L., Wagner, M., Knokey, A.-M., Marder, C.,
Nagle, K., Shaver, D., & Wei, X. (2011). The post-high
school outcomes of young adults with disabilities up to
8 years after high school: A Report from the National
Longitudinal Transition Study-2 (Nlts2). Ncser 2011–
3005. Washington, DC: National Center For Special
Education Research.
Nurmi, J.-E. (1999). 15 self-definition and mental
health during adolescence and young adulthood.
In J. Schulenberg, J. L. Maggs, & K. Hurrelmann
(Eds.), Health risks and developmental transitions
during adolescence (p. 395). New York: Cambridge
University Press.
Oeffinger, K. C., Mertens, A. C., Sklar, C. A., Kawashima,
T., Hudson, M. M., Meadows, A. T., Friedman, D. L.,
Marina, N., Hobbie, W., & Kadan-Lottick, N. S.
(2006). Chronic health conditions in adult survivors of
childhood cancer. New England Journal of Medicine,
355, 1572–1582.
Osgood, D. W., Ruth, G., Eccles, J. S., Jacobs, J. E. & Barber,
B. L. (2005). Six paths to adulthood: Fast starters, parents without careers, educated partners, educated singles, working singles, and slow starters. na.
Reiss, J. G., Gibson, R. W., & Walker, L. R. (2005). Health
care transition: Youth, family, and provider perspectives. Pediatrics, 115, 112–120.
Rifkin, J. (2011). The Thired industrial revolution.
New York: St. Martin’s Press.
Roisman, G. I., Aguilar, B., & Egeland, B. (2004).
Antisocial behavior in the transition to adulthood:
The independent and interactive roles of developmental history and emerging developmental tasks.
Development and Psychopathology, 16, 857–871.
Rotundo, E. A. (1994). American manhood:
Transformations in masculinity from the revolution to
the modern era. New York: Basic Books.
Roux, A. M., Shattuck, P. T., Rast, J. E., Rava, J. A., &
Anderson, K.,. A. (2015). National autism indicators
report: Transition into young adulthood. Philadelphia:
Life Course Outcomes Research Program/A.J. Drexel
Autism Institute/Drexel University.
Ryan, R. M., & Lynch, J. H. (1989). Emotional autonomy
versus detachment: Revisiting the vicissitudes of adolescence and young adulthood. Child Development,
60(2), 340–356.
Saran, R., Li, Y., Robinson, B., Ayanian, J., Balkrishnan,
R., Bragg-Gresham, J., Chen, J., Cope, E., Gipson,
D. Wood et al.
D., & He, K. (2015). US renal data system 2014
annual data Report: Epidemiology of kidney disease
in the United States. American Journal of Kidney
Diseases: The Official Journal of the National Kidney
Foundation, 65, A7.
Sarnak, M. J. (2003). Cardiovascular complications in
chronic kidney disease. American Journal of Kidney
Diseases, 41, 11–17.
Scheer, S. D., Unger, D. G., & Brown, M. B. (1996).
Adolescents becoming adults: Attributes for adulthood. Adolescence, 31, 127.
Schlegel, A., & Barry Iii, H. (1991). Adolescence: An
anthropological inquiry. New York: Free Press.
Schulenberg, J. E., & Maggs, J. L. (2002). A developmental perspective on alcohol use and heavy drinking
during adolescence and the transition to young adulthood. Journal of Studies on Alcohol, Supplement No.
14, 54–70.
Schulenberg, J. E., Sameroff, A. J., & Cicchetti, D. (2004).
The transition to adulthood as a critical juncture in
the course of psychopathology and mental health.
Development and Psychopathology, 16, 799–806.
Shanahan, M. J. (2000). Pathways to adulthood in changing societies: Variability and mechanisms in life
course perspective. Annual Review of Sociology, 26,
667–692.
Shroff, R., Weaver, D. J., & Mitsnefes, M. M. (2011).
Cardiovascular complications in children with chronic
kidney disease. Nature Reviews Nephrology, 7, 642–649.
Silbereisen, R. K., & Lerner, R. M. (2007). Approaches
to positive youth development. Los Angeles/London:
Sage.
Skokauskas, N., & Gallagher, L. (2010). Psychosis,
affective disorders and anxiety in autistic Spectrum
disorder: Prevalence and Nosological considerations.
Psychopathology, 43, 8–16.
So, T.-Y., Bradley Layton, J., Bozik, K., Farrington, E.,
Gipson, P. E., Gibson, K., Primack, W., Conley Iii, W.,
Gipson, D. S., & Ferris, M. (2011). Cognitive pharmacy services at a pediatric nephrology and hypertension clinic. Renal Failure, 33, 19–25.
Spear, L. P. (2000). The adolescent brain and agerelated behavioral manifestations. Neuroscience &
Biobehavioral Reviews, 24(4), 417–463.
Steelman, L. C., & Powell, B. (1991). Sponsoring the
next generation: Parental willingness to pay for higher
education. American Journal of Sociology, 96(6),
1505–1529.
Steinberg, L., Dahl, R., Keating, D., Kupfer, D. J., Masten,
A. S., & Pine, D. S. (2006). The study of developmental psychopathology in adolescence: Integrating
affective neuroscience with the study of context. In
D. Cicchetti & D. Cohen (Eds.), Handbook of developmental psychopathology (Vol. 2, pp. 710–741).
New York: Wiley.
Taylor, J. L., & Seltzer, M. M. (2010). Changes in the
autism behavioral phenotype during the transition
to adulthood. Journal of Autism and Developmental
Disorders, 40, 1431–1446.
Taylor, J. L., & Seltzer, M. M. (2011a). Changes in the
mother–child relationship during the transition to
Emerging Adulthood as a Critical Stage in the Life Course
adulthood for youth with autism Spectrum disorders.
Journal of Autism and Developmental Disorders, 41,
1397–1410.
Taylor, J. L., & Seltzer, M. M. (2011b). Employment and
post-secondary educational activities for young adults
with autism Spectrum disorders during the transition
to adulthood. Journal of Autism and Developmental
Disorders, 41, 566–574.
Wadsworth, M. E., et al. (2016). Poverty and the development of psychopathology. In: M. E. Wadsworth, G.
W. Evans, K. Grant, J. S. Carter, & S. Duffy (Eds.),
Development and psychopathology.Volume Four. Risk,
resilience, and intervention, Ch 8 (pp. 215–237).
Hoboken: John Wiley & Sons.
Walker, J. S., & Gowen, L. K. (2011). Community-based
approaches for supporting positive development in
youth and young adults with serious mental health
conditions. Portland: Research and Training Center
for Pathways to Positive Futures, Portland State
University.
Watson, A. R. (2000). Non-compliance and transfer
from Paediatric to adult transplant unit. Pediatric
Nephrology, 14, 469–472.
Watson, A. R. (2005). Problems and pitfalls of transition from Paediatric to adult renal care. Pediatric
Nephrology, 20, 113–117.
143
Watson, A. R., Harden, P., Ferris, M., Kerr, P. G., Mahan,
J., & Ramzy, M. F. (2011). Transition from pediatric to adult renal services: A consensus statement by
the international society of nephrology (Isn) and the
international pediatric nephrology Association (Ipna).
Pediatric Nephrology, 26, 1753–1757.
Webster, B. H., Jr., & Bishaw, A. (2006). U.S. Census
Bureau, American Community Survey Reports, ACS02, Income, Earnings, and Poverty Data From the
2005 American Community Survey. Washington, DC:
U.S. Government Printing Office.
Whitbeck, L., Hoyt, D. R., & Huck, S. M. (1994). Early
family relationships, intergenerational solidarity, and
support provided to parents by their adult children.
Journal of Gerontology, 49, S85–S94.
White, S. W., Oswald, D., Ollendick, T., & Scahill, L. (2009).
Anxiety in children and adolescents with autism Spectrum
disorders. Clinical Psychology Review, 29, 216–229.
Yeager, D. S., & Dweck, C. S. (2012). Mindsets that
promote resilience: When students believe that personal characteristics can be developed. Educational
Psychologist, 47(4), 302–314.
Yeung, E., Kay, J., Roosevelt, G. E., Brandon, M., &
Yetman, A. T. (2008). Lapse of care as a predictor
for morbidity in adults with congenital heart disease.
International Journal of Cardiology, 125, 62–65.
Open Access This chapter is licensed under the terms of the Creative Commons Attribution 4.0 International License
(http://creativecommons.org/licenses/by/4.0/), which permits use, sharing, adaptation, distribution and reproduction in
any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license and indicate if changes were made.
The images or other third party material in this chapter are included in the chapter’s Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the chapter’s Creative Commons license
and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain
permission directly from the copyright holder.
Pregnancy Characteristics
and Women’s Cardiovascular
Health
Abigail Fraser, Janet M. Catov, Deborah A. Lawlor,
and Janet W. Rich-Edwards
1
Introduction
Growing evidence suggests that pregnancy is a
“critical period” in a woman’s life when her health
development is especially sensitive to certain internal and external stimuli. As a normal response to
pregnancy and in order to support the developing
fetus, women become more insulin resistant and
hyperlipidemic and experience an increase in blood
pressure (BP; after an initial drop) and upregulation
This chapter contains a modified version of a previously
published review and analysis of existing research that
appeared in Epidemiologic Reviews. Reprinted with permission from:
Janet W. Rich-Edwards, Abigail Fraser, Deborah
A. Lawlor, Janet M. Catov; Pregnancy Characteristics and
Women’s Future Cardiovascular Health: An Underused
Opportunity to Improve Women’s Health?. Epidemiol
Rev 2014;36(1):57–70. doi: 10.1093/epirev/mxt006
AF and DAL work in a unit that receives infrastructure
funding from the United Kingdom Medical Research
Council (MC_UU_12013), and AF is funded by a United
Kingdom Medical Research Council fellowship (MR/
M009351/1). A grant to DAL from the Wellcome Trust
also supports this collaborative work (WT094529MA).
JMC is funded by RO1HL103825 and K12HD43441. JRE
is supported by an American Heart Association Founder’s
Grant (13GRNT17070022). The authors have no relevant
disclosures, financial or otherwise.
A. Fraser (*) • D.A. Lawlor
Medical Research Council Integrative Epidemiology
Unit at the University of Bristol, University of
Bristol, Bristol, UK BS8 2BN
e-mail: abigail.fraser@bristol.ac.uk
of coagulation factors and the inflammatory cascade (Sattar 2004). This is perhaps the prime example of health as an emergent property as it is one
that enables the bearing of offspring and thus the
perpetuation of humanity. While for a majority of
women, this adaptation to pregnancy remains
“healthy,” in some women it develops into a complication of pregnancy such as gestational diabetes
mellitus (GDM), preeclampsia, fetal growth
restriction (FGR), and preterm delivery.
It has long been understood that pregnancy
complications are important for the life course
health development of offspring, but much less
appreciated that these complications also have
key implications for the long-term health development of the mother. An accumulating body of
research has shown that these common pregnancy
J.M. Catov
Department of Obestetrics and Gynecology,
University of Pittsburgh, Pittsburgh, PA 15213, USA
University of Pittsburgh, Department of
Epidemiology, Pittsburgh, PA 15261, USA
Magee-Womens Research Institute,
Pittsburgh, PA 15213, USA
J.W. Rich-Edwards
Connors Center for Women’s Health and Gender
Biology, Brigham and Women’s Hospital,
Boston, MA 02120, USA
Harvard Medical School, Boston, MA 02115, USA
Harvard School of Public Health,
Boston, MA 02120, USA
© The Author(s) 2018
N. Halfon et al. (eds.), Handbook of Life Course Health Development,
DOI 10.1007/978-3-319-47143-3_8
145
A. Fraser et al.
146
complications predict the future risk of chronic
diseases in women, including cardiovascular disease, diabetes, and breast cancer (Rich-Edwards
2009). In this chapter we use life course health
development theoretical principles as a lens for an
examination of the implications of pregnancy history for cardiovascular disease (CVD), a leading
cause of female mortality (Oblast 1999; Yusuf
et al. 2001).
Globally, one out of three women dies from
CVD (Shah et al. 2009; Mathers et al. 2008). We
do a worse job of recognizing and predicting
CVD in women than in men, in part because CVD
presents itself differently between the sexes
(Mosca et al. 2011; Shaw et al. 2006). This has
important implications for the prevention of CVD.
Primary prevention, if applied to high risk populations early enough to avert the cumulative damage
of chronic disease, can reduce CVD incidence
(Scarborough and Weissberg 2011; Shay et al.
2012; MMWR 1989). In response to the growing
appreciation that many preventive efforts start too
late to be effective, there has been a call for “primordial prevention”—the prevention of the major
CVD risk factors themselves (Labarthe 1999;
Weintraub et al. 2011). In this context, pregnancy
complications have the potential to be effective
CVD risk “stress tests” to identify women who
would most benefit from primordial or primary
prevention efforts to reduce CVD risk (Sattar and
Greer 2002). The concept of primordial prevention is consistent with the life course health development principle that health development is an
emergent phenomenon and the best way to prevent future disease is to build health assets that
have long-term salutary benefits.
On average, more than 80% of women in highincome countries bear at least one child (Martinez
et al. 2012; OECD Family D 2014), as do upward
of 90% of women in most lower- and middleincome nations (United Nations DoEaSA,
Population Division 2009). A high proportion of
women will, in the course of their reproductive
career, have a pregnancy complicated by GDM, a
hypertensive disorder of pregnancy, FGR macrosomia, or preterm delivery. The prevalence of any
one of these conditions in any given pregnancy
ranges from 2% to greater than 12%. In one UK
study, 36% of singleton pregnancies were compli-
cated by at least one of these factors (Fraser et al.
2012). In the U.S. national Nurses’ Health Study 2,
we estimated that 29% of parous study participants
have had one of these pregnancy complications. As
reviewed below, each of these complications has
been associated with roughly a twofold increase in
the risk of CVD events. If 80% of women are parous and 30% of them have had a pregnancy complication predictive of CVD, then about 25% of
women are at heightened risk for future CVD risk.
We begin with a review of the evidence for
associations of parity and common pregnancy
complications (low birth weight, fetal growth
restriction, preterm delivery, hypertensive disorders of pregnancy, and GDM) with future CVD
risk. We conducted MEDLINE searches for
English-language cohort and case-control studies published in the peer-reviewed literature
through December 2012, as described in detail
elsewhere (Rich-Edwards et al. 2014). Whether
pregnancy is a sensitive period in terms of cardiovascular health development across the life
course and complications per se contribute to
long term CVD risk, whether they simply
unmask women with an underlying propensity
for CVD, or whether both pathways are in play
remains unclear. As suggested by life course
health development, longitudinal studies are
needed to untangle these temporal effects. In the
second part of the chapter, we further explore
the physiologic mechanisms that might explain
the associations between pregnancy complications and CVD. Finally, we discuss the implications of these findings for future research as
well as for health care design and policy.
2
Associations of Parity
and Pregnancy
Complications with CVD Risk
in Mothers
2.1
Parity and CVD
Most (Green and Moser 1988; Ness et al. 1993)
but not all studies (Steenland et al. 1996) have
found a positive association between parity
(number of children) and later CVD. In the largest study to date, the association was examined in
Pregnancy Characteristics and Women’s Cardiovascular Health
1.3 million with a median follow-up time of 9.5
years (range 0–24) women using Swedish registry data (Parikh et al. 2010). Parity was associated with CVD in a J-shaped fashion, with two
births representing the nadir of risk. Compared
with women with two births, the multivariableadjusted hazard ratios (95% confidence interval
(CI)) for women with 0 and ≥5 births were 1.11
(1.09–1.14) and 1.57 (1.52–1.64), respectively.
Desired family size may affect the shape of the
parity-CVD risk distribution in different societies
and is an example of how societal norms and social
structures may affect health development and its
determinants. In Sweden, the modal family size
(two children) coincides with the nadir of maternal
cardiovascular risk (Parikh et al. 2010). This suggests that many women who bore only one child
suffered from secondary infertility, first pregnancy
complications that precluded further pregnancies,
or severe neonatal outcomes that discouraged further childbearing. To the extent that subfertility and
severe pregnancy complications predict future
CVD risk, they may explain the low-parity “hook”
of the J-shaped association of parity and maternal
CVD. The increase in CVD risk with increasing
parity after two children may be the result of different phenomena. These include rival, but not mutually exclusive, theories that (1) adverse physiologic
change accumulates over pregnancies; (2) adverse
lifestyle habits accrue with more children; and/or
(3) selection bias in which women at higher CVD
risk opt for larger families. Thus, it is unclear
whether the association of higher parity with CVD
risk is causal or correlational.
Some insight into the association of parity
with maternal CVD risk may be gleaned by
examining the association of number of children
with paternal CVD risk. Similar associations for
mothers and fathers would suggest that the association between parity and maternal CVD is not
causal, but is more likely a result of confounding
by socioeconomic position and/or behaviors
related to child-rearing. Three reports examined
associations of number of children with CVD in
fathers. In general, men who have fathered the
most children appear to have small increased
CVD risk, though this association is not always
statistically significant and is weaker than the
147
associations observed among mothers (Dekker
and Schouten 1993; Lawlor et al. 2003; Ness
et al. 1995). Adjustment for lifestyle factors tends
to reduce the associations in both mothers and
fathers (Lawlor et al. 2003; Catov et al. 2007a).
These results suggest that the association between
high parity and CVD in later life may be largely
the result of socioeconomic position and/or
behavioral risk factors associated with childrearing that are shared by both parents.
2.2
Common Pregnancy
Complications and CVD
in Mothers
Offspring birth weight predicts maternal lifespan
(Catov et al. 2007a; Davey Smith et al. 1997,
2000a, b, 2007). Figure 1 presents the findings
from studies that have examined associations of
offspring birth weight or fetal growth—a function
of birth weight and gestation length—with maternal CVD risk (Davey Smith et al. 1997, 2007,
2000a, b, 2005; Bellamy et al. 2007, 2011;
Friedlander et al. 2007; Lykke et al. 2010a;
Mongraw-Chaffin et al. 2010; Smith et al. 2001;
Wikström et al. 2005; Fraser et al. 2012; Ness et al.
1993). One meta-analysis has calculated that, for
every standard deviation (roughly 500 g) higher
birth weight of the firstborn child, maternal CVD
mortality is decreased by 25% (Davey Smith et al.
2007). It is unclear whether the inverse association
of offspring birth weight with mortality is constant
across the entire range of birth weight, as the association of high birth weight with maternal CVD
risk varies by study. In some populations, the mothers of the largest infants (>4000 g or >4500 g) have
the lowest risks of CVD (Davey Smith et al. 1997,
2000b), while in other populations there is an
uptick in CVD risk for the mothers of macrosomic
newborns (Davey Smith et al. 2007; Bonamy et al.
2011; Friedlander et al. 2007; Lykke et al. 2012).
Given the strong associations of macrosomia with
GDM and later type 2 diabetes (Metzger et al.
1993), the presence and magnitude of the association of large birth weight with future CVD risk may
depend on the population prevalence of GDM and
chronic diabetes during pregnancy (in other words,
A. Fraser et al.
148
Relative Risk
(95% CI)
First Author, Year (Reference No.)
Mean or
Median
Years of
Follow-up
Caption
Notes
Birthweight
Davey Smith,1997
1.23 (0.97, 1.57)
15
a, h
Davey Smith, 2000
1.49 (1.21, 1.84)
10
a, h
Davey Smith, 2000
1.30 (1.12, 1.51)
34
a, h
Smith, 2001
2.20 (1.80, 2.80)
17
b, i
Davey Smith, 2007
1.25 (1.14, 1.37)
45
a, j
Friedlander, 2007
2.11 (1.38, 3.21)
34
c, j
d, h
Fetal Growth
Davey Smith, 2005
1.39 (1.23, 1.57)
20
Wikstrom, 2005
1.80 (1.60, 2.30)
19–28
e, i
Lykke, 2010
2.56 (2.19, 3.00)
15
e, h
Mongraw-Chaffin, 2010
1.44 (1.03, 2.03)
37
f, h
Bonamy, 2011
2.13 (1.87, 2.44)
12
g, k
0.5
1
1.0
2.0
3.0
4.0
Relative Risk
Fig. 1 Results from studies of offspring birth weight or
fetal growth and relative risk of maternal cardiovascular
disease (CVD). a Per 1 standard deviation (SD) lower
birth weight; b lowest birth weight quintile compared to
all others; c <2500 g compared to 1500–3999 g birth
weight; d per 1 SD lower birth weight, adjusted for gesta-
tional age; e small for gestational age; f intrauterine
growth retardation; g ~2 SD below mean birth weight
adjusted for gestational age; h CVD mortality; i coronary
heart disease (CHD) events ; j CHD mortality; k CVD
events (CI confidence interval)
the extent to which large infant size is pathological). Indeed, the association of macrosomia with
CVD risk is attenuated by adjustment for GDM(36)
(Bonamy et al. 2011), indicating that a substantial
portion of the association of macrosomia and CVD
is explained by metabolic risk.
It is abundantly clear, however, that the 8% of
deliveries that are low birth weight (<2500 g) are
associated with twice the maternal CVD incidence and mortality of other deliveries (Davey
Smith et al. 1997, 2000a, b, 2007). Associations
of offspring birth weight with maternal CVD
are only modestly diminished by adjustment for
cigarette smoking and not affected by control for
prepregnancy body mass index (Bonamy et al.
2011; Davey Smith et al. 2005).
Birth weight is the product of fetal growth rate
and gestation length. Fetal growth, represented as
birth weight corrected for gestation length, predicts
maternal CVD risk (Bonamy et al. 2011; Lykke
et al. 2012), as does gestation length (discussed
below). In fact, the coincidence of restricted fetal
growth and prematurity yields a more than threefold increased CVD risk (Bonamy et al. 2011).
The curvilinear association of offspring birth
weight with maternal CVD risk observed in many
populations may be the product of competing
pathological phenomena. At one end of the birth
weight spectrum, the association of macrosomia
with maternal CVD risk may be explained by
underlying metabolic risk; at the other end of the
spectrum, the association of low birth weight with
maternal CVD risk may be driven by endothelial
dysfunction and other pathologies associated
with restricted fetal growth and preterm birth.
First offspring birth weight also predicts paternal CVD, although the magnitude of the positive
association of offspring birth weight with paternal
Pregnancy Characteristics and Women’s Cardiovascular Health
CVD risk is less than a third of that for the infant’s
mother (Davey Smith et al. 2007). The fact that
the birth weight of their first child predicts CVD
events in both parents suggests that shared lifestyle or environmental factors, such as cigarette
smoking, might influence both the growth of the
fetus and CVD risk in the parents and/or that
pleiotropic genetic variants affect both growth
and CVD risk. Birth weight is passed down
through maternal and paternal lines (Lie et al.
2006), opening the possibility that paternal CVD/
fetal growth genes could affect both the pregnancy outcome and long-term chronic disease
risk in the father (Freathy et al. 2007). However,
the stronger association in mothers than in fathers
suggests either parent-specific genomic imprinting or—as seems more parsimonious—that
maternal health during pregnancy affects fetal
growth and is a marker of her future CVD risk.
Preterm delivery (<37 weeks’ gestation)
accounts for 6–12% of deliveries in the developed
world (Beck et al. 2010). The hazard ratios for
CVD associated with total preterm delivery are
depicted in Fig. 2 and are on the order of 1.3–2.6
for births <37 completed weeks compared with
term births (Davey Smith et al. 2000b, 2005;
Bonamy et al. 2011; Lykke et al. 2010a, b; Smith
et al. 2001; Wikström et al. 2005; Catov et al.
2010a; Irgens et al. 2001; Nardi et al. 2006; Pell
et al. 2004; Rich-Edwards et al. 2012). There is a
greater range of relative risk when distinct preterm phenotypes are examined separately. While
most preterm deliveries follow spontaneous labor
or preterm premature rupture of membranes, a
significant and growing fraction results from
medically induced labor or Caesarean section
without labor. The chief reasons for these medically indicated deliveries include preeclampsia
and FGR, both of which have been associated
with increased maternal CVD risk. In studies that
have distinguished them, hypertensive preterm
deliveries consistently have a stronger association
with maternal CVD outcomes than do normotensive preterm deliveries, though the latter are still
associated with a 1.2- to threefold increased risk
compared with term deliveries (Catov et al.
2010a; Irgens et al. 2001). In the two studies that
have contrasted CVD risk among mothers with
149
spontaneous versus indicated preterm deliveries
(Rich-Edwards et al. 2012; Hastie et al. 2011a),
indicated delivery was associated with higher
risks of CVD mortality than spontaneous preterm
delivery. Nevertheless, spontaneous preterm
delivery—compared with term delivery—was
associated with doubling of CVD risk (RichEdwards et al. 2012; Hastie et al. 2011a).
Unlike the associations of parity or birth
weight with paternal CVD risk, two studies
(Davey Smith et al. 2005; Irgens et al. 2001) have
reported that preterm delivery is not associated
with paternal risk of CVD, implying that the
association of preterm delivery with maternal
CVD risk is not the product of a high-CVD risk
lifestyle or genetic variants shared between both
parents and their offspring. Of relevance, preterm
birth risk appears to be passed only through the
maternal line (Wilcox et al. 2008). These observations suggest that maternal intrauterine environment and health determine the risk of preterm
delivery and explain its association with maternal
CVD risk, rather than shared lifestyle or environment of the mother and father.
Gestational diabetes mellitus is a common and
growing pregnancy complication that affects as
many as 5% of pregnancies. It is well established that
women with GDM are at increased risk of developing diabetes later in life (Bellamy et al. 2009);
between 3% and 70% of women with a history of
GDM will develop type 2 diabetes within three
decades of the pregnancy (Kim et al. 2002), with a
meta-analysis of 675,455 women finding a sevenfold increase in risk of later type 2 diabetes (Bellamy
et al. 2009). Type 2 diabetes is an important CVD
risk factor, having a markedly higher relative and
absolute association with CVD in women than it
does in men (Sarwar et al. 2010). Given these associations, it seems self-evident that a history of GDM
would be associated with increased CVD risk.
However, due largely to the fact that GDM screening during pregnancy was neither routine nor standardized until recent decades, there are few cohorts
with long enough follow-up of screened populations to detect CVD incidence or mortality among
women with a history of GDM (Shah et al. 2008;
Carr et al. 2006). These are displayed in Fig. 3. The
only large population-based study of this topic is a
A. Fraser et al.
150
Relative Risk
(95% CI)
Mean or
Median
Years of
Follow-up
Caption
Notes
Davey Smith, 2000
2.06 (1.22, 3.47)
30
a, b
Irgens, 2001
2.95 (2.12, 4.81)
13
c, d
Smith, 2001
2.10 (1.50, 3.00)
15–19
a, e
Davey Smith, 2005
2.45 (2.06, 2.91)
20
a, b
Wikstrom, 2005
1.30 (1.10, 1.50)
19–28
a, e
Nardi, 2006
2.12 (1.19, 3.78)
Unknown
a, f
Catov, 2010
1.36 (1.31, 1.41)
28
a, g
Lykke, 2010
1.49 (1.36, 1.64)
14
h, e
Bonamy, 2011
1.68 (1.50, 1.88)
12
h, g
Hastie, 2011
1.58 (1.47, 1.71)
22
a, e
First Author, Year (Reference No.)
0.5
1.0
2.0
3.0
4.0
Relative Risk
Fig. 2 Results from studies of preterm delivery and relative risk of maternal cardiovascular disease (CVD). a <37
weeks’ gestation weeks compared with term; b CVD mortality; c <37 weeks’ gestation length compared with term
normotensive pregnancies; d CVD mortality, excluding
stroke mortality; e coronary heart disease (CHD) events; f
myocardial infarction; g CVD events; h 32–36 weeks’
gestation length compared with term (CI confidence
interval)
record linkage study conducted in Ontario, Canada,
with a median follow-up of 11.5 years (Shah et al.
2008). In that study, a history of GDM was associated with a greater risk of hospital admission for
acute myocardial infarction, coronary bypass, coronary angioplasty, stroke, or carotid endarterectomy
(hazard ratio (HR) = 1.71; 95% CI, 1.08–2.69).
Upon adjustment for diabetes after pregnancy, the
association was attenuated toward the null (adjusted
HR, 1.13; 0.67–1.89). A smaller, cross-sectional
study found that women with a history of GDM had
a higher CVD risk than women without a history of
GDM (adjusted OR = 1.85, 1.21–2.82) and experienced CVD events 7 years earlier, on average (Carr
et al. 2006).
Lesser degrees of antepartum hyperglycemia
have also been associated with an elevated risk of
subsequent diabetes and CVD. In the Ontario
study, women with evidence of elevated glycemia short of GDM criteria were at an increased
risk of diabetes (HR = 2.56, 2.28–2.87)
(Retnakaran and Shah 2009a) and CVD
(HR = 1.19, 1.02–1.39) (Retnakaran and Shah
2009b) compared to normoglycemic women.
Hypertensive disorders of pregnancy (HDPs)
are common pregnancy complications that presage CVD. Preeclampsia, the combination of
hypertension and proteinuria, affects approximately 2–5% of pregnancies, with a predominance among first pregnancies (Fraser et al. 2012;
Wallis et al. 2008; North et al. 2011). Estimates
of the prevalence of gestational hypertension,
new-onset hypertension without proteinuria, vary
from 3% to 14% (Fraser et al. 2012; Wallis et al.
2008; Roberts et al. 2005). Women with a history
of preeclampsia have roughly fourfold higher
incidence of hypertension and twofold elevated
risks of heart disease, stroke, and venous thromboembolism (Bellamy et al. 2007; McDonald
et al. 2008). Two systematic reviews, one of
cohort studies (n = 25) and the other of both
cohort (n = 10) and case-control (n = 5) studies,
Pregnancy Characteristics and Women’s Cardiovascular Health
First Author, Year
(Reference No.)
151
Relative Risk
(95% CI)
Mean or
Median
Years of Caption
Follow-up Notes
Carr, 2006
1.58 (1.00, 2.49)
30
a
Shah, 2008
1.71 (1.08, 2.69)
12
b
0.5
1.0
2.0
3.0
4.0
Relative Risk
Fig. 3 Results from studies of gestational diabetes mellitus and relative risk of maternal cardiovascular disease (CVD).
a Self-reported coronary artery disease, b CVD events (CI confidence interval)
have both reported a doubling of risk for different
measures of CVD comparing women with preeclampsia to normotensive women over a median
of 10–12-year follow-up (Bellamy et al. 2007;
McDonald et al. 2008). Figure 4 depicts the relative risk of coronary heart disease (CHD) and
CVD outcomes among mothers with a history of
preeclampsia (Mongraw-Chaffin et al. 2010;
Smith et al. 2001; Wikström et al. 2005; Irgens
et al. 2001; Funai et al. 2005; Hannaford et al.
1997; Jónsdóttir et al. 1995; Kestenbaum et al.
2003; Lin et al. 2011; Lykke et al. 2009; Wilson
et al. 2003).
Publications from three cohort studies published since those reviews give some insight into
the onset and duration of CVD risk following
HDP (Mongraw-Chaffin et al. 2010; Smith et al.
2001; Lin et al. 2011; Lykke et al. 2009). In a
short-term follow-up of over 1,000,000 pregnancies in Taiwan, women with preeclampsia/
eclampsia were at double the risk of major CVD
from the third trimester of pregnancy up to three
years postpartum, with particularly high relative
risks for stroke (HR = 14.5, 1.3–165.1) and myocardial infarction (HR = 13.0, 4.6–6.3) (Lin et al.
2011). While these results suggest a high relative
risk immediately following HDP, the confidence
intervals are wide, and the absolute risk of CVD
events is very small at this age, so that this immediate risk is unlikely to account for a large number of CVD events. The Child Health and
Development Study in California has provided
some of the longer follow-up; over 37 years after
pregnancy, women with a history of preeclampsia in any pregnancy had double the risk of CVD
death (HR = 2.14; 1.29–3.57) (Mongraw-Chaffin
et al. 2010). This doubling of risk is consistent
with studies with shorter duration of follow-up.
Considering the exponential increase in the absolute numbers of CVD events with increasing age,
this suggests that the elevated risk of CVD among
women with a history of HDP is not limited to the
early years postpartum.
Thus, studies repeatedly report a doubling
of CVD risk among women with a history of
preeclampsia and suggest lesser degrees of
excess risk among women with a history of gestational hypertension, despite the strong association of gestational hypertension with development
of chronic hypertension (Lykke et al. 2009). The
combination of preterm delivery and preeclampsia—a likely marker of the severity of preeclampsia—is a particularly potent predictor of CVD
risk. Compared to normotensive term pregnancies, women delivering preterm preeclamptic
pregnancies have very high relative risks of future
A. Fraser et al.
152
Relative Risk
(95% CI)
First Author, Year (Reference No.)
Mean or
Median
Years of Caption
Follow-up Notes
Jonsdottir, 1995
1.90 (1.02, 3.52)
Unknown
a
Hannaford, 1997
1.65 (1.26, 2.16)
Up to 26
b
Irgens, 2001
3.61 (0.76, 17.18)
13
c, e
Smith, 2001
2.10 (1.60, 2.60)
15–19
Kestenbaum, 2003
2.55 (1.70, 3.83)
Wilson, 2003
1.95 (0.90, 4.22)
Funai, 2005
3.01 (2.18, 4.33)
30
d
Wikstrom, 2005
2.21 (1.56, 3.31)
19–28
b, e
Lykke, 2009
1.82 (1.65, 2.00)
15
b
Mongraw-Chaffin, 2010
2.73 (1.78, 4.18)
37
d
Skjaerven, 2012
1.90 (1.60, 2.20)
25
d
0.5
1.0
2.0
3.0
4.0
8
Unknown
b
c, e
a
4.5
Relative Risk
Fig. 4 Results from studies of hypertensive disorders of
pregnancy and relative risk of maternal cardiovascular disease (CVD). a Coronary heart disease (CHD) mortality, b
CHD events, c CVD events, d CVD mortality, e composite
estimate provided by Bellamy review (18). A 2011 study
by Lin (62) reported a relative risk of 23.0 (95% confidence interval (CI), 5.1–103.7) for CVD events (except
stroke) during pregnancy and up to three years after delivery. We omitted that study from the figure so that we could
keep the relative risk scale consistent across figures
CVD ranging from 2.5 to 9.5. (Mongraw-Chaffin
et al. 2010; Smith et al. 2001; Lin et al. 2011;
Skjaerven et al. 2012)
pregnancy has been associated with a modestly
increased risk of type 2 diabetes (adjusted
HR = 1.16, 1.01–1.34) and each non-GDM pregnancy with a reduced risk of diabetes (HR = 0.34,
0.27–0.41) (Retnakaran et al. 2011). In fact, this
highlights an intriguing pattern that is emerging
with respect to last births: having preeclampsia
(Skjaerven et al. 2012), preterm delivery (RichEdwards et al. 2012), or GDM (Retnakaran
et al. 2011) in the last pregnancy appears to be
associated with especially high risk of future
CVD in mothers. Perhaps reflecting the same
phenomenon, women who have one preterm
delivery and one term delivery in their first two
births appear to be at higher risk of CHD if the
preterm delivery was the second birth (Catov
et al. 2010a; Lykke et al. 2010b). This suggests
that pregnancy complications severe enough to
contraindicate or discourage a subsequent
pregnancy may be particularly potent predictors
of future CVD risk.
2.3
Recurrent Pregnancy
Complications, Last
Pregnancy Complications,
and Maternal CVD Risk
Much of the literature is based on first pregnancies, precluding examination of the association
of recurring pregnancy complications with CVD
risk. There is evidence that recurrent preeclampsia (Lykke et al. 2009) and preterm
delivery(Catov et al. 2010a; Lykke et al. 2010b)
are associated with a greater risk of CVD than a
single complicated pregnancy in multiparous
women. Although the association of recurrent
GDM with CVD risk has not been studied, after
a first GDM pregnancy, each subsequent GDM
Pregnancy Characteristics and Women’s Cardiovascular Health
3
Physiological Mechanisms
Linking Pregnancy
Complications to Maternal
CVD Risk
Pathways that link pregnancy exposures to later
life CVD are not well understood. Considerable
evidence supports the existence of common predisposing factors for both pregnancy complications and CVD risk suggesting that pregnancy
complications can be thought of as a failed stress
test, with pregnancy being the stressor. There
have been almost no studies examining the alternative that pregnancy complications might cause
increased CVD risk. To address this issue, we
summarize evidence that compares CVD risk
before, during, and after pregnancies with and
without complications.
3.1
Cardiovascular Risk Factors
Preceding Pregnancy
Complications
Higher prepregnancy blood pressure is a risk
factor for preeclampsia (Magnussen et al. 2007)
and preterm delivery (Catov et al. 2013). Chronic
hypertension has a well-established relation to
increased risk of preeclampsia—known as
“superimposed” preeclampsia. Even within the
normotensive range, there is a positive doseresponse association of prepregnancy systolic
and/or diastolic blood pressure with preeclampsia, (Magnussen et al. 2007) and women who
develop hypertensive disorders of pregnancy
have higher blood pressure at 8 weeks’ gestation
than normotensive (Macdonald-Wallis et al. 2012).
Preexisting hypertension has also been associated with FGR, especially in cases that were also
preterm (Catov et al. 2008a). Risks for these
complications also rise with increasing maternal
age, suggesting that the aging endothelium may
less successfully adapt to the profound vascular
demands of pregnancy.
Prepregnancy lipid concentrations are also
associated with pregnancy complications and
offspring birth weight; the nature of the associa-
153
tion varies with the pregnancy outcome in question. Lipid profiles consistent with elevated
CVD risk, including higher prepregnancy triglyceride levels, total cholesterol, and lower
HDL cholesterol, have been associated with
preeclampsia and preterm delivery in the study
in Norway (Magnussen et al. 2007, 2011). The
US Coronary Artery Risk Development in
Young Adults Study (CARDIA) found a curvilinear association of prepregnancy cholesterol
levels with risk of delivering preterm (Catov
et al. 2010b). With respect to fetal growth,
women with a more atherogenic lipid profile
may bear larger infants (Romundstad et al.
2007); this suggests that the association of low
birth weight—at least the fetal growth component
of low birth weight—with maternal CVD risk
may not operate via dyslipidemia.
Prepregnancy adiposity and glucose/insulin
dysregulation is strongly implicated in the etiology of GDM, based on the observation that
women with GDM tend to have a family history
of type 2 diabetes and higher body mass index
(BMI) before pregnancy (Solomon et al. 1997),
as well as higher levels of glucose and insulin and
lower levels of adiponectin before the onset of
the midpregnancy hyperglycemia that defines
GDM (Nanda et al. 2011; Riskin-Mashiah et al.
2010; Sacks et al. 2003; Williams et al. 2004).
Higher BMI and family history of diabetes are
also associated with increased risk of preeclampsia (Qiu et al. 2003; O’Brien et al. 2003). The risk
of preeclampsia doubles with every 5–7 kg/m2
increase in body mass index before pregnancy
(O’Brien et al. 2003).
Thus, subclinical elevations in the classic
CVD risk factors of blood pressure, lipid levels,
elevated BMI, and glucose/insulin dysregulation
appear to predate both preeclampsia and
GDM. Less clear is the extent to which CVD risk
factors precede spontaneous preterm deliveries or
FGR in normotensive pregnancies. Furthermore,
the roles of prepregnancy inflammatory and
coagulation factors with respect to pregnancy
complications remain to be elucidated, despite
the importance of these systems for both reproduction and CVD risk (Romero et al. 2007).
A. Fraser et al.
154
3.2
Cardiovascular Risk Factors
During Pregnancy
3.2.1
Cardiovascular Adaptation
in Normal Pregnancy
In normal gestation, maternal blood volume
increases progressively from 6 to 8 weeks’ gestation, peaking at an increase of 45% by 32 weeks
(Monga and Creasy 1994). Cardiac output
increases by 30–50%, with half of this increase
occurring very early in gestation. Pulse rate
increases 17%, and there are striking alterations
in renal physiology. Although the insulin
response to glucose is augmented in early pregnancy, insulin resistance emerges in the second
half of pregnancy (Butte 2000). In addition, cholesterol and triglyceride profiles change after gestation week 9 to support steroid synthesis and
fetal growth (Butte 2000). In uncomplicated
pregnancy, there is a tendency for low-density
lipoprotein (LDL) to shift across gestation from
large, buoyant particles to smaller, denser, and
more atherogenic particles (Hubel et al. 1998a).
Fat is accumulated during the second trimester
and then mobilized to support the dramatic fetal
growth of the third trimester (Herrera 2000).
3.2.2
Cardiovascular Risk Factors
During Pregnancy
Complications
Vascular and endothelial dysfunction is characteristic of pregnancies complicated by preeclampsia or growth restriction. Placental
underperfusion is common, and there are elevated
markers of endothelial dysfunction in the maternal circulation. Women with hypertensive disorders of pregnancy demonstrate increased
resistance in the uterine arteries (Ducey et al.
1987; Campbell and Griffin 1983), vascular stiffness, and impaired endothelial response
(Savvidou et al. 2003, 2011). In addition, placental vascular lesions indicative of failed spiral
artery remodeling, ischemia, or hemorrhage have
also been reported in cases of both medically
indicated and spontaneous preterm birth (Kelly
et al. 2009).
During pregnancy, lipid aberrations accompany several pregnancy complications. Again, the
direction of the associations appears to depend on
the nature of the pregnancy complication. The
dyslipidemias associated with atherosclerosis
(hypertriglyceridemia, hypercholesterolemia, elevated free fatty acids, and excess oxidized LDL)
are frequently seen during preeclampsia …
(Clausen et al. 2001; Hubel et al. 1996, 1998b;
Sattar et al. 1997). There is also emerging evidence to suggest that this atherogenic lipid profile
is associated with both spontaneous and indicated
preterm births (Edison et al. 2007; Catov et al.
2007b). Similarly, women with GDM exhibit elevations in triglycerides and, less consistently, total
cholesterol and LDL during pregnancy
(Enquobahrie et al. 2005). On the other hand, low
maternal total and LDL-cholesterol concentrations appear in the third trimester in pregnancies
complicated by FGR (Sattar et al. 1999). Placental
studies are conflicting, with some suggesting
reduced expression of lipoprotein receptors in
placentas from FGR vs. appropriate weight for
gestational age births (Wadsack et al. 2007) and
others suggesting overexpression of these receptors (Stepan et al. 1999). FGR studies are hampered by nonstandard phenotyping, and thus
findings may represent differing levels of severity.
Despite these limitations, these data suggest that
extremes of lipid concentrations are associated
with adverse pregnancy outcomes. Longitudinal
studies are needed to better understand how the
relative contributions of low or high cholesterol
are related to failed or compensatory lipid adaptation required to optimize fetal growth.
Metabolic dysregulation in pregnancy defines
GDM and is a strong risk factor for preeclampsia;
there is considerable overlap of the two conditions, with twice the rate of preeclampsia in diabetic versus nondiabetic pregnancies (Ostlund
et al. 2004). However, GDM has only a modest
association with spontaneous preterm birth
(Hedderson et al. 2003). Higher early-pregnancy
BMI is associated with increased risk of HDP
and GDM (Solomon et al. 1997; O’Brien et al.
2003; Lawlor et al. 2012), but with reduced risk
of SGA and spontaneous preterm birth in most
studies (Smith et al. 2007).
Systemic inflammation during pregnancy may
be important in the pathogenesis of several preg-
Pregnancy Characteristics and Women’s Cardiovascular Health
nancy complications. Elevated serum levels of
C-reactive protein and/or leukocytes have been
detected in women who experience GDM, FGR,
and both spontaneous and indicated preterm
deliveries (Catov et al. 2007c; Ernst et al. 2011;
Freeman et al. 2004; Pitiphat et al. 2005; Wolf
et al. 2001). However, neither mid-gestation circulating levels of C-reactive protein nor proinflammatory cytokines have proven to have
prognostic value for specific pregnancy outcomes
(Curry et al. 2007; Gammill et al. 2010).
Normal pregnancy is a state of hypercoagulability, and complications such as preeclampsia
and preterm birth are characterized by particularly high biomarkers of an activated fibrinolytic
cascade, as well as perhaps an impaired ability to
mount this response appropriately (Catov et al.
2008b; Hackney et al. 2010; Heilmann et al.
2007). It has been hypothesized that aberrations
in the cross talk between inflammation and the
coagulation cascades could contribute to the
pathophysiology of these pregnancy complications (Girardi 2011).
3.3
3.3.1
Cardiovascular Risk Factors
After Pregnancy
Enduring Cardiovascular
Impact of Normal Pregnancy
Most of the cardiovascular adaptations to normal
pregnancy resolve in the postpartum period,
although there are some detectable and lasting
pregnancy effects. Blood pressure is modestly
decreased in the postpartum period after a first
uncomplicated pregnancy (Hackney et al. 2010).
However, other lingering effects are not as
salutary. Importantly, women retain, on average,
0.5–5.0 kg of weight following each pregnancy
(Heilmann et al. 2007; Girardi 2011). Lactation
may help resolve the cardiometabolic adaptations
and fat accumulation associated with pregnancy
(Agatisa et al. 2004; Berends et al. 2008; Catov
et al. 2011).
The first birth may be a sentinel marker for
complications in later pregnancies and future
CVD risk (Lauenborg et al. 2005; Meyers-Seifer
and Vohr 1996; Verma et al. 2002). Several fac-
155
tors distinguish first births. First, longitudinal
studies suggest that the lasting blood pressure
and lipid changes associated with pregnancy
occur after first but not subsequent births
(Hackney et al. 2010). In addition, first births are
at higher risk for the major obstetric complications of preterm delivery, HDP, FGR, and stillbirth. Women with any of these complications are
at higher risk in subsequent pregnancies for
recurrence of the same complication as well as
the onset of other complications. Importantly,
complications during a first pregnancy impact the
likelihood of having a subsequent pregnancy. As
noted above, complications in a last pregnancy
appear to be associated with especially high relative risks of CVD events. Thus, health status of
the first and last pregnancies may be particularly
telling of future maternal health.
The cumulative effect of these adaptations and
resolutions and risks may contribute to the
above-noted J-shaped association between parity
and maternal CVD risk, with lowest risk for
women who have delivered two infants. It is not
clear whether pregnancies exert a cumulative cardiovascular burden with increasing parity,
whether higher-order pregnancies at more
advanced maternal age exert more cardiovascular
risk, or whether women at high cardiovascular
risk bear more children.
3.3.2
Cardiovascular Risk
After Pregnancy Complications
The association of vascular and endothelial dysfunction with pregnancy complications continues after delivery. Women with preeclampsia
have impaired endothelial function after pregnancy (Agatisa et al. 2004). This may also be
true, although to a lesser extent, of women who
deliver small babies due to FGR or preterm
delivery. For example, lower offspring birth
weight is associated with higher maternal blood
pressure in the years after pregnancy (Davey
Smith et al. 2005). Some (Catov et al. 2013;
Berends et al. 2008) but not all (Catov et al.
2011) studies report higher blood pressure and
atherosclerotic carotid vessel remodeling among
women who have delivered an FGR neonate.
Although studies are not unanimous (Macdonald-
156
Wallis et al. 2012; Lauenborg et al. 2005),
women with a history of GDM are more likely to
have hypertension (Meyers-Seifer and Vohr
1996; Verma et al. 2002), vascular dysfunction
(Heitritter et al. 2005), impaired endotheliumdependent vasodilatation (Anastasiou et al.
1998), and higher carotid artery intima-media
thickness (Tarim et al. 2006). These differences
are not fully explained by the higher BMI typical
of women with a history of GDM.
Studies of lipid profiles after pregnancies
complicated by preeclampsia are consistent with
increased atherogenesis risk, including consistently reported higher total cholesterol, LDL cholesterol, and triglycerides, although these
differences are not always statistically significant
(Fraser et al. 2012; Manten et al. 2007; Hubel
et al. 2008; Laivuori et al. 1996; Magnussen et al.
2009; Romundstad et al. 2010; Sattar et al. 2003;
Smith et al. 2009). Associations of reduced HDL
cholesterol after preeclampsia have been reported
by some (Fraser et al. 2012; Magnussen et al.
2009; Romundstad et al. 2010; Smith et al. 2009)
but not all (Manten et al. 2007; Laivuori et al.
1996; Sattar et al. 2003) studies. One study has
reported dyslipidemia among women with a history of spontaneous and indicated preterm births
(Catov et al. 2011). Some (Lauenborg et al. 2005;
Meyers-Seifer and Vohr 1996; Verma et al. 2002;
Di Cianni et al. 2007) but not all (Schwarz et al.
2009) have reported elevated total cholesterol,
LDL cholesterol, and/or triglycerides in women
with a history of GDM. As with the studies of
lipid concentrations before and during pregnancy,
studies of lipid concentrations in women in
the years after FGR are conflicting, with some
reporting hyperlipidemia (Kanagalingam et al.
2009) and others reporting no differences compared to women with uncomplicated births
(Catov et al. 2011).
It is now firmly established that women with
a history of GDM have a manifold higher risk of
developing type 2 diabetes than women with
normoglycemic pregnancies (Bellamy et al.
2009). It is less widely appreciated that women
with a history of preeclampsia are also at high
risk of type 2 diabetes. After preeclampsia,
mothers are three times more likely to develop
A. Fraser et al.
diabetes within 16 years (Lykke et al. 2009), an
observation bolstered by evidence of dysregulated glucose and insulin, as well as insulin resistance as early as 2 years after preeclamptic
pregnancy (Fraser et al. 2012; Manten et al.
2007; Laivuori et al. 1996; Magnussen et al.
2009; Smith et al. 2009; Wolf et al. 2004).
However, not all pregnancy complications are
associated with risk of future metabolic disorder:
in the Nurses’ Health Study 2, although 2% of
women who delivered a very preterm infant (<32
weeks’ gestation) had a 35% higher risk of developing type 2 diabetes, moderate preterm delivery
was not associated with increased diabetes risk
(James-Todd et al. 2010).
After pregnancy, plasma C-reactive protein is
elevated among women with prior eclampsia and
indicated preterm births suggesting that systemic
low-grade inflammation may link some adverse
pregnancy outcomes and later CVD (Hastie et al.
2011b; Hubel et al. 2006). Several studies have
documented higher C-reactive protein levels
among women with a history of GDM (Heitritter
et al. 2005; Di Cianni et al. 2007; Winzer et al.
2004). Although inflammation seems a likely
culprit to explain the association of spontaneous
preterm delivery with CVD risk, the only study to
date that has examined this question has reported
no differences in plasma C-reactive protein levels
of women with a history of spontaneous preterm
delivery compared to term delivery (Hastie et al.
2011b).
Women with a history of pregnancies complicated by preeclampsia may maintain a procoagulation state in the years after pregnancy,
predisposing them to vascular and thrombotic
events (Portelinha et al. 2009), although this
pathway is less studied than others linking pregnancy complications to maternal CVD risk.
Thus, the associations of pregnancy complications with future CVD events in women are likely
explained, at least in part, by their associations
with classic CVD risk factors of hypertension,
dyslipidemia, type 2 diabetes, and perhaps
inflammation and thrombosis, which are evident
before, during, and after such complicated pregnancies. Pregnancy provides a challenge to women’s cardiovascular system, and pregnancy
Pregnancy Characteristics and Women’s Cardiovascular Health
complications may serve as precursors, i.e., early
indications that a woman is on a high-CVD risk
trajectory, before these classic CVD risk factors
are clinically detected. That said, this trajectory
is complex and likely to be nonlinear. Given that
most pregnancies occur in early adulthood, long
before most CVD events occur, there is ample
opportunity to influence that trajectory by recognizing that health develops continuously over the
lifespan and that health development is a process
resulting from the ongoing interactions between
person and environment.
4
Recommendations
for Future Research
4.1
Major Themes and Findings
The associations of pregnancy complications
with CVD events are remarkably consistent.
Several pregnancy complications are more
common among racial minority groups, who are
also at higher risk of metabolic and cardiovascular disease. Although untested, the use of pregnancy complication history to screen women for
targeted CVD prevention has potential to improve
public health, given the magnitude of the associations, the prevalence of the pregnancy complications, and the importance of CVD in women.
Pregnancy complications occur early enough in a
woman’s life course to offer a significant meaningful “runway” for primordial CVD prevention
by lifestyle intervention and primary prevention
by statins and antihypertensive drugs. In 2011,
both the American Heart Association and the
European Society of Cardiology included histories of preeclampsia and (in the case of the
American Heart Association) GDM as part of
CVD risk assessment that would trigger closer
monitoring and control of CVD risk factors
(Mosca et al. 2011; Regitz-Zagrosek et al. 2011).
Pregnancy appears to be a critical transition
period for a woman that stresses her cardiovascular system in ways that may shed light on future
disease risk.
4.2
157
Research Priorities
(a) Epidemiologic research
• Establishing whether pregnancy complications per se contribute to CVD risk
We need to establish whether pregnancy complications act as stress tests to unmask women
who are already at increased risk of CVD in
later life and/or whether (and which) pregnancy
complications have a causal, direct contribution
to a woman’s CVD risk. If pregnancy complications per se contribute to CVD risk independently of prepregnancy cardiovascular health,
prevention strategies and treatment of pregnancy complications could be important not
only for women and their offspring during the
pregnancy but also later in life.
(b) Mechanism research
• Identifying mechanistic pathways to pregnancy complications and CVD
Should epidemiologic evidence suggest that
pregnancy complications are causally associated
with greater future CVD risk, it will be important
to ascertain the underlying pathways responsible
for these effects in order to identify potential
treatment pathways. Gaining better understanding of the mechanisms underlying pregnancy
complications themselves will also be important
in informing prevention strategies.
(c) Translational research
• Improving risk stratification
Irrespective of whether pregnancy complications
causally contribute to future CVD risk, a key
question is the extent to which pregnancy history
can be used to improve CVD risk scoring systems
for women, such as the Framingham Risk Score.
At present, these scoring systems are of debatable
utility for women under age 70 (Greenland et al.
2010), and addition of pregnancy complications
to prediction at these relatively younger ages may
be particularly important. Several pregnancy
A. Fraser et al.
158
complications are more common among racial
minority groups, who are also at higher risk of
metabolic and cardiovascular disease. Pregnancy
history may be of particular importance in identifying risk in these groups.
• Establishing strategies to mitigate future CVD
risk in women with pregnancy complications
If pregnancy complications are useful for early
CVD risk prediction, the next question is whether
earlier risk identification—as early as at the time
of pregnancy—is a cost-effective way of reducing future risk. To do so it is important to examine whether women who experienced pregnancy
complications should undergo more intense or
earlier screening and monitoring in the postpartum years in order to determine whether they
cross thresholds for treatment (e.g., with statins)
earlier than women without pregnancy complications. We would also need to test the extent to
which different lifestyle or pharmacologic preventions are effective at preventing future CVD
in young or middle-aged women with a history of
pregnancy complications. Key to this is identifying stages in the lifespan when women are (or are
not) receptive to CVD prevention, including the
postpartum year.
4.3
Data and Methods
Development Priorities
(a) Importance of linking research across life
stages: need for extended follow-up of
women with known reproductive histories
The bulk of the research associating pregnancy
history to CVD risk is derived from the linkage
of large, often national, vital statistics registries
for birth, hospitalization, and mortality statistics. These exercises have yielded consistent
associations of pregnancy complications with
CVD risk. However, as most registries were
founded in the 1950s or 1960s, the longest-running has been able to follow women only into
the early postmenopausal years. The data on
GDM are further limited by the lack of consistent methods for screening and diagnosing
GDM. Further follow-up will determine the
extent to which the associations of pregnancy
complications are maintained into the age range
at which CVD events are most common in
women. In the meantime, the stratification of
risk by time since pregnancy is a helpful way to
examine the extent to which risk associated with
pregnancy complications changes over time
(Hastie et al. 2011a). Although relative risk of
CVD events may weaken with time, the absolute risks associated with a history of pregnancy
complications is likely to grow with time since
pregnancy, as women age. In addition, we
should incorporate pregnancy history data into
existing CVD cohorts with decades of followup. By illuminating the timing with which particular CVD risk factors emerge in the wake of
specific pregnancy complications, we may be
able to leverage the information contained by
pregnancy history to predict CVD risk earlier
than conventional risk screening protocols.
(b) Importance of linking research across life
stages: need for studies with established
CVD risk factors measured before, during,
and after pregnancy
To understand the trajectory of CVD risk and
the role of pregnancy complications in that trajectory, we need more studies to measure CVD
risk factors prior to conception, particularly as
evidence suggests that changes in blood pressure,
augmentation index, and pulse wave velocity
occur as early as 6 weeks’ gestation, indicating
that maternal adaptations occur very early in
gestation (Mahendru et al. 2014). This raises
the question of whether “booking” first trimester
measures that are available in several birth
cohorts is representative of prepregnancy values
and emphasizes the importance of ascertaining
cardiovascular health trajectories from pre- to
postpartum.
(c) Need for innovative analytical approaches to
improve causal inference
Methods for improving our understanding of
whether pregnancy complications are causally
related to later maternal health need to go beyond
Pregnancy Characteristics and Women’s Cardiovascular Health
conventional multivariable approaches in prospective cohorts. For example, if it is found that
genetic variants associated with high blood pressure and glucose intolerance/type 2 diabetes in
general populations of men and nonpregnant
women are also associated with HDP and GDM,
this would lend some support to the hypothesis of
a common etiology and pregnancy unmasking a
preexisting (genetic) risk. There is some evidence
that several type 2 diabetes mellitus variants from
genome-wide association studies show robust
associations with GDM (Karlsson et al. 2007;
Lauenborg et al. 2009; Cho et al. 2009; Kwak
et al. 2012).
Although it is not feasible to randomize
women to pregnancy complications, long-term
follow-up of women who have been in randomized controlled trials that have effectively prevented or treated the pregnancy complication will
also address some of the research questions
above. Lastly, experimental induction of pregnancy complications in animal models and following the mothers after delivery to examine
whether vascular damage was sustained or metabolic risk increased are important for examining
the question of a pregnancy causal effect
(Bytautiene et al. 2010). However, the generalizability of the animal models depends on the
fidelity with which the human pregnancy complications, such as preeclampsia, can be mimicked
in other species, where they may not occur
naturally.
4.4
Translational Priorities
We are just beginning to investigate the clinical
implications of this growing body of research.
First, we need to establish the role of pregnancy
complications in determining maternal chronic
disease risk. Independently of that, we also need
to determine our ability to change the health trajectories of women with histories of complicated
pregnancy. We will then have to consider the
many issues of integrating the findings into clinical and public health systems. Some potential
clinical implications include the need to link pre-
159
natal with primary care medical records, development of clinical screening strategies, prevention
and treatment protocols after pregnancy complications, and increasing awareness among clinicians of these associations that span typical
clinical silos between obstetrics and medicine
(Rich-Edwards et al. 2010).
5
Conclusions
The stress test of pregnancy provides glimpses
into the otherwise silent early adult years in
which health development and chronic disease
trajectories are set. Research to characterize the
ways in which pregnancy complications inform
us about subclinical and clinical vascular and
metabolic risk in the mother is in its infancy.
Future research will require large datasets that
have prospectively collected accurate data on
cardiovascular risk factors before, during, and
after pregnancy, into middle age and beyond,
when disease begins to emerge; data on pregnancy complications is also required. Only with
such detailed information can we determine the
extent to which specific pregnancy complications
are related to future CVD, over and above prepregnancy risk factors, and whether they add to
established risk factor scores calculated in middle age. With large birth cohorts increasingly recognizing the importance of long-term follow-up
of the mother as well as their infant, the potential
for this research is increasing. Ultimately, randomized controlled trials will be necessary to
establish whether pregnancy advice and/or continued monitoring and early treatment of women
identified as at risk during pregnancy is a costeffective way of reducing CVD risk in women.
Research in this area will require integration
across such diverse specialties including obstetrics, primary care, pediatrics, endocrinology, and
cardiology. This broader perspective may yield
novel insights into the determinants of pregnancy
outcomes and health development across the
lifespan, perhaps creating a large shift in the
ways in which we promote the health of women
and children.
160
References
Agatisa, P. K., Ness, R. B., Roberts, J. M., Costantino,
J. P., Kuller, L. H., & McLaughlin, M. K. (2004).
Impairment of endothelial function in women with a
history of preeclampsia: An indicator of cardiovascular risk. American Journal of Physiology – Heart and
Circulatory Physiology, 286, H1389–H1393.
Anastasiou, E., Lekakis, J. P., Alevizaki, M., et al. (1998).
Impaired endothelium-dependent vasodilatation in
women with previous gestational diabetes. Diabetes
Care, 21, 2111–2115.
Beck, S., Wojdyla, D., Say, L., et al. (2010). The worldwide incidence of preterm birth: A systematic review
of maternal mortality and morbidity. Bulletin of the
World Health Organization, 88, 31–38.
Bellamy, L., Casas, J. P., Hingorani, A. D., & Williams,
D. J. (2007). Pre-eclampsia and risk of cardiovascular
disease and cancer in later life: Systematic review and
meta-analysis. British Medical Journal, 335, 974.
Bellamy, L., Casas, J. P., Hingorani, A. D., & Williams, D.
(2009). Type 2 diabetes mellitus after gestational diabetes: A systematic review and meta-analysis. Lancet,
373, 1773–1779.
Berends, A. L., de Groot, C. J., Sijbrands, E. J., et al.
(2008). Shared constitutional risks for maternal
vascular-related pregnancy complications and future
cardiovascular disease. Hypertension, 51, 1034–1041.
Bonamy, A. K., Parikh, N. I., Cnattingius, S., Ludvigsson,
J. F., & Ingelsson, E. (2011). Birth characteristics
and subsequent risks of maternal cardiovascular
disease: Effects of gestational age and fetal growth.
Circulation, 124, 2839–2846.
Butte, N. F. (2000). Carbohydrate and lipid metabolism
in pregnancy: Normal compared with gestational
diabetes mellitus. The American Journal of Clinical
Nutrition, 71, 1256S–1261S.
Bytautiene, E., Lu, F., Tamayo, E. H., et al. (2010). Longterm maternal cardiovascular function in a mouse
model of sFlt-1-induced preeclampsia. American
Journal of Physiology. Heart and Circulatory
Physiology, 298, H189–H193.
Campbell, S., Griffin, D. R., Pearce, J. M., et al. (1983).
New doppler technique for assessing uteroplacental
blood flow. Lancet, 321, 675–677.
Carr, D. B., Utzschneider, K. M., Hull, R. L., et al. (2006).
Gestational diabetes mellitus increases the risk of cardiovascular disease in women with a family history of
type 2 diabetes. Diabetes Care, 29, 2078–2083.
Catov, J., Newman, A., Roberts, J., et al. (2007a).
Association between infant birth weight and maternal
cardiovascular risk factors in the health, aging, and
body composition study. Annals of Epidemiology, 17,
36–43.
Catov, J. M., Bodnar, L. M., Kip, K. E., et al. (2007b).
Early pregnancy lipid concentrations and spontaneous preterm birth. American Journal of Obstetrics and
Gynecology, 197, 610. e611–610. e617.
A. Fraser et al.
Catov, J. M., Bodnar, L. M., Ness, R. B., Barron, S. J.,
& Roberts, J. M. (2007c). Inflammation and dyslipidemia related to risk of spontaneous preterm
birth. American Journal of Epidemiology, 166,
1312–1319.
Catov, J. M., Nohr, E. A., Olsen, J., & Ness, R. B. (2008a).
Chronic hypertension related to risk for preterm and
term small-for-gestational-age births. Obstetrics and
Gynecology, 112, 290.
Catov, J., Bodnar, L., Hackney, D., Roberts, J., &
Simhan, S. (2008b). Activation of the fibrinolytic
cascade early in pregnancy among women with spontaneous preterm birth. Obstetrics and Gynecology,
112, 1116–1122.
Catov, J. M., Wu, C. S., Olsen, J., Sutton-Tyrrell, K., Li,
J., & Nohr, E. A. (2010a). Early or recurrent preterm
birth and maternal cardiovascular disease risk. Annals
of Epidemiology, 20, 604–609.
Catov, J. M., Ness, R. B., Wellons, M. F., Jacobs,
D. R., Roberts, J. M., & Gunderson, E. P. (2010b).
Prepregnancy lipids related to preterm birth risk:
The coronary artery risk development in young
adults study. Journal of Clinical Endocrinology and
Metabolism, 95, 2009–2028.
Catov, J. M., Dodge, R., Yamal, J. M., Roberts, J. M.,
Piller, L. B., & Ness, R. B. (2011). Prior preterm or
small-for-gestational-age birth related to maternal
metabolic syndrome. Obstetrics and Gynecology, 117,
225–232.
Catov, J. M., Dodge, R., Barinas-Mitchell, E., et al.
(2013). Prior preterm birth and maternal subclinical
cardiovascular disease 4 to 12 years after pregnancy.
Journal of Women’s Health, 22(2002), 835–843.
Cho, Y. M., Kim, T. H., Lim, S., et al. (2009). Type 2
diabetes-associated genetic variants discovered in the
recent genome-wide association studies are related to
gestational diabetes mellitus in the Korean population.
Diabetologia, 52, 253–261.
Clausen, T., Djurovic, S., & Henriksen, T. (2001).
Dyslipidemia in early second trimester is mainly a feature of women with early onset pre-eclampsia. BJOG,
108, 1081–1087.
Curry, A., Vogel, I., Drews, C., et al. (2007). Midpregnancy maternal plasma levels of interleukin 2, 6,
and 12, tumor necrosis factor-alpha, interferon-gamma,
and granulocyte-macrophage colony-stimulating factor
and spontaneous preterm delivery. Acta Obstetricia et
Gynecologica Scandinavica, 86, 1103–1110.
Davey Smith, G., Hart, C., Ferrell, C., et al. (1997). Birth
weight of offspring and mortality in the Renfrew and
Paisley study: Prospective observational study. British
Medical Journal, 315, 1189–1193.
Davey Smith, G., Harding, S., & Rosato, M. (2000a).
Relation between infants’ birth weight and mothers’
mortality: Prospective observational study. British
Medical Journal, 320, 839–840.
Davey Smith, G., Whitley, E., Gissler, M., & Hemminki,
E. (2000b). Birth dimensions of offspring, premature birth, and the mortality of mothers. Lancet, 356,
2066–2067.
Pregnancy Characteristics and Women’s Cardiovascular Health
Davey Smith, G., Sterne, J., Tynelius, P., Lawlor, D. A., &
Rasmussen, F. (2005). Birth weight of offspring and
subsequent cardiovascular mortality of the parents.
Epidemiology, 16, 563–569.
Davey Smith, G., Hypponen, E., Power, C., & Lawlor,
D. A. (2007). Offspring birth weight and parental
mortality: Prospective observational study and metaanalysis. American Journal of Epidemiology, 166,
160–169.
Dekker, J. M., & Schouten, E. G. (1993). Number of pregnancies and risk of cardiovascular disease. The New
England Journal of Medicine, 329, 1893–1894. author
reply 1894–1895.
Di Cianni, G., Lencioni, C., Volpe, L., et al. (2007).
C-reactive protein and metabolic syndrome in
women with previous gestational diabetes. Diabetes/
Metabolism Research and Reviews, 23, 135–140.
Ducey, J., Schulman, H., Farmakides, G., et al. (1987). A
classification of hypertension in pregnancy based on
Doppler velocimetry. American Journal of Obstetrics
and Gynecology, 157, 680–685.
Edison, R. J., Berg, K., Remaley, A., et al. (2007). Adverse
birth outcome among mothers with low serum cholesterol. Pediatrics, 120, 723–733.
Enquobahrie, D. A., Williams, M. A., Qiu, C., & Luthy,
D. A. (2005). Early pregnancy lipid concentrations
and the risk of gestational diabetes mellitus. Diabetes
Research and Clinical Practice, 70, 134–142.
Ernst, G. D. S., de Jonge, L. L., Hofman, A., et al. (2011).
C-reactive protein levels in early pregnancy, fetal
growth patterns, and the risk for neonatal complications: The Generation R Study. American Journal
of Obstetrics and Gynecology, 205, 132.e131–132.
e112.
Fraser, A., Nelson, S. M., Macdonald-Wallis, C., et al.
(2012). Associations of pregnancy complications with
calculated cardiovascular disease risk and cardiovascular risk factors in middle age/clinical perspective.
Circulation, 125, 1367–1380.
Freathy, R. M., Weedon, M. N., Bennett, A., et al. (2007).
Type 2 diabetes TCF7L2 risk genotypes alter birth
weight: A study of 24,053 individuals. American
Journal of Human Genetics, 80, 1150–1161.
Freeman, D. J., McManus, F., Brown, E. A., et al. (2004).
Short-and long-term changes in plasma inflammatory
markers associated with preeclampsia. Hypertension,
44, 708–714.
Friedlander, Y., Paltiel, O., Manor, O., et al. (2007).
Birthweight of offspring and mortality of parents:
The Jerusalem perinatal study cohort. Annals of
Epidemiology, 17, 914–922.
Funai, E. F., Friedlander, Y., Paltiel, O., et al. (2005). Longterm mortality after preeclampsia. Epidemiology, 16,
206–215.
Gammill, H. S., Powers, R. W., Clifton, R. G., et al.
(2010). Does C-reactive protein predict recurrent preeclampsia? Hypertension in Pregnancy, 29, 399–409.
Girardi, G. (2011). Role of tissue factor in pregnancy
complications: Crosstalk between coagulation and
161
inflammation. Thrombosis Research, 127(Suppl 3),
S43–S46.
Green, A. B. V., & Moser, K. (1988). Mortality in women
in relation to their childbearing history. BMJ (Clinical
research ed), 297, 391–395.
Greenland, P., Alpert, J. S., Beller, G. A., et al. (2010).
2010 ACCF/AHA guideline for assessment of
cardiovascular risk in asymptomatic adults: A report
of the American College of Cardiology Foundation/
American Heart Association Task Force on Practice
Guidelines. Circulation, 122, e584–e636.
Hackney, D. N., Catov, J. M., & Simhan, H. N. (2010).
Low concentrations of thrombin-inhibitor complexes
and the risk of preterm delivery. American Journal of
Obstetrics and Gynecology, 203, 184.e181–184.e186.
Hannaford, P., Ferry, S., & Hirsch, S. (1997).
Cardiovascular sequelae of toxaemia of pregnancy.
Heart, 77, 154–158.
Hastie, C. E., Smith, G. C., MacKay, D. F., & Pell, J. P.
(2011a). Maternal risk of ischaemic heart disease following elective and spontaneous pre-term delivery:
Retrospective cohort study of 750 350 singleton pregnancies. International Journal of Epidemiology, 40,
914–919.
Hastie, C. E., Smith, G. C. S., Mackay, D. F., & Pell,
J. P. (2011b). Association between preterm delivery
and subsequent C-reactive protein: A retrospective
cohort study. American Journal of Obstetrics and
Gynecology, 205, 556.e551–556.e554.
Hedderson, M. M., Ferrara, A., & Sacks, D. A. (2003).
Gestational diabetes mellitus and lesser degrees of
pregnancy hyperglycemia: Association with increased
risk of spontaneous preterm birth. Obstetrics and
Gynecology, 102, 850–856.
Heilmann, L., Rath, W., & Pollow, K. (2007). Hemostatic
abnormalities in patients with severe preeclampsia.
Clinical and Applied Thrombosis/Hemostasis, 13,
285–291.
Heitritter, S. M., Solomon, C. G., Mitchell, G. F.,
Skali-Ounis, N., & Seely, E. W. (2005). Subclinical
inflammation and vascular dysfunction in women
with previous gestational diabetes mellitus. Journal
of Clinical Endocrinology and Metabolism, 90,
3983–3988.
Herrera, E. (2000). Metabolic adaptations in pregnancy
and their implications for the availability of substrates
to the fetus. European Journal of Clinical Nutrition,
54, S47.
Hubel, C. A., McLaughlin, M. K., Evans, R. W., Hauth,
B. A., Sims, C. J., & Roberts, J. M. (1996). Fasting
serum triglycerides, free fatty acids, and malondialdehyde are increased in preeclampsia, are positively
correlated, and decrease within 48 hours post partum.
American Journal of Obstetrics and Gynecology, 174,
975–982.
Hubel, C., Shakir, Y., Gallaher, M., McLaughlin, M., &
Roberts, J. (1998a). Low-density lipoprotein particle
size decreases during normal pregnancy in association
with triglyceride increases. Journal of the Society for
Gynecologic Investigation, 5, 244–250.
162
Hubel, C. A., Lyall, F., Weissfeld, L., Gandley, R. E., &
Roberts, J. M. (1998b). Small low-density lipoproteins
and vascular cell adhesion molecule-1 are increased
in association with hyperlipidemia in preeclampsia.
Metabolism, 47, 1281–1288.
Hubel, C. A., Powers, R., Snaedal, S., et al. (2006).
C-reactive protein is increased 30 years after eclamptic pregnancy. J Soc Gynecol Invest, 13(2 Suppl),
292A.
Hubel, C. A., Powers, R. W., Snaedal, S., et al. (2008).
C-reactive protein is elevated 30 years after eclamptic
pregnancy. Hypertension, 51, 1499–1505.
Irgens, H. U., Reisaeter, L., Irgens, L. M., & Lie, R. T.
(2001). Long term mortality of mothers and fathers
after pre-eclampsia: Population based cohort study.
BMJ (Clinical research ed), 323, 1213–1217.
James-Todd, T. K. A., Hibert, E., Mason, S., Vadnais, M.,
Hu, F., & Rich-Edwards. J. (2010). Gestation length,
birth weight and subsequent risk of type 2 diabetes in
mothers. Oral presentation. Presented at American
Diabetes Association’s 70th Scientific Sessions, June
2010, Orlando, FL.
Jónsdóttir, L., Arngrimsson, R., Geirsson, R. T.,
Slgvaldason, H., & Slgfússon, N. (1995). Death rates
from ischemic heart disease in women with a history
of hypertension in pregnancy. Acta Obstetricia et
Gynecologica Scandinavica, 74, 772–776.
Kanagalingam, M. G., Nelson, S. M., Freeman, D. J.,
et al. (2009). Vascular dysfunction and alteration of
novel and classic cardiovascular risk factors in mothers of growth restricted offspring. Atherosclerosis,
205, 244–250.
Kelly, R., Holzman, C., Senagore, P., et al. (2009).
Placental vascular pathology findings and pathways to
preterm delivery. American Journal of Epidemiology,
170, 148–158.
Kestenbaum, B., Seliger, S. L., Easterling, T. R., et al.
(2003). Cardiovascular and thromboembolic events
following hypertensive pregnancy. American Journal
of Kidney Diseases, 42, 982–989.
Kim, C., Newton, K. M., & Knopp, R. H. (2002). Gestational
diabetes and the incidence of type 2 diabetes a systematic review. Diabetes Care, 25, 1862–1868.
Kwak, S. H., Kim, S. H., Cho, Y. M., et al. (2012). A
genome-wide association study of gestational diabetes
mellitus in Korean women. Diabetes, 61, 531–541.
Labarthe, D. R. (1999). Prevention of cardiovascular risk
factors in the first place. Preventive Medicine, 29,
S72–S78.
Laivuori, H., Tikkanen, M. J., & Ylikorkala, O. (1996).
Hyperinsulinemia 17 years after preeclamptic first
pregnancy. The Journal of Clinical Endocrinology and
Metabolism, 81, 2908–2911.
Lauenborg, J., Mathiesen, E., Hansen, T., et al. (2005).
The prevalence of the metabolic syndrome in a danish
population of women with previous gestational diabetes mellitus is three-fold higher than in the general
population. The Journal of Clinical Endocrinology
and Metabolism, 90, 4004–4010.
A. Fraser et al.
Lauenborg, J., Grarup, N., Damm, P., et al. (2009).
Common type 2 diabetes risk gene variants associate with gestational diabetes. The Journal of Clinical
Endocrinology and Metabolism, 94, 145–150.
Lawlor, D. A., Emberson, J. R., Ebrahim, S., et al. (2003).
Is the association between parity and coronary heart
disease due to biological effects of pregnancy or adverse
lifestyle risk factors associated with child-rearing?
Circulation, 107, 1260–1264.
Lawlor, D. A., Relton, C., Sattar, N., & Nelson, S. M.
(2012). Maternal adiposity—a determinant of perinatal and offspring outcomes? Nature Reviews
Endocrinology, 8, 679–688.
Lie, R. T., Wilcox, A. J., & Skjaerven, R. (2006). Maternal
and paternal influences on length of pregnancy.
Obstetrics and Gynecology, 107, 880–885.
Lin, Y.-S., Tang, C.-H., Yang, C.-Y. C., et al. (2011).
Effect of pre-eclampsia–eclampsia on major cardiovascular events among peripartum women in Taiwan.
The American Journal of Cardiology, 107, 325–330.
Lykke, J. A., Langhoff-Roos, J., Sibai, B. M., Funai, E. F.,
Triche, E. W., & Paidas, M. J. (2009). Hypertensive
pregnancy disorders and subsequent cardiovascular
morbidity and type 2 diabetes mellitus in the mother.
Hypertension, 53, 944–951.
Lykke, J. A., Langhoff-Roos, J., Lockwood, C. J.,
Triche, E. W., & Paidas, M. J. (2010a). Mortality of
mothers from cardiovascular and non-cardiovascular
causes following pregnancy complications in first
delivery. Paediatric and Perinatal Epidemiology,
24, 323–330.
Lykke, J., Paidas, M., Damm, P., Triche, E., Kuczynski,
E., & Langhoff-Roos, J. (2010b). Preterm delivery and risk of subsequent cardiovascular morbidity and type-II diabetes in the mother. BJOG: An
International Journal of Obstetrics & Gynaecology,
117, 274–281.
Lykke, J. A., Paidas, M. J., Triche, E. W., & LANGHOFFROOS, J. (2012). Fetal growth and later maternal death,
cardiovascular disease and diabetes. Acta Obstetricia
et Gynecologica Scandinavica, 91, 503–510.
Macdonald-Wallis, C., Lawlor, D. A., Fraser, A., May,
M., Nelson, S. M., & Tilling, K. (2012). Blood pressure change in normotensive, gestational hypertensive,
preeclamptic, and essential hypertensive pregnancies.
Hypertension, 59, 1241–1248.
Magnussen, E. B., Vatten, L. J., Lund-Nilsen, T. I.,
Salvesen, K. A., Smith, G. D., & Romundstad, P. R.
(2007). Prepregnancy cardiovascular risk factors as
predictors of pre-eclampsia: Population based cohort
study. BMJ (Clinical research ed), 335, 978.
Magnussen, E. B., Vatten, L. J., Smith, G. D., &
Romundstad, P. R. (2009). Hypertensive disorders
in pregnancy and subsequently measured cardiovascular risk factors. Obstetrics and Gynecology, 114,
961–970.
Magnussen, E. B., Vatten, L. J., Myklestad, K., Salvesen,
K. Å., & Romundstad, P. R. (2011). Cardiovascular
risk factors prior to conception and the length of
Pregnancy Characteristics and Women’s Cardiovascular Health
pregnancy: Population-based cohort study. American
Journal of Obstetrics and Gynecology, 204, 526.
e521–526.e528.
Mahendru, A. A., Everett, T. R., Wilkinson, I. B., Lees,
C. C., & McEniery, C. M. (2014). A longitudinal
study of maternal cardiovascular function from preconception to the postpartum period. Journal of
Hypertension, 32, 849–856.
Manten, G. T., Sikkema, M. J., Voorbij, H. A., Visser,
G. H., Bruinse, H. W., & Franx, A. (2007). Risk factors
for cardiovascular disease in women with a history of
pregnancy complicated by preeclampsia or intrauterine growth restriction. Hypertension in Pregnancy, 26,
39–50.
Martinez, G., Daniels, K., & Chandra, A. (2012). Fertility
of men and women aged 15–44 years in the United
States: National survey of family growth, 2006–2010.
Natl Health Stat Report, 12, 1–28.
Mathers, C., Boerma, J. T., Fat, D. M., & World Health
Organization. (2008). The global burden of disease :
2004 update. Geneva: World Health Organization.
McDonald, S. D., Malinowski, A., Zhou, Q., Yusuf, S.,
& Devereaux, P. J. (2008). Cardiovascular sequelae
of preeclampsia/eclampsia: A systematic review
and meta-analyses. American Heart Journal, 156,
918–930.
Metzger, B. E., Cho, N. H., Roston, S. M., & Radvany,
R. (1993). Prepregnancy weight and antepartum
insulin secretion predict glucose tolerance five years
after gestational diabetes mellitus. Diabetes Care,
16(1wik), 42–1605.
Meyers-Seifer, C. H., & Vohr, B. R. (1996). Lipid levels
in former gestational diabetic mothers. Diabetes Care,
19, 1351–1356.
MMWR. (1989). Chronic disease reports in the Morbidity
and Mortality Weekly Report (MMWR). MMWR
Morb Mortal Wkly Rep, 38, 1–8.
Monga, M., & Creasy, R. (1994). Cardiovascular and
renal adaptation to pregnancy. In R. Creasy, R. Resnik,
J. D. Iams, et al. (Eds.), Maternal-fetal medicine:
Principles and practice (pp. 758–767). Philadelphia:
WB Saunders.
Mongraw-Chaffin, M. L., Cirillo, P. M., & Cohn, B. A.
(2010). Preeclampsia and cardiovascular disease
death. Hypertension, 56, 166–171.
Mosca, L., Benjamin, E. J., Berra, K., et al. (2011).
Effectiveness-based guidelines for the prevention
of cardiovascular disease in women – 2011 update:
A guideline from the american heart association.
Circulation, 123, 1243–1262.
Nanda, S. S. M., Syngelaki, A., Akolekar, R., &
Nicolaides, K. H. (2011). Prediction of gestational
diabetes mellitus by maternal factors and biomarkers
at 11 to 13 weeks. Prenatal Diagnosis, 31, 135–141.
Nardi, O., Zureik, M., Courbon, D., Ducimetière, P.,
& Clavel-Chapelon, F. (2006). Preterm delivery of
a first child and subsequent mothers’ risk of ischaemic heart disease: A nested case–control study.
European Journal of Cardiovascular Prevention &
Rehabilitation, 13, 281–283.
163
Ness, R. B., Harris, T., Cobb, J., et al. (1993). Number of
pregnancies and the subsequent risk of cardiovascular
disease. The New England Journal of Medicine, 328,
1528–1533.
Ness, R. B., Cobb, J., Harm, T., & D’Agostino, R. B.
(1995). Does number of children increase the rate
of coronary heart disease in men? Epidemiology, 6,
442–445.
North, R. A., McCowan, L. M., Dekker, G. A., et al.
(2011). Clinical risk prediction for pre-eclampsia in
nulliparous women: Development of model in international prospective cohort. BMJ (Clinical research ed),
342, d1875.
O’Brien, T. E., Ray, J. G., & Chan, W. S. (2003). Maternal
body mass index and the risk of preeclampsia: A systematic overview. Epidemiology, 14, 368–374.
Oblast, T. I. (1999). Decline in deaths from heart disease
and stroke—United States, 1900–1999. Heart Disease
and Stroke, 63(1900), 593–597.
OECD Family D. (2014). Childlessness (SF 2.5) In:
Social Policy Division, Organisation for Economic
Co-operation and Development. Paris, France: 2010.
SF2.5 report on childlessness. www.oecd.org/social/
family/database. Accessed 24 June, 2013.
Ostlund, I., Haglund, B., & Hanson, U. (2004). Gestational
diabetes and preeclampsia. European Journal of
Obstetrics, Gynecology, and Reproductive Biology,
113, 12–16.
Parikh, N. I., Cnattingius, S., Dickman, P. W., Mittleman,
M. A., Ludvigsson, J. F., & Ingelsson, E. (2010).
Parity and risk of later-life maternal cardiovascular
disease. American Heart Journal, 159, 215–221.
e216.
Pell, J. P., Smith, G. C., & Walsh, D. (2004). Pregnancy
complications and subsequent maternal cerebrovascular events: A retrospective cohort study of 119,668
births. American Journal of Epidemiology, 159,
336–342.
Pitiphat, W., Gillman, M. W., Joshipura, K. J., Williams,
P. L., Douglass, C. W., & Rich-Edwards, J. W. (2005).
Plasma C-reactive protein in early pregnancy and preterm delivery. American Journal of Epidemiology,
162, 1108–1113.
Portelinha, A., Cerdeira, A. S., Belo, L., et al. (2009).
Haemostatic factors in women with history of
Preeclampsia. Thrombosis Research, 124, 52–56.
Qiu, C., Williams, M. A., Leisenring, W. M., et al. (2003).
Family history of hypertension and type 2 diabetes
in relation to preeclampsia risk. Hypertension, 41,
408–413.
Regitz-Zagrosek, V., Lundqvist, C. B., Borghi, C., et al.
(2011). ESC Guidelines on the management of cardiovascular diseases during pregnancy. The Task Force
on the Management of Cardiovascular Diseases during Pregnancy of the European Society of Cardiology
(ESC). European Heart Journal, 32, 3147–3197.
Retnakaran, R., & Shah, B. R. (2009a). Abnormal screening glucose challenge test in pregnancy and future risk
of diabetes in young women. Diabetic Medicine, 26,
474–477.
164
Retnakaran, R., & Shah, B. R. (2009b). Mild glucose
intolerance in pregnancy and risk of cardiovascular
disease: A population-based cohort study. Canadian
Medical Association Journal, 181, 371–376.
Retnakaran, R., Austin, P. C., & Shah, B. R. (2011). Effect
of subsequent pregnancies on the risk of developing
diabetes following a first pregnancy complicated
by gestational diabetes: A population-based study.
Diabetic Medicine, 28, 287–292.
Rich-Edwards, J. W. (2009). Reproductive health as a sentinel of chronic disease in women. Women’s Health
(London, England), 5, 101–105.
Rich-Edwards, J. W., McElrath, T. F., Karumanchi,
S. A., & Seely, E. W. (2010). Breathing life into
the lifecourse approach: Pregnancy history and cardiovascular disease in women. Hypertension, 56,
331–334.
Rich-Edwards, J. W. K. K., Wilcox, A., & Skjaerven, R.
(2012). Duration of first pregnancy predicts maternal
cardiovascular death, whether delivery was medically indicated or spontaneous. American Journal of
Epidemiology, 175(Suppl 11), S64.
Rich-Edwards, J. W., Fraser, A., Lawlor, D. A., & Catov,
J. M. (2014). Pregnancy characteristics and women’s
future cardiovascular health: An underused opportunity to improve women’s health? Epidemiologic
Reviews, 36, 57–70.
Riskin-Mashiah, S., Damti, A., Younes, G., & Auslender,
R. (2010). First trimester fasting hyperglycemia as a
predictor for the development of gestational diabetes
mellitus. European Journal of Obstetrics, Gynecology,
and Reproductive Biology, 152, 163–167.
Roberts, C. L., Algert, C. S., Morris, J. M., Ford, J. B.,
& Henderson-Smart, D. J. (2005). Hypertensive disorders in pregnancy: A population-based study. The
Medical Journal of Australia, 182, 332–335.
Romero, R., Espinoza, J., Gonçalves, L. F., Kusanovic,
J. P., Friel, L., & Hassan, S. (2007). The role of inflammation and infection in preterm birth. Seminars in
Reproductive Medicine, 25, 021–039.
Romundstad, P. R., Davey Smith, G., Nilsen, T. I., &
Vatten, L. J. (2007). Associations of prepregnancy
cardiovascular risk factors with the offspring’s birth
weight. American Journal of Epidemiology, 166,
1359–1364.
Romundstad, P. R., Magnussen, E. B., Smith, G. D., &
Vatten, L. J. (2010). Hypertension in pregnancy and
later cardiovascular risk: Common antecedents?
Circulation, 122, 579–584.
Sacks DA, C. W., Wolde-Tsadik, G., & Buchanan, T. A.
(2003). Fasting plasma glucose test at the first prenatal
visit as a screen for gestational diabetes. Obstetrics
and Gynecology, 101, 1197–1203.
Sarwar, N., Gao, P., Seshasai, S. R., et al. (2010). Diabetes
mellitus, fasting blood glucose concentration, and risk
of vascular disease: A collaborative meta-analysis of
102 prospective studies. Lancet, 375, 2215–2222.
Sattar, N. (2004). Do pregnancy complications and
CVD share common antecedents? Atherosclerosis
Supplements, 5, 3–7.
A. Fraser et al.
Sattar, N., & Greer, I. A. (2002). Pregnancy complications
and maternal cardiovascular risk: Opportunities for
intervention and screening? British Medical Journal,
325, 157–160.
Sattar, N., Bendomir, A., Berry, C., Shepherd, J., Greer, I. A.,
& Packard, C. J. (1997). Lipoprotein subfraction concentrations in preeclampsia: Pathogenic parallels to atherosclerosis. Obstetrics and Gynecology, 89, 403–408.
Sattar, N., Greer, I., Galloway, P., et al. (1999). Lipid and
lipoprotein concentrations in pregnancies complicated
by intrauterine growth restriction. Journal of Clinical
Endocrinology and Metabolism, 84, 128–130.
Sattar, N., Ramsay, J., Crawford, L., Cheyne, H., & Greer,
I. A. (2003). Classic and novel risk factor parameters in
women with a history of preeclampsia. Hypertension,
42, 39–42.
Savvidou, M. D., Hingorani, A. D., Tsikas, D., Frölich,
J. C., Vallance, P., & Nicolaides, K. H. (2003).
Endothelial dysfunction and raised plasma concentrations of asymmetric dimethylarginine in pregnant
women who subsequently develop pre-eclampsia. The
Lancet, 361, 1511–1517.
Savvidou, M. D., Kaihura, C., Anderson, J. M., &
Nicolaides, K. H. (2011). Maternal arterial stiffness
in women who subsequently develop pre-eclampsia.
PLoS ONE, 6, e18703.
Scarborough, P., & Weissberg, P. (2011). Trends in coronary heart disease, 1961–2011. London: British Heart
Foundation.
Schwarz, E. B., Ray, R. M., Stuebe, A. M., et al. (2009).
Duration of lactation and risk factors for maternal cardiovascular disease. Obstetrics and Gynecology, 113,
974–982.
Shaat, N. L. A., Karlsson, E., Ivarsson, S., Parikh, H.,
Berntorp, K., & Groop, L. (2007). A variant in the
transcription factor 7-like 2 (TCF7L2) gene is associated with an increased risk of gestational diabetes
mellitus. Diabetologia, 50, 972–979.
Shah, B. R., Retnakaran, R., & Booth, G. L. (2008).
Increased risk of cardiovascular disease in young
women following gestational diabetes mellitus.
Diabetes Care, 31, 1668–1669.
Shah, R. U., Klein, L., & Lloyd-Jones, D. M. (2009).
Heart failure in women: Epidemiology, biology and
treatment. Women’s Health (London, England), 5,
517–527.
Shaw, L. J., Bairey Merz, C. N., Pepine, C. J., et al.
(2006). Insights from the NHLBI-Sponsored Women’s
Ischemia Syndrome Evaluation (WISE) Study: Part I:
Gender differences in traditional and novel risk factors, symptom evaluation, and gender-optimized diagnostic strategies. Journal of the American College of
Cardiology, 47, S4–S20.
Shay, C. M., Ning, H., Allen, N. B., et al. (2012). Status
of cardiovascular health in US adults clinical perspective prevalence estimates from the National Health and
Nutrition Examination Surveys (NHANES) 2003–
2008. Circulation, 125, 45–56.
Skjaerven, R., Wilcox, A. J., Klungsoyr, K., et al. (2012).
Cardiovascular mortality after pre-eclampsia in one
Pregnancy Characteristics and Women’s Cardiovascular Health
child mothers: Prospective, population based cohort
study. BMJ (Clinical research ed), 345, e7677.
Smith, G. C., Pell, J. P., & Walsh, D. (2001). Pregnancy
complications and maternal risk of ischaemic heart
disease: A retrospective cohort study of 129,290
births. Lancet, 357, 2002–2006.
Smith, G. C. S., Shah, I., Pell, J. P., Crossley, J. A., &
Dobbie, R. (2007). Maternal obesity in early pregnancy and risk of spontaneous and elective preterm
deliveries: A retrospective cohort study. American
Journal of Public Health, 97, 157–162.
Smith, G. N., Walker, M. C., Liu, A., et al. (2009). A history of preeclampsia identifies women who have underlying cardiovascular risk factors. American Journal of
Obstetrics and Gynecology, 200(58), e51–e58.
Solomon, C. G., Willett, W. C., Carey, V. J., et al. (1997).
A prospective study of pregravid determinants of gestational diabetes mellitus. JAMA, 278, 1078–1083.
Steenland, K., Lally, C., & Thun, M. (1996). Parity and
coronary heart disease among women in the American
Cancer Society CPS II population. Epidemiology, 7,
641–643.
Stepan, H., Faber, R., & Walther, T. (1999). Expression
of low density lipoprotein receptor messenger ribonucleic acid in placentas from pregnancies with
intrauterine growth retardation. British Journal of
Obstetrics and Gynaecology, 106, 1221–1222.
Tarim, E., Yigit, F., Kilicdag, E., et al. (2006). Early onset
of subclinical atherosclerosis in women with gestational diabetes mellitus. Ultrasound in Obstetrics &
Gynecology, 27, 177–182.
United Nations DoEaSA, Population Division. (2009).
World fertility report. New York: United Nations
Publications.
Verma, A., Boney, C. M., Tucker, R., & Vohr, B. R.
(2002). Insulin resistance syndrome in women with
prior history of gestational diabetes mellitus. The
Journal of Clinical Endocrinology and Metabolism,
87, 3227–3235.
Wadsack, C., Tabano, S., Maier, A., et al. (2007).
Intrauterine growth restriction is associated with
alterations in placental lipoprotein receptors and
maternal lipoprotein composition. American Journal
of Physiology - Endocrinology and Metabolism, 292,
E476–E484.
165
Wallis, A. B., Saftlas, A. F., Hsia, J., & Atrash, H. K.
(2008). Secular trends in the rates of preeclampsia,
eclampsia, and gestational hypertension, United States,
1987–2004. American Journal of Hypertension, 21,
521–526.
Weintraub, W. S., Daniels, S. R., Burke, L. E., et al.
(2011). Value of primordial and primary prevention
for cardiovascular disease: A policy statement from
the American Heart Association. Circulation, 124,
967–990.
Wikström, A. K., Haglund, B., Olovsson, M., & Lindeberg,
S. N. (2005). The risk of maternal ischaemic heart disease after gestational hypertensive disease. BJOG: An
International Journal of Obstetrics & Gynaecology,
112, 1486–1491.
Wilcox, A. J., Skjaerven, R., & Lie, R. T. (2008). Familial
patterns of preterm delivery: Maternal and fetal
contributions. American Journal of Epidemiology,
167, 474–479.
Williams, M. A., Qiu, C., Muy-Rivera, M., Vadachkoria,
S., Song, T., & Luthy, D. A. (2004). Plasma adiponectin concentrations in early pregnancy and subsequent
risk of gestational diabetes mellitus. The Journal
of Clinical Endocrinology and Metabolism, 89,
2306–2311.
Wilson, B. J., Watson, M. S., Prescott, G. J., et al. (2003).
Hypertensive diseases of pregnancy and risk of hypertension and stroke in later life: Results from cohort
study. British Medical Journal, 326, 845.
Winzer, C., Wagner, O., Festa, A., et al. (2004). Plasma
adiponectin, insulin sensitivity, and subclinical inflammation in women with prior gestational diabetes mellitus. Diabetes Care, 27, 1721–1727.
Wolf, M., Kettyle, E., Sandler, L., Ecker, J. L., Roberts,
J., & Thadhani, R. (2001). Obesity and preeclampsia:
The potential role of inflammation. Obstetrics and
Gynecology, 98, 757–762.
Wolf, M., Hubel, C. A., Lam, C., et al. (2004).
Preeclampsia and future cardiovascular disease:
Potential role of altered angiogenesis and insulin
resistance. The Journal of Clinical Endocrinology and
Metabolism, 89, 6239–6243.
Yusuf, S., Reddy, S., Ôunpuu, S., & Anand, S. (2001).
Global burden of cardiovascular diseases. Circulation,
104, 2746–2753.
Open Access This chapter is licensed under the terms of the Creative Commons Attribution 4.0 International License
(http://creativecommons.org/licenses/by/4.0/), which permits use, sharing, adaptation, distribution and reproduction in
any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license and indicate if changes were made.
The images or other third party material in this chapter are included in the chapter’s Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the chapter’s Creative Commons license
and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain
permission directly from the copyright holder.
Part III
The Life Course Origins and Consequences
of Select Major Health Conditions
and Issues
Early in the Life Course: Time
for Obesity Prevention
Summer Sherburne Hawkins, Emily Oken,
and Matthew W. Gillman
1
Introduction
Although the dramatic rise in obesity in the USA
experienced in recent decades (Wang and
Beydoun 2007) may have now stabilized (Ogden
et al. 2014; Wen et al. 2012), the prevalence of
obesity remains high. The most recent nationally
representative data collected in 2012 showed that
17% of children ages 2–19 years were obese
(Ogden et al. 2014). From 2003 through 2010,
the prevalence of obesity in low-income children
aged 2–4 years remained at approximately 15%
(Pan et al. 2012), while one regional study suggested that from 2004 through 2008, obesity
prevalence started to drop among 0 to 6-year-olds
from non-low-income families (Wen et al. 2012).
The reasons for this potential slowing or slight
reversal of the trend among some subgroups in
the USA remain unknown. However, obesity has
not affected all segments of the population
equally (Wang and Beydoun 2007; Wang et al.
2011), and racial/ethnic differences in obesity are
already evident by the preschool years (Ogden
et al. 2014).
The short- and long-term consequences of
obesity include conditions involving nearly every
organ system, such as asthma, type 2 diabetes,
high blood pressure, cardiovascular disease,
depression, and orthopedic problems, starting in
childhood and escalating among adults (Han
et al. 2010; Lobstein et al. 2004). Children who
are obese are much more likely to be obese
adults, and obesity at any age is very difficult to
treat (Lobstein et al. 2004). Furthermore, the
population-level effects of obesity are substantial. Nationwide, 9.1% of annual medical spending is attributable to adult obesity, representing a
cost of $147 billion per year (Finkelstein et al.
2009). This combination of evidence on the epidemiology, health consequences, and public
health impact of obesity suggests that prevention
is essential.
2
S.S. Hawkins (*)
Boston College, Chestnut Hill, MA, USA
e-mail: summer.hawkins@bc.edu
E. Oken • M.W. Gillman
Harvard Medical School and Harvard Pilgrim Health
Care Institute, Boston, MA, USA
e-mail: emily_oken@harvardpilgrim.org;
matthew.gillman@nih.gov
Conceptual Framework
One of the reasons for the intractability of childhood obesity is the failure to take into account the
complexity and interconnectedness of contributing factors ranging from the social, built, and
economic environments to behavior, physiology,
and epigenetics. These factors may also interact
with each other creating a self-perpetuating cycle
© The Author(s) 2018
N. Halfon et al. (eds.), Handbook of Life Course Health Development,
DOI 10.1007/978-3-319-47143-3_9
169
S.S. Hawkins et al.
170
of obesity, as we illustrate using gestational
diabetes mellitus (Sect. 4.3; Gillman 2016).
Based on the life course health development
approach to chronic disease epidemiology, biological, behavioral, and psychosocial exposures
that occur at particular stages in the life course
may have differential and/or lasting effects on
later outcomes (Ben-Shlomo and Kuh 2002;
Halfon and Forrest 2017).
The effects of these exposures can be cumulative across an individual’s life as well as operate
across generations to influence future generation’s
risk for chronic disease. Factors can also interact
with each other over the life course and be more
or less important at particular stages. These periods of particular influence are often termed critical or sensitive (Ben-Shlomo and Kuh 2002).
Throughout the chapter, we demonstrate that multiple critical and sensitive periods for obesity risk
exist across the life course, although practically
they may be hard to identify. Given the plasticity
inherent in early human development, the preand perinatal periods may present the most important opportunity for critical or sensitive period
effects. Added to the fact that treatment of obesity
is impeded by cultural, behavioral, and physiological feedback loops, modifying environment,
behaviors, and physiology early in life is likely an
especially effective strategy for preventing obesity and its consequences.
Consistent with LCHD principles (see Halfon
and Forrest 2017), Glass and McAtee (2006) propose that the multilevel approach shown in Fig. 1
places obesity prevention in a complex system with
individual risk factors being influenced by multiple
“above-water” levels (families, neighborhood, policies) as well as by the interaction with biology and
“underwater” levels (genes, epigenetics, physiology) over time. This framework adds to a life
course health development approach by emphasizing feedback loops and cross-level influences, such
as gene-environment interactions, thus highlighting
the need for methodology that takes into account
these complex relationships to help identify the
important and modifiable levers of change (Huang
and Glass 2008).
We have organized this chapter to reflect the
current thinking on periods in the life course that
appear to be most important for the development
of obesity. We focus primarily on the early portion of the life course, but also discuss later periods that may be key time points for intervening.
These periods often represent times of active
growth or turning points when the primary
sources of influence change.
At each stage of the life course, we discuss
specific macro-level factors if they are directly
relevant to that time period. Most macro-level
factors, however, either indirectly or directly
influence all age groups, so we present them
together after reviewing each life course stage
individually. While this chapter does not represent a systematic review of the literature, we
present key examples at each stage of the life
course to illustrate important risk factors, mechanisms, and gaps in research. We conclude with
recommendations for future work on methodology, research in emerging areas, and implications
for practice and policy.
3
Measurement of Overweight
and Obesity
Body mass index (BMI; weight/height2) is the
most commonly used indirect measure of adiposity, or fatness, at the population level. The US
Centers for Disease Control and Prevention (CDC)
recommends monitoring growth in children aged
0–2 years using the World Health Organization
weight for recumbent length growth standards and
defines excess weight as at or above the 97.7th
percentile (Grummer-Strawn et al. 2010). The
CDC defines obesity in children aged 2–19 years
as a BMI at or above the 95th percentile for age
and sex, with overweight between the 85th and
95th percentiles, using the CDC sex-specific BMIfor-age growth charts from 2000 (Kuczmarski
et al. 2002; Ogden and Flegal 2010).
4
Prenatal Period
4.1
Birth Weight
Numerous studies have confirmed the associations of higher birth weight with risk of later obesity and type 2 diabetes mellitus; lower birth
Early in the Life Course: Time for Obesity Prevention
171
Fig. 1 Conceptual framework for etiology of childhood obesity (Reproduced from Glass and McAtee (2006))
weight is consistently associated with increased
risk for central adiposity and its metabolic correlates, including type 2 diabetes mellitus and cardiovascular disease (Parsons et al. 1999; Yu et al.
2011). Birth weight, however, is not an etiologic
factor in itself. Some have used birth weight as a
marker of in utero programming during sensitive
periods of development (Gillman 2005).
However, many factors could explain the relationship between birth weight and later obesity.
For example, maternal obesity, a risk factor for
higher birth weight, is also highly related to
childhood obesity (Perng et al. 2014). This association may be due to genomic inheritance and
shared postnatal environment of eating habits and
physical activity or inactivity in addition to in
utero effects (Gillman and Poston 2012).
However, even after controlling for maternal obesity, sociodemographic characteristics, and other
risk factors, the birth weight-obesity association
remains, raising the possibility of a lasting effect
of fetal programming on later health (Gillman
2004; Oken and Gillman 2003).
4.2
Maternal Prepregnancy
Obesity
Many women start their pregnancy already
overweight or obese. In 2009, a representative
survey from 20 states found that 21% of women
were obese prepregnancy and an additional
25% were overweight (Fisher et al. 2013).
Mothers who are overweight or obese prepregnancy are more likely to have infants with
higher birth weight and an increased risk for
being large-for-gestational age (LGA; >90th
percentile of weight-for-gestational age) or
macrosomic (birth weight > 4000 g) (Institute
of Medicine 2009).
S.S. Hawkins et al.
172
Women who are overweight or obese going
into pregnancy are at higher risk for developing
gestational diabetes mellitus (Torloni et al. 2009)
and more likely to gain excessive amounts of
weight during pregnancy (Battista et al. 2011;
Dalenius et al. 2012). Studies must take into
account these related factors to tease apart the
roles of potentially modifiable risk factors.
Furthermore, these risk factors may also be markers for shared genes and/or the postnatal environment (including lifestyle behaviors), which could
also influence children’s risk for obesity. This
issue of interrelated factors—which may serve as
mediators, moderators, or confounders of each
other—is applicable not just to the prenatal
period, but also to all stages of life course health
development.
4.3
Gestational Diabetes Mellitus
(GDM)
GDM is defined as diabetes first diagnosed in
pregnancy, typically between 24 and 28 weeks
gestation. Data on 59 million births from the
National Hospital Discharge Survey showed that
GDM increased from 1.9% to 4.2% from 1989
through 2004 (Getahun et al. 2008). Over this
time period, the relative increase in GDM was
94% for white women (from 2.2% to 4.2%), but
260% for black women (from 0.6% to 2.1%)
(Getahun et al. 2008).
GDM may be fueling the obesity epidemic
(Battista et al. 2011; Herring and Oken 2011).
Figure 2 shows the interrelationships of maternal,
fetal, and child factors across the life course that
may propagate the intergenerational transmission
of obesity and diabetes (Gillman 2016). Both
maternal prepregnancy obesity and excessive
gestational weight gain (GWG) independently
increase mothers’ risk for developing GDM. A
systematic review found that higher maternal
prepregnancy BMI was associated with an
increase in risk for GDM, such that overweight,
moderately obese, and morbidly obese women
were 2, 3, and 5.5 times more likely to develop
GDM compared to women with normal BMI
(Torloni et al. 2009). Excess GWG during preg-
nancy may also increase mothers’ risk for GDM,
independent of women’s prepregnancy weight
(Hedderson et al. 2010). Compared to women
with adequate levels of weight gain, women with
excessive GWG are also more likely to have
greater postpartum weight retention (Nehring
et al. 2011). All of these factors increase women’s risk for subsequent health problems. Women
with a history of GDM are more likely to develop
type 2 diabetes mellitus, particularly within the
first decade after delivery (Kim et al. 2002).
Developing GDM in one pregnancy increases
mothers’ risk for the recurrence of GDM in subsequent pregnancies (Kim et al. 2007). If women
do not lose the excess weight they gained before
they become pregnant again, this increased adiposity reinforces the cycle in the subsequent
pregnancy.
Prepregnancy obesity, excessive GWG, and
GDM are associated with higher fetal growth and
subsequent increases in children’s adiposity and
risk for obesity (Fig. 2) (Gillman 2016). Infants
born to mothers with prepregnancy obesity or
GDM are more likely to be of higher birth weight
or be macrosomic (Battista et al. 2011; Institute
of Medicine 2009), and trials of GDM treatment
show reductions in macrosomia (Crowther et al.
2005; Gillman et al. 2010; Landon et al. 2009). A
study found that children born to mothers with
GDM had higher levels of adiposity and insulin
secretion at age 5–10 years, independent of current BMI (Chandler-Laney et al. 2012). Boney
et al. (2005) found that children who were LGA
at birth and exposed to either GDM or maternal
obesity in utero were at an increased risk for
developing metabolic syndrome. The cycle continues when girls who are obese mature and have
children of their own.
Plausible developmental mechanisms exist for
the influence of GDM on childhood obesity.
Pancreatic β-cells normally increase their insulin
secretion to compensate for the insulin resistance
during pregnancy. Glucose intolerance results
when the β-cells are not able to respond to this
increasing demand (Battista et al. 2011). Since
glucose crosses the placenta but insulin does not,
the fetus is exposed to greater levels of glucose
(Freinkel 1980). The developing fetal pancreas
Early in the Life Course: Time for Obesity Prevention
173
Postpartum
Pregnancy
Prepregnancy
obesity
Fetus
Postpartum
weight
retention
Excess
gestational
weight gain
Mother
Later Life
Type 2
diabetes,
cardiovascular
disease,etc.
Gestational
diabetes
Altered fetal
growth &
metabolism
Child
Altered
body
composition
Child
obesity
Adult
obesity,
type 2
diabetes,
CVD,
etc.
Fig. 2 Intergenerational transmission of obesity and gestational diabetes mellitus (Reproduced from Gillman (2016))
responds to the additional glucose by producing
insulin, which can increase fetal adiposity
(Freinkel 1980). Current research on the mechanisms of GDM is focusing on the areas of genetics, glucose, amino acid, lipid transport, and
adipokines (Harlev and Wiznitzer 2010).
Methodological challenges abound in studying the association between GDM and later obesity. Kim et al. (2011) have argued that many
studies combine pregestational diabetes mellitus
and GDM into one category, examine only
exposed or high-risk infants without an appropriate control group, and fail to control for potential
confounding factors, most notably BMI. Their
systematic review identified 12 studies of children exposed prenatally to GDM (excluding pregestational diabetes), with resulting crude odds
ratios of 0.7–6.3 for offspring obesity. However,
in many studies, it is not possible to distinguish
women with GDM with true onset during pregnancy from undiagnosed pregestational diabetes
(Herring and Oken 2011; Kim et al. 2011). In the
two studies that adjusted for maternal prepreg-
nancy BMI, results attenuated and were no longer significant after the covariate was included
(Kim et al. 2011), which suggests that GDM may
be a marker for preexisting maternal factors.
Randomized controlled trials (RCTs) of risk
factor manipulation in pregnancy with childhood
follow-up are especially valuable because they
minimize the effect of confounding and thus are
the most direct assessment of in utero programming in humans. In a US-based RCT of the treatment for mild GDM on pregnancy-related
outcomes, Landon et al. (2009) found reduction
in LGA and macrosomia; however, no childhood
follow-up has been done. Gillman et al. (2010)
followed up a subset of children who participated
in an Australian-based multicenter RCT of treatment for mild GDM. Although the parent trial
showed halving of macrosomia at birth, there
were no differences in child BMI at age 4–5 years.
One possible explanation for the null result is that
offspring of mothers with GDM appear to lose
their excess fat in the first year of life and do not
again begin to diverge from their unexposed
S.S. Hawkins et al.
174
peers until school age; thus, continued follow-up
of these children may be needed to demonstrate a
longer-lasting effect of prenatal GDM treatment.
Furthermore, as this study included women with
mild GDM only, a more dramatic benefit of treatment may be experienced among offspring of
women with more extreme hyperglycemia
(Gillman et al. 2010).
Another method for minimizing confounding
is sibling-pair designs, which partially control for
differences in the pre- and postnatal environment
(Brion 2010). This method relies on discrepant in
utero exposure between siblings, such that the
mother experienced GDM during her pregnancy
with one sibling but not the other. Lawlor and
colleagues (2011b) conducted a sibling-pair
study of over 280,000 Swedish men through a
record linkage study of their military conscription medical examination with birth information.
They found that BMI at age 18 was higher in men
whose mothers had GDM during pregnancy than
those who did not; these differences were still
evident in the within-sibling analyses and independent of maternal BMI. The authors concluded
that GDM may influence later obesity risk
through intrauterine mechanisms (Lawlor et al.
2011b).
4.4
Gestational Weight Gain
(GWG)
In 1990, the Institute of Medicine (IOM) (1990)
issued guidelines for the appropriate amount of
weight women should gain during pregnancy.
However, over the last two decades, excessive
GWG has been common and increasing. In 2010,
based on the Pregnancy Nutrition Surveillance
System (PNSS) from 29 states and the District of
Columbia, approximately 48% of low-income
women gained more weight than recommended
(Dalenius et al. 2012). Excessive weight gain is
associated with adverse infant outcomes, including macrosomia and LGA infants, as well as
postpartum weight retention in mothers (SiegaRiz et al. 2009). In 2009, the IOM published new
guidelines, which for the first time include a
weight gain range for obese women that recom-
mends lower gains than for women with lower
weight status (Institute of Medicine 2009).
GWG may also have a lasting effect on body
size across the life course through in utero conditioning. Although some evidence suggests that
extreme undernutrition during pregnancy may be
associated with higher obesity risk in offspring,
the more common occurrence is overnutrition
during pregnancy (Herring et al. 2012). Recent
systematic reviews found that excessive GWG
was associated with an increased risk for offspring obesity compared to adequate GWG (Lau
et al. 2014; Mamun Mamun et al. 2014; Nehring
et al. 2013), with an overall pooled odds ratio of
1.4 (Mamun et al. 2014; Nehring et al. 2013). For
example, Perng et al. (2014) found that every 5
kilograms of GWG was associated with greater
adiposity (measured total fat and trunk fat) and
higher leptin in children at ages 6–10 years, independent of maternal prepregnancy BMI.
GWG may influence childhood obesity
through several potential developmental pathways. Mothers who are more prone to gain
weight through genetic risk, poor diet, or other
behavioral factors may have children who are
also themselves exposed to these same risks.
However, associations still remain after adjustment for maternal and paternal BMI, reducing
some influence of shared genes and the postnatal environment. An alternative explanation
may be through fetal conditioning (Gillman
2005), similar to that proposed for GDM. In
animal models, overnutrition during pregnancy
has resulted in insulin resistance, increased adiposity, and hypertension in offspring (Alfaradhi
and Ozanne 2011).
One methodology to test causality is through
an intervention aimed at influencing maternal
weight gain during pregnancy through positive
lifestyle changes. Three recent meta-analyses
summarized the evidence on RCTs for prenatal
dietary, physical activity, and behavioral or lifestyle interventions on maternal weight gain and
infant outcomes (Agha et al. 2014; Oteng-Ntim
et al. 2012; Thangaratinam et al. 2012). The
reviews found that dietary and lifestyle interventions resulted in a small reduction in GWG
(1.42–2.21 kg), but together the interventions did
Early in the Life Course: Time for Obesity Prevention
not appear to influence birth weight or LGA
(Agha et al. 2014; Oteng-Ntim et al. 2012;
Thangaratinam et al. 2012). However, none of the
trials to date have looked at longer-term outcomes of child adiposity. Several ongoing RCTs
targeted at changing GWG will follow offspring
for development of obesity and its consequences
(Dodd et al. 2011; Moholdt et al. 2011; Vesco
et al. 2012). Vesco et al. (2014) found that obese
women randomized to a dietary intervention
gained less weight during pregnancy and had a
lower proportion of babies born LGA, with further follow-up planned.
Two separate sibling-pair analyses have examined GWG and offspring obesity. Lawlor and colleagues (2011a) examined the association of
maternal weight gain during pregnancy with obesity in men at age 18 years. They found that among
mothers with a normal BMI, there was no relationship between GWG and later obesity.
However, among mothers who were already overweight or obese, GWG was associated with an
increased risk for later obesity, even in the withinsibling analyses, suggesting an influence of the
intrauterine environment (Lawlor et al. 2011a).
Although Branum et al. (2011) found that women
with higher prepregnancy BMI and gestational
weight gain had children with a higher BMI at age
4, differences were no longer significant in the sibling fixed-effects analysis. The interaction
between maternal prepregnancy BMI and GWG
on childhood obesity risk is an area of active
investigation (Institute of Medicine 2009).
4.5
Maternal Smoking
During Pregnancy
In 2010, approximately 9% of US women smoked
during pregnancy, with white mothers and mothers with lower education more likely to smoke
during pregnancy (Hawkins and Baum 2014).
Many short- and long-term health effects of
smoking during pregnancy on mothers and
infants are well known, including lower fetal
growth (U.S. Department of Health and Human
Services 2004). It may seem paradoxical then
that numerous studies, as summarized in recent
175
meta-analyses, have shown that prenatal smoking
exposure is associated with an increased risk for
later obesity, even in studies that adjusted for
potential confounding factors (Oken et al. 2008;
Weng et al. 2012). Although mechanisms for the
relationship between maternal smoking during
pregnancy and childhood obesity are not fully
understood, animal studies have shown an association between prenatal nicotine exposure and
increased adiposity in offspring (Gao et al. 2005).
However, the extent to which this association,
seen mainly in observational studies, is causal
remains uncertain, especially since smoking is
so strongly socially patterned. Several
approaches have been used to minimize confounding, including accounting for paternal
smoking, a proxy for sociodemographic risk that
is likely to provide little direct exposure to the
fetus. One study with information on both maternal smoking during pregnancy and paternal
smoking status postpartum found that the association remained after adjustment for sociodemographic characteristics and paternal smoking
(von Kries et al. 2008), while in another study
the association was no longer evident after
adjustment (Fleten et al. 2012).
Others have used alternative study designs to
test whether the association may be causal.
Although RCTs have demonstrated that smoking
cessation during pregnancy reduces rates of low
birth weight and preterm birth (Lumley et al.
2009), none have examined later child obesity
risk. Two studies have used sibling-pair designs to
examine smoking during pregnancy (Gilman et al.
2008; Iliadou et al. 2010). Although both found a
relationship between smoking and later obesity,
the association was no longer evident after including the sibling fixed-effect. These results raise the
possibility that the association may be confounded
by unmeasured factors that are shared within families rather than being causal.
4.6
Hormonal Influences: Leptin
In 1973, Douglas L. Coleman published his seminal work in two papers, discussing an unknown
circulating factor responsible for the obese/diabetic
S.S. Hawkins et al.
176
state in the ob/ob mouse, later called leptin
(Grayson and Seeley 2012). Leptin is a hormone
primarily produced by adipose tissue known to be
responsible for the regulation of appetite, energy
expenditure, and neuroendocrine function
(Hauguel-de Mouzon et al. 2006). It is sometimes
called the satiety hormone because of its effects on
inducing a sense of fullness.
The developmental role of leptin in the perinatal period may differ from that later in the life
course. The placenta releases leptin into maternal
and fetal circulation, which may influence appetite and weight regulation. Maternal plasma
leptin concentrations during gestation are two
times higher than during non-gravid periods
(Hauguel-de Mouzon et al. 2006). Fetuses born
to obese mothers and mothers with GDM have
higher cord leptin levels than fetuses of lean
mothers or mothers without GDM, respectively
(Catalano et al. 2009; Okereke et al. 2002).
Umbilical cord leptin levels are positively associated with fetal fat mass, percent body fat, and
birth weight (Okereke et al. 2002).
Sensitive period(s) may exist for the role of
leptin in later obesity risk. A series of studies
from Project Viva, a US pre-birth cohort, has
examined relationships of leptin with early
growth. Lower cord blood leptin levels were
associated with smaller size at birth, but higher
weight gain from birth to 6 months and higher
BMI at age 3 years (Mantzoros et al. 2009; Parker
et al. 2011). In a follow-up study, Boeke et al.
(2013) found that lower leptin levels during pregnancy and in cord blood were associated with
more adiposity at age 3 years; however, higher
leptin levels at age 3 were associated with greater
weight gain and higher adiposity through age 7.
These findings were independent of maternal
BMI and birth weight. The authors concluded
there may be a sensitive period of leptin influence
during the prenatal period followed by accumulation of leptin tolerance during early childhood,
with different effects of leptin exposure by timing
(Boeke et al. 2013). These observations are consistent with animal studies showing conditioning
impact of heightened leptin sensitivity on later
obesity, with differences evident across the life
course (Bouret et al. 2004). There is great interest
in identifying the correct sensitive period(s) for
leptin to influence fetal development and reduce
later obesity risk. To date, however, little is
known about modifiable factors influencing fetal
leptin regulation in humans (Boeke et al. 2013).
5
Infancy
5.1
Rapid Weight Gain
Infants grow in both length and weight, and weight
changes include growth in both fat-free mass and
fat mass. Many studies have used change in weight
as a proxy for gain in adiposity, which may or may
not be a valid assumption. There are many different definitions of rapid growth, which are often
based on country- or population-specific references (Monteiro and Victora 2005).
Several meta-analyses have demonstrated that
infants who gain weight more quickly than average during the first 2 years of life are at higher
risk for later obesity (Baird et al. 2005; Monteiro
and Victora 2005; Weng et al. 2012). Taveras and
colleagues (2011c) examined the number of
major weight-for-length percentiles crossed on
the CDC 2000 growth chart during each of the
6-month periods from birth to 2 years with outcomes of obesity at ages 5 and 10 years. They
found that crossing upward 2 or more percentiles
in the first 2 years was associated with an
increased risk for obesity at both ages. The highest prevalence of later obesity was seen in children with the crossing upward of 2 or more
weight-for-length percentiles within the first
6 months of life (Taveras et al. 2011c).
Debate continues as to the exact time frame
that rapid weight gain matters the most for later
obesity risk. A major limitation of current
research is the lack of repeated measurements at
small intervals during infancy. Studies with
repeat detailed measurements often include
small, homogeneous samples of children, and
thus their generalizability is limited. An important area of future research is to more precisely
define the critical window for rapid gain in adiposity (or length) as well as modifiable determinants of rapid weight gain.
Early in the Life Course: Time for Obesity Prevention
Although based on the research we summarize here a potential conclusion might be to limit
rapid weight gain in infants to prevent later obesity, an important consideration is the potential
detrimental effects of restricting weight gain for
other organ systems. For infants born preterm,
more rapid postnatal weight gain has an important benefit for neurodevelopment and attained
size (Ehrenkranz et al. 2006) Although no association was seen between slower infant weight
gain and poorer neurodevelopmental outcomes in
a study of healthy, term infants (Belfort et al.
2008), this area requires more study to help determine optimal length and BMI patterns to promote
healthy neurodevelopment as well as reduce
childhood obesity.
One important modifiable determinant of the
rate of infant weight gain is infant feeding, both
the type and the approach. Although exclusively
breastfed infants gain weight more rapidly in the
early postnatal period, infants who are formula
fed or fed a combination of breast milk and formula gain BMI more rapidly during the latter
half of the first year of life (Kramer et al. 2004).
In an Australian RCT promoting positive feeding
practices, Mihrshahi et al. (2011) found that formula feeding and feeding on a schedule were
independently associated with rapid weight gain
between birth and age 4–7 months. Although the
association between breastfeeding and childhood obesity is discussed in the next section,
infant gain in length or adiposity and nutrition
are closely linked and, in fact, may be challenging to tease apart.
5.2
Breastfeeding
Despite the numerous demonstrated health benefits of breastfeeding (Section on Breastfeeding
2012), according to the CDC’s 2014 Breastfeeding
Report Card, only 79% of US mothers initiated
breastfeeding and only 49% breastfed for at least
6 months (Centers for Disease Control and
Prevention 2014). Moreover, racial/ethnic disparities in breastfeeding are substantial. Using
national data from 2004 to 2008, there was a 20%
point differential between black and white moth-
177
ers for both breastfeeding initiation (54% versus
74%) and breastfeeding for at least 6 months
(27% versus 43%) (Centers for Disease Control
and Prevention 2010).
Systematic reviews and meta-analyses of
observational studies have demonstrated an
inverse association of breastfeeding initiation or
duration with later obesity (Arenz et al. 2004;
Harder et al. 2005; Owen et al. 2005b). Two
reviews found that an inverse relationship still
remained after adjusting for confounding factors,
such as parental obesity or social class (Arenz
et al. 2004; Owen et al. 2005b). However, a third
showed attenuation of the association with BMI
to null after such adjustment (Owen et al. 2005a).
There was also evidence for a dose-response relationship, such that a longer duration of breastfeeding conferred a greater reduction in obesity
risk (Harder et al. 2005; Owen et al. 2005b). As
the majority of study samples were often homogeneous with children of mostly white European
descent, there is debate about the extent to which
these relationships apply to all racial/ethnic
groups (Gillman 2011; Harder et al. 2005).
Plausible mechanisms abound for the relationship between breastfeeding and childhood obesity (Bartok and Ventura 2009). Breastfed infants
may be better at self-regulation than bottle-fed
infants because they come off the breast when
they are full, which may help them learn to regulate energy intake (Li et al. 2010). Breast milk
contains hormones, such as leptin, involved in
regulating growth and development during
infancy (Savino and Liguori 2008). After the first
3 months of life, breastfed infants have slower
weight gain for the remainder of the year than
formula-fed infants (Savino and Liguori 2008).
Nevertheless, evidence is mounting to question
the extent to which this relationship is causal
(Gillman 2011). The majority of research has been
based on observational studies, and associations
may still be due to unmeasured factors. The few
studies using a sibling-pair design showed that
breastfeeding duration reduced risk for later obesity, but because mothers often breastfeed similarly across siblings, there are a limited number of
discordant pairs to detect effects (Gillman et al.
2006; Metzger and McDade 2010; Nelson et al.
S.S. Hawkins et al.
178
2005; O’Tierney et al. 2009). Furthermore, these
designs may not eliminate the role of infant growth
and behavior as a predictor—rather than result—
of breastfeeding intensity and duration.
Another methodological design to infer causality is to examine the association in a context
with different social gradients for both the exposure and outcome measures (Brion 2010; Gillman
2011). Brion et al. (2011) compared the association between breastfeeding duration and BMI
from two cohorts—in England where breastfeeding is associated with more advantaged social
circumstances and in Brazil where there is little
social patterning. There was an inverse association between breastfeeding duration and obesity
risk for children in England, but no such association for children in Brazil, from which the authors
concluded the effects are likely due to residual
confounding (Brion et al. 2011). Similarly, a
study in Hong Kong also found no association
between breastfeeding and childhood obesity,
where breastfeeding and obesity follow different
social patterns (Kwok et al. 2010).
The Promotion of Breastfeeding Intervention
Trial (PROBIT) in Belarus is a cluster-randomized
trial of breastfeeding promotion (Kramer et al.
2001). Mothers who started breastfeeding were
randomly assigned to an intervention group that
provided additional breastfeeding support or a
standard care control group. Although mothers in
the breastfeeding promotion group showed
increases in duration and exclusivity of breastfeeding, there was no evidence for a protective
effect of breastfeeding on skinfold thickness or
obesity in children at age 6.5 or 11 years (Kramer
et al. 2007; Martin et al. 2013). Based on the
existing evidence, breastfeeding may only confer
modest protection against obesity rather than
being a major determinant (Gillman 2011).
5.3
Disparities in Obesity Partially
Explained by Early-Life
Factors
Beyond breastfeeding, many other early-life obesogenic exposures may be socially patterned
(Dixon et al. 2012). Several investigators have
attempted to investigate whether observed racial/
ethnic disparities in obesity rates (Ogden et al.
2014) are related to early-life factors. Taveras
and colleagues (2010) have shown that compared
to white children, black and Hispanic children
were more likely to have risk factors for childhood obesity including excessive weight gain
during infancy and early introduction of solid
foods, while after 2 years, they were more likely
to have a television in their bedrooms, shorter
sleep duration, and higher intake of sugarsweetened beverages and fast food (Taveras et al.
2010). In contrast, they were less likely to be
exposed to factors associated with protection
against obesity, including exclusive breastfeeding (Taveras et al. 2010). The same investigators
more recently examined whether racial/ethnic
differences in childhood obesity at age 7 were
explained by these early risk factors (Taveras
et al. 2013). Although black and Hispanic children had higher BMI z-scores than white children, there were no longer differences in BMI
after adjustment for infancy- and childhoodrelated risk factors. The authors conclude that
racial/ethnic disparities in obesity may be determined by modifiable factors in early life (Taveras
et al. 2013).
6
Early to Mid-childhood
6.1
Family
Although family members have similar levels of
adiposity, meaning that children are more likely
to have a high BMI if their parents have a high
BMI (Patel et al. 2011), the influence of parents
on child adiposity almost certainly extends
beyond shared genes. Parents (including caregivers) also share a similar family, neighborhood,
and social environment. During the early years,
parents are the primary influence on children’s
dietary and physical activity/inactivity choices.
Parents not only physically provide children with
food and opportunities for physical activity, but
they also influence children’s preferences through
modeling or other experiences (Birch and
Davison 2001; Van Der Horst et al. 2007). In the
next two sections on early childhood to midchildhood and adolescence, we focus on “above-
Early in the Life Course: Time for Obesity Prevention
water” macro-level factors where there is
potential for population-level interventions and
public health impact.
6.2
Diet, Physical Activity,
and Inactivity
In the simplest terms, excessive weight gain
occurs when there is more “energy in” than
“energy out.” The dietary and physical activity
patterns of children and adolescents in the USA
have changed substantially over the last few
decades. Total energy intake and portion sizes
from energy-dense, nutrient-poor foods have
increased, and more meals are being eaten away
from home (Duffey and Popkin 2013; Piernas
and Popkin 2011). Among school-aged children,
daily calories from sugar-sweetened beverages
have increased from 130 to 212 kcal/day over the
last 20 years (Lasater et al. 2011). Although children and adolescents are recommended to participate in 60 min or more of physical activity
daily (U.S. Department of Health and Human
Services 2008), many children are not achieving
this goal. In 2013, nationwide, 15% of students
did not engage in at least 60 min of physical
activity on any of the prior 7 days (Kann et al.
2014). As children transition from childhood
through adolescence, physical activity levels
decrease (Kahn et al. 2008), and media exposure
and sedentary behaviors increase (Kann et al.
2014; Rideout and Hamel 2006). In recent
decades, screen time and media use have
increased. Currently, 41% of students play video or
computer games 3 or more hours per day, and an
additional 33% watch television 3 or more hours
per day (Kann et al. 2014). Even by age 1, children are using approximately 50 min of screen
media daily, which increases to nearly 2 hours by
age 4–6 years (Rideout and Hamel 2006).
As media use has increased, so has children’s
exposure to advertisements and food marketing.
Food advertising has been linked to influencing
children’s food preferences, purchasing requests,
and consumption patterns (McGinnis et al. 2006),
suggesting a potential mechanism by which television and media use may increase children’s risk for
179
obesity. US guidelines for the responsible advertising to children are voluntary self-regulatory initiatives, and the Rudd Center for Food Policy and
Obesity (2013) suggested that loopholes in industry pledges may provide for more public relations
benefits than health benefits.
6.3
Food Insecurity
In 2013, 14% of US households with children
were food insecure at least once during the year,
meaning they were unable to provide adequate
food for one or more household members due to
insufficient means (Coleman-Jensen et al. 2014).
Black and Hispanic households, low-income
households, and single-parent households with
children had rates of food insecurity higher than
the national average (Coleman-Jensen et al.
2014). Since children from lower-income families are at higher risk for obesity, the challenge
for many families is to dependably provide nutritious and high-quality food rather than obtaining
enough food (Ludwig et al. 2012). The
Supplemental Nutrition Assistance Program
(SNAP) has no regulations on the quality of
foods purchased (US Department of Agriculture,
2013b), and families may be spending their limited food budget on foods that are high in calories
but with low nutrient quality such as sugarsweetened beverages.
Three systematic reviews found that although
food insecurity was associated with an increased
risk for obesity in adults, particularly women, the
evidence was mixed for children (Dinour et al.
2007; Eisenmann et al. 2011; Franklin et al.
2012). A challenge for studies is identifying
whether the measure of food insecurity is at the
family or child level as parents may protect children from being food insecure. Echoing the conclusion from a review by Eisenmann et al. (2011),
even if the association is not causal, both food
insecurity and obesity exist in low-income
households. It is essential, therefore, to learn
more about purchasing patterns, diet, and physical activity in these families to learn why food
insecurity and obesity coexist and identify areas
for intervention.
S.S. Hawkins et al.
180
6.4
Child Care and School
Policies and programs in child care and school
can influence children’s dietary intake, physical
activity patterns, and risk for obesity.
Approximately 30% of children are in centerbased programs by age 2 and 40% of children by
age 3, spending nearly 30 hours in nonparental
care each week (National Center for Education
Statistics 2005). Thus, child care is a setting in
which obesogenic behaviors can be allowed or
prevented (Larson et al. 2011a). A review by
Larson and colleagues (2011b) identified 18 obesity prevention interventions that take place in
child care centers/preschools and 2 of the 5 interventions that used weight status as an outcome
found evidence for reduced weight.
In 2007, Benjamin et al. (2008) reviewed state
child care regulations for policies related to nutrition and physical activity that may contribute to
childhood obesity. They found that 80% of states
had regulations to ensure that water is freely
available in child care centers, 33% regulated
screen time, 18% had regulations for the provision of breast milk, 14% restricted sugarsweetened beverages, and only three states
required a specified number of minutes of physical activity. Four states had no policies related to
obesogenic practices in child care. The proportion of states with these regulations for family
child care homes—as opposed to centers—was
either the same or less. As of 2013, up to one
quarter of states did not comply with any of the
five recommendations from the IOM to promote
physical activity among infants in child care centers or family child care homes (Slining et al.
2014). Benjamin et al. (2009) showed that infants
who attended home-based child care early in life
had an increased weight for length at 1 year and
BMI at 3 years, suggesting that more research is
needed into the food and physical activity policies of family child care homes. Additional studies in Denmark (Neelon et al. 2015) and Finland
(Lehto et al. 2015) have demonstrated an association between starting child care before age 1 and
an increased risk for later obesity. In 2011, the
White House Task Force on Childhood Obesity
supported the development of new national stan-
dards for healthy eating, encouraging breastfeeding, promoting physical activity, and limiting
screen time in early-care settings (American
Academy of Pediatrics, American Public Health
Association, and National Resource Center for
Health and Safety in Child Care and Early
Education 2012).
Since children spend most of their time in
school and may eat two daily meals there, the
school environment can play an important role in
shaping health behaviors (Institute of Medicine
2007; Story et al. 2009). A systematic review and
meta-analysis by Waters et al. (2011) identified 55
interventions for preventing obesity in children
aged 0–18 years, with 5 of the 6 most promising
strategies related to the school environment:
school curricula that include healthy eating and
physical activity, increases in opportunities for
physical activity throughout the school week,
improvements in the nutritional quality of the food
served in schools, environments and cultural practices that support children eating healthier foods
and being active throughout the day, and support
for teachers and staff to implement health promotion strategies and activities. They also concluded
that interventions did not increase disparities.
There have been a number of reviews that examined the impact of school-based interventions
overall or school food and physical activity, separately, on obesity risk (Gonzalez-Suarez et al.
2009; Katz et al. 2008; Kropski et al. 2008).
Food and beverages available in schools are
either part of the federal school lunch and breakfast programs or are competitive foods sold
outside the federal programs. Meals served in the
National School Lunch Program and School
Breakfast Program must adhere to federally
defined nutrition standards in order for schools to
be eligible for federal subsidies (Institute of
Medicine 2007). In 2013, the US Department of
Agriculture issued the new “Smart Snacks in
Schools” nutrition standards for competitive
foods and beverages, which limit calories, fat,
sugar, and sodium (U.S. Department of
Agriculture 2013a). A study by Masse et al.
(2013) found that between 2003 and 2008, states
significantly strengthened their school nutritionrelated laws, particularly those related to the sale
Early in the Life Course: Time for Obesity Prevention
of competitive foods. Overall, laws for competitive food policies were stronger for elementary
schools than for middle and high schools. As of
2008, 7 states had no school nutrition laws across
the 16 categories they examined (Masse et al.
2013).
The National Association for Sport and
Physical Education (NASPE) sets standards for
physical education, including time allotment,
curriculum, and staffing (National Association
for Sport and Physical Education 2004). Perna
et al. (2012) found that public schools in states
with specific and stringent physical education
laws reported more weekly time for physical education: specifically, elementary schools reported
40 more minutes and middle schools reported 60
more minutes than schools within states with no
laws. There were no differences between high
schools. However, overall, only 8.5% of schools
fully met the NASPE guidelines for physical education time (Perna et al. 2012). Without federal
legislation for physical education, policies vary
widely by state and even by schools themselves.
7
Adolescence
7.1
Social Influences
Adolescence is a developmental period characterized by hormonal changes and a period of
rapid growth both physically and psychologically
(Viner et al. 2012). As children become more
independent, their sphere of influence moves
from parents to peers with increasing autonomy
in making choices. Although influences of dietary
and physical activity habits on obesity risk are
similar to those described during the early- to
mid-childhood periods, these habits are often
influenced by peer norms. Strong peer relationships are an important developmental change
during adolescence, and peer groups can influence health behaviors both positively and negatively (Viner et al. 2012).
Using social network theory and analysis,
Christakis and colleagues (2007) found that
weight gain in one person spread through social
ties and influenced risk of obesity in a friend, sib-
181
ling, or spouse among adults in the Framingham
Study. Trogdon et al. (2008) used the National
Longitudinal Study of Adolescent Health (Add
Health) to examine the influence of peer effects
on adolescents’ own BMI by constructing a
detailed definition of peer groups based on nominated friendship relationships. They found that
mean peer BMI was associated with adolescent
BMI, females were more sensitive to peer BMI
and overweight status than males, and peer
weight was more influential among adolescents
with the highest BMI (Trogdon et al. 2008).
Ali et al. (2011) also used data from the Add
Health study to understand potential mechanisms
of peer effects on obesity-related health behaviors. They found evidence for the influence of
peers on adolescents pursuing an active sport,
regular exercise, and eating in fast-food restaurants, but no consistent relationships with television viewing, short sleep duration, or other
dietary factors (Ali et al. 2011). Although additional research is needed to confirm these findings, the potential implications are that obesity
prevention programs aimed at adolescents will
need to consider the influence of peer groups.
8
Macro-level Factors
Although we have touched upon policies and
social issues that have their strongest influence in
particular age groups, this section presents
macro-level factors that either indirectly or
directly influence obesity risk across all stages in
life course health development.
8.1
Environment
The term “obesogenic environment” often refers
to features of the built and natural environments
that limit healthful behaviors related to eating,
sleeping, screen time, and physical activity. The
definition used to study the built environment
varies widely across studies (Dunton et al. 2009;
Lovasi et al. 2009; Papas et al. 2007). The most
common measures of access to food are distances
to the nearest fast-food restaurants or grocery
182
stores or density of food outlets within a defined
area. The most common measures of access to
physical activity resources are measured distance
to facilities for physical activity such as parks,
density of such facilities, walkability, or aspects
of community design. Related measures included
assessments of aesthetics or neighborhood safety.
Papas et al. (2007) found that 17 of the 20 studies
identified found a significant association between
some aspect of the built environment and risk for
obesity across the life course, with the majority
of studies looking at features of the environment
related to physical activity. Dunton et al. (2009)
examined aspects of the built environment related
to physical activity and childhood obesity. While
they found few consistent findings in children, in
adolescents obesity was associated with access to
equipment and facilities, neighborhood type
(urban/rural), and urban sprawl. Lovasi et al.
(2009) examined how the built environment may
be contributing to disparities in obesity and its
risk factors. They focused on studies that included
individuals who were poor or of low socioeconomic status, black, or Hispanic. The authors
found that lack of access to supermarkets, exercise facilities or places to be active, and safety
because of crime or traffic was all associated with
BMI or related behaviors, and they concluded
that prevention strategies should focus on these
areas (Lovasi et al. 2009).
There are many methodological challenges
to understanding the effects of the neighborhood environment on children because of the
diversity of populations, measures, and outcomes across studies (Dunton et al. 2009;
Lovasi et al. 2009; Papas et al. 2007). A further
limitation is that nearly all of the studies have
been cross-sectional, so the temporality of associations is not clear. Effects of the built environment on health vary across the life course
because exposure to and interaction with the
environment change by age. For example, distance to playgrounds matters more for children
than adolescents, whereas walkability may have
less impact on young children. While subjective
measures of the built environment are often
from parents or children themselves, more studies are using objective measures of the environ-
S.S. Hawkins et al.
ment, such as geographic information system
(GIS) mapping or even personal devices to
determine time, place, and activity simultaneously. The most important aspect is the definition of the built environment. Studies that
examine the same construct, such as walkability, may be using different definitions and measurements, which limit the ability to synthesize
the evidence. As Dunton et al. (2009) conclude,
it is imperative to identify “modifiable and specific” features of the built environment to inform
the development of interventions.
Neighborhoods vary in many ways, most of
which are unmeasured but likely interrelated;
observed associations that remain after adjustment are still subject to residual confounding.
RCTs, which balance both measured and unmeasured confounders, are uncommon in this area
primarily due to the feasibility of manipulating
aspects of the built environment. The Department
of Housing and Urban Development conducted a
unique social experiment from 1994 through
1998 to better understand the effects of neighborhood characteristics on health and social outcomes (Sanbonmatsu et al. 2011). Women with
children living in public housing in high-poverty
areas were randomly assigned to one of three
groups: experimental housing vouchers redeemable only if they moved to a low-poverty area,
unrestricted traditional vouchers, and a control
group that offered neither opportunity (Ludwig
et al. 2011). Ludwig et al. (2011) followed up the
participants from 2008 to 2010 and found that
women assigned to the experimental group had
lower levels of extreme obesity and diabetes than
women in the control group, although there were
no baseline measures of these outcomes.
However, among youth, there were no differences in physical health outcomes, including
obesity (Sanbonmatsu et al. 2011). The authors
concluded that although the mechanisms for
these reductions were unknown, the intervention
provides some causal evidence for the impact of
the neighborhood environment on health (Ludwig
et al. 2011).
A methodology for future research in this area
is quasi-experimental designs that evaluate a
clearly defined change—often a wide-ranging
Early in the Life Course: Time for Obesity Prevention
policy—that are referred to as natural experiments. Taking measurements of residents before
and after this change, or using secondary measures such as data from electronic medical
records, especially in comparison with an unexposed control group, can help assess the impact
of neighborhood factors on obesity itself or
related health behaviors. Such alternative study
designs are needed to identify important levers of
change to help inform the development of interventions conducted at the neighborhood or community levels.
8.2
Local and State
In recent years, city and state governments have
taken bold actions to change the environment and
encourage healthy choices. New York City
(NYC) has been a pioneer in this effort by using
a multiagency approach to tackle obesity and its
risk factors (Mello 2009). In 2006, NYC passed
the first regulation in the USA banning trans fat
use in restaurants. An evaluation comparing food
purchase data before and after the ban found a
significant decrease in the trans fat content of
purchases from fast-food chains after the law,
with benefits for patrons from both high- and
low-poverty neighborhoods (Angell et al. 2012).
In 2008, NYC required chain restaurants to post
calorie information prominently on menu boards
and menus. Although children and adolescents’
reported they noticed the posted information,
there were no differences in calories purchased
after versus before the introduction of calorie
postings (Elbel et al. 2011). NYC initiatives have
also extended to schools and the built environment (Office of the Mayor 2012). NYC established nutritional standards for every city agency
that purchases or serves meals to clients, including the 1.1 million students that attend city
schools, as well as standards for city vending
machines. The most controversial proposal was
to limit the size of sugar-sweetened beverages
sold in food service establishments to 16 ounces,
which caused much debate both for and against
this measure and was ultimately blocked
(Grynbaum 2012).
183
Although these policies in NYC provide
assessable natural experiments to allow for evaluation of specific environmental factors that might
promote weight gain (or loss), a challenge is
learning what may be the critical lever(s) for
change. When policies and programs are implemented simultaneously or within short time periods, it may not be possible to tease apart the
individual effects of each policy. Furthermore,
these efforts may not be generalizable; budgets
are often limited, and other cities or states may
not have the resources to implement the whole
suite of policies that have been enacted in NYC.
Nevertheless, these efforts provide valuable data
to clinicians and policy makers to help elucidate
what policies or programs will have the biggest
effect on obesity levels.
A relatively new area is using legislation to
financially penalize the purchase and consumption of obesogenic foods and beverages. One
approach draws from the lessons of tobacco control and the success of cigarette taxes. Taxes on
sugar-sweetened beverages are a prime example,
although taxes have also been proposed to reduce
the consumption of fast food (Powell et al. 2013).
The consumption of sugar-sweetened beverages
is strongly related to increased body weight (Te
Morenga et al. 2012). As of January 2014, 34
states and DC had a sales tax on regular soda sold
in food stores, with a mean tax rate of 5.2%
(Chriqui et al. 2014). A systematic review by
Powell et al. (2013) found that soda taxes had
little impact on weight outcomes, but the authors
argued that they were based on state-level sales
taxes that were relatively low and therefore
unlikely to effect substantial change in behavior.
Brownell et al. (2009) have proposed a
national excise tax on sugar-sweetened beverages
as a public health strategy to address the obesity
epidemic. They recommended implementing an
excise tax of 1 cent per ounce for beverages that
have any added caloric sweetener, which would
increase the cost of a 20-ounce soda by 15–20%
(Brownell et al. 2009). This type of tax, which is
similar to cigarette excise taxes, is preferable to a
sales tax because it provides an incentive to
reduce the amount of sugar per ounce of sugarsweetened beverage (Brownell et al. 2009). Since
S.S. Hawkins et al.
184
the cost would be passed onto retailers, they
would likely increase the retail price, and consumers would be aware of the cost when they are
deciding to purchase the product. Andreyeva
et al. (2011) estimated that a 1 cent per ounce tax
on sugar-sweetened beverages could reduce consumption by 24% and daily per capita caloric
intake from sugar-sweetened beverages from current levels of 190–200 calories to 145–150 calories. Brownell et al. (2009) suggest that the public
health impact could be greater for groups at
higher risk for obesity, such as children and lowincome groups, who consume greater amounts of
sugar-sweetened beverages.
An alternative approach to promoting a healthful diet is a subsidy to reduce price and encourage the consumption of healthful foods.
Unfortunately, in recent decades, just the opposite situation has occurred. The real inflationadjusted price of fruits and vegetables has
increased, while soda prices have declined and
fast-food prices have remained stable (Powell
et al. 2013). In the USA, subsidies on food have
been designed to alleviate food insecurity for
low-income families rather than increasing consumption of healthful foods by everyone. Powell
et al. (2013) found that lower fruit and vegetable
prices were associated with lower body weight
among low-income populations; however, cohort
studies are mixed when it comes to the effects of
fruits and vegetables on weight gain (Casazza
et al. 2013). Based on the long history of tobacco
control, the area of taxes and subsidies related to
obesity is still in its infancy.
8.3
ing healthy foods in schools; improving access to
healthy, affordable food; and getting children
more physically active. The strategy emphasized
that changes are needed at many macro levels to
promote healthy behaviors, including improvements to schools and the built environment. The
White House Task Force reported some progress
on many of these areas after just 1 year (White
House Task Force on Childhood Obesity 2011).
The IOM’s Standing Committee on Childhood
Obesity Prevention (Institute of Medicine 2011)
and the US Department of Health and Human
Services’ National Prevention Strategy (National
Prevention Council 2011) echo these priorities.
9
Recommendations
9.1
Major Themes
A multilevel approach for obesity prevention is
needed, which takes into account individual risk
factors that operate at multiple levels (“abovewater” and “underwater” influences) and recognizes that these factors also interact across the
life course (Fig. 1). We reviewed phases of life
course health development, prenatal through adolescence, that appear to be most important for the
development of obesity. We also presented alternative methodological approaches to observational studies that can help disentangle causal
associations. The following sections outline
research priorities and data/methods development priorities, and conclude with recommendations for practice and policy.
National
9.2
Reducing and preventing obesity is a federal priority. At a national level, the White House Task
Force on Childhood Obesity and the accompanying Let’s Move! campaign aim to reduce childhood obesity and raise a healthier generation
through an intra-agency collaboration (White
House Task Force on Childhood Obesity 2010).
The 70 recommendations were summarized into
five themes: getting children a healthy start on
life; empowering parents and caregivers; provid-
Research Priorities
Of the many emerging risk factors for obesity, we
briefly highlight sleep duration, endocrinedisrupting chemicals, epigenetic markers, and
microbial colonization.
• Sleep duration and quality: Disruption of sleep
may have adverse health consequences, including childhood obesity. Biologically plausible
mechanisms exist for why short sleep duration
Early in the Life Course: Time for Obesity Prevention
could increase children’s risk for obesity: tiredness and fatigue lead to reduced physical activity; hormone changes associated with insufficient
sleep, particularly lower leptin levels and higher
ghrelin levels, result in increased hunger and
eating; and with less sleep, there is more awake
time to eat (Taheri 2006). Although most of the
studies in this area have been cross-sectional,
prospective cohort studies do provide some evidence for causality. Chen et al. (2008) identified
17 studies from 1980 to 2007 on sleep duration
and adiposity in children, of which 3 were prospective cohort studies. Nielsen et al. (2011)
reviewed the literature from 2007 to 2009 and
identified an additional 13 studies in children.
Of the eight prospective cohort studies, all found
a significant inverse relationship between duration of sleep and adiposity (Nielsen et al. 2011).
Recent studies from Project Viva have shown
that chronic sleep curtailment from infancy was
associated with an increase in adiposity and
metabolic risk at age 7 (Cespedes et al. 2014;
Taveras et al. 2014).
• Early relational environment: Two aspects of
children’s early relational environment—the
quality of parental relationships and children’s
exposure to adverse experiences—have been
identified as potential risk factors for child and
adolescent obesity. The psychological and
physiological consequences of insecure attachments and trauma are well established, including poor emotional regulation and heightened
stress responses, which may directly lead to
obesity through biological changes (Glaser and
Kiecolt-Glaser 2005) or indirectly through
emotional eating (Michopoulos et al. 2015;
Torres and Nowson 2007) and sleep disturbances (Vgontzas et al. 2008). Studies in children and adolescents have found that those
with poor-quality maternal-child relationships
(Anderson et al. 2012) or insecure attachment
(Anderson and Whitaker 2011) in early childhood were more likely to be obese compared to
those with higher-quality relationships. Danese
and Tan (2014) conducted a meta-analysis of
41 studies and found that childhood maltreatment was associated with an increased risk of
obesity across the life course with an overall
pooled odds ratio of 1.36 and associations
185
remained after adjustment for socioeconomic
status and health behaviors. An additional
meta-analysis of 23 cohort studies found that
adults who reported physical, emotional, sexual, or general abuse during childhood were
more likely to be obese (pooled odds ratios of
1.28–1.45) and four studies reported doseresponse relationships (Hemmingsson et al.
2014). While this evidence suggests a role of
children’s early relational environment in the
development of obesity, further research is
needed to better understand the mechanisms to
help inform interventions.
• Endocrine-disrupting chemicals (EDCs): EDCs
are compounds that mimic or interfere with the
normal actions of endocrine hormones, including estrogens, androgens, and thyroid and pituitary hormones (Newbold et al. 2007). Although
some EDCs are naturally occurring, man-made
organic compounds pose greater risks to human
health and include flame retardants, bisphenol
A (BPA), pesticides, and polychlorinated
biphenyls (PCBs). EDCs that are lipophilic,
resistant to metabolism, and/or able to bioconcentrate up the food chain become stored in
body fats and are of particular concern (Elobeid
and Allison 2008). Thus, in utero exposure to
environmental chemicals, including EDCs,
during critical periods may play a role in the
development of obesity through fetal programming (Newbold et al. 2007). Only recently has
evidence been synthesized from animal models
and epidemiologic studies in humans to suggest a possible link between EDCs and later
obesity (Elobeid and Allison 2008). Trasande
et al. (2012) found that urinary BPA concentrations were associated with obesity in children
and adolescents, although alternative explanations cannot be ruled out because the study was
cross-sectional.
• Epigenetics: The notion that early environmental influences, such as maternal diet, or toxic
substances, like air pollution, alter offspring
outcomes through epigenetic changes that influence gene regulation could unite several strands
of human and animal observations about the
origins of obesity. Proof of principle emanates
from agouti mice (Waterland and Jirtle 2004).
The epigenetic mechanism associated with dif-
S.S. Hawkins et al.
186
ferences in body fat and cardiometabolic disease risk of the offspring involves switching off
the agouti gene by methyl groups from a dietary
supplement (Waterland and Jirtle 2004). These
and similar findings show, in principle, how
genetically identical individuals raised in similar postnatal environments can nonetheless
develop widely differing phenotypes.
• Gut microbiota: Infants get their first priming
dose of microbes in utero via the placenta, followed by a more thorough colonization as they
pass through the birth canal and are exposed to
the mother’s skin, so that by the time they are
a few days or weeks old, their gastrointestinal
tracts are colonized by a population of
microbes notable not only for their abundance
but also variety. Gut microbiota are likely to
affect many organs and systems, including the
regulation of energy balance and weight gain.
Although research on animal models suggests
a role of gut microbiota in the development of
obesity, studies in humans are limited (DiBaise
et al. 2008). One model system for evaluating
the role of gut microbiota is the route of delivery. Microbes from the mother and the environment colonize the infant’s intestinal tract
during delivery (Neu and Rushing 2011).
During cesarean delivery, the direct contact
with maternal vaginal and intestinal flora is
absent. The intestinal microbial composition
of infants via cesarean delivery resembles that
of the mother’s skin, whereas the intestinal
flora of infants born vaginally resembles the
mother’s vaginal flora and intestinal tract
(Dominguez-Bello et al. 2010). A systematic
review by Li et al. (2013) found that delivery
by cesarean section was associated with an
increased risk for overweight/obesity across
the life course compared to vaginal delivery,
with an overall pooled odds ratio of 1.33.
9.3
Data/Methods Development
Priorities
There are a number of major challenges to understanding causal influences on obesity risk
throughout the life course. Here, we discuss some
of these challenges as well as approaches to overcoming or minimizing them.
• One recurrent issue is the extent to which
associations described in observational studies are causal. One notable example is breastfeeding (Gillman 2011). Mothers who choose
to breastfeed often have substantially different
social and economic circumstances from
mothers who do not breastfeed their infants.
Furthermore, factors that predict successful
initiation and long-term maintenance of
breastfeeding, such as maternal obesity and
cesarean delivery, are themselves putative
causes of child obesity. Also, it is possible that
infant characteristics themselves predict
breastfeeding duration, since mothers may be
more likely to supplement faster growing
infants, who appear hungrier.
• In cross-sectional studies, both confounding
and reverse causation are important considerations. Longitudinal cohort studies with adjustment for multiple measured characteristics can
go part of the way in minimizing confounding,
but other approaches such as sibling-pair
design, maternal versus paternal effects,
cohorts with different confounding structures,
and RCTs can help control for unmeasured
confounding. Others have applied Mendelian
randomization, a method that takes advantage
of variation in genes of known function to
examine the causal effect of a modifiable exposure on disease in nonexperimental studies
(Smith and Ebrahim 2003). The genotype must
affect the disease status only via its effect on
the exposure of interest and should be randomly distributed with respect to other covariates (Ding and Hu 2008). This approach can
produce unbiased estimates of the effects of a
putative causal variable without a traditional
RCT. One example is the study of the maternal
and offspring FTO genotype and offspring
obesity risk, which suggests that maternal obesity affects childhood obesity only through
pathways other than the intrauterine environment (Lawlor et al. 2008). However, many
times these approaches are not feasible. For
example, data may not be readily available on
Early in the Life Course: Time for Obesity Prevention
large numbers of siblings with discordant
exposures; RCTs are costly and may not be
ethical; and the few common genes that influence obesity risk do not have strong associations and also may influence multiple metabolic
pathways.
• Another challenge is that of appropriate exposure assessment, which is relevant for multiple
factors of great interest including diet, physical activity, and the toxic, built, and social
environments. Multiple factors may interact
among each other. For example, breastfeeding
may particularly affect childhood obesity risk
only in the presence of an obesogenic environment, whereas two important null studies of
breastfeeding and obesity (Brion et al. 2011;
Kramer et al. 2007; Martin et al. 2013) were
both conducted in middle-income countries
with relatively low population obesity rates
(Belarus and Brazil).
• Current statistical techniques typically cannot
take into account the multitude of factors both
hierarchically and across the life course that
impact obesity. Traditional longitudinal analyses, even those that account for multiple levels
of influence, are often not powerful enough to
account for the complexity and interconnectedness of obesity. Systems science approaches
such as agent-based and system dynamics
modeling can include not only longitudinal and
multiple levels but also more complex features
of relationships like nonlinearity, path dependence, loops, and tipping (Hammond 2009;
Huang et al. 2009). These approaches have just
recently been imported into public health from
disciplines such as engineering (Mani et al.
2010) and evolutionary biology (Kitano 2002)
and may very well contribute to understanding
and ultimately solving childhood obesity.
9.4
Translational Priorities
The key to reducing childhood obesity is finding
the right level and time in the life course to intervene for the maximal effectiveness and efficiency. Many intervention studies early in the life
course are getting underway or are ongoing, and
will be invaluable in informing not only what fac-
187
tors to change but how to change them. Such
interventions may be complex and costly, but
ultimately may be what is required to reverse the
tide of obesity.
• Interventions to modify determinants of obesity through life course health development
may invoke multiple settings, e.g., medical
care, homes, child care, and school (Foster
et al. 2010; Taveras et al. 2011a, b, 2012);
involve multiple components, e.g., system
redesign and individual behavior change strategies including e-technology (Lubans et al.
2010; Taveras et al. 2012); and target single or
multiple factors (Dodd et al. 2011; Taveras
et al. 2011b; Vesco et al. 2012). Interventions
within medical care may be especially valuable during pregnancy and infancy, when individuals see their providers more often than
any other time during the life course.
• Some recommendations are already clear. For
prenatal factors, smoking avoidance has been
a priority for decades (U.S. Department of
Health and Human Services 2004). Guidelines
exist for appropriate GWG (Institute of
Medicine 2009). For mild-moderate GDM,
treatment with lifestyle and insulin reduces
neonatal complications (Crowther et al. 2005;
Landon et al. 2009); the protocol and criteria
for diagnosing GDM are undergoing new
scrutiny.
• Among early childhood factors, “feeding up”
small-for-gestational age infants should be
abandoned for most because such infants who
gain weight rapidly in infancy are at higher
risk of chronic disease and derive no neurocognitive benefits (Belfort and Gillman 2011).
The World Health Organization recommends
exclusive breastfeeding for 6 months, but in
the developed world, recommendations are
moving toward 4–6 months (Section on
Breastfeeding 2012); that interval appears to
be appropriate for introducing solid foods for
obesity prevention (Huh et al. 2011; Pearce
et al. 2013).
• Among dietary factors, evidence is strongest
for intake of sugary beverages at many ages.
Avoiding introducing these into infant diets
may be especially valuable, given most
188
humans’—especially
children’s—inherent
“sweet tooth” (Ventura and Mennella 2011).
• The American Academy of Pediatrics recommends zero screen time under age 2 years and
no more than 2 hours/day of screen time for
child age 2 years and older (Strasburger 2011).
Avoiding from the outset having a TV in the
room where a child sleeps appears to be key to
reducing screen time (Schmidt et al. 2012).
• Among the newer risk factors, improving
sleep duration and quality may be an especially effective maneuver—and preliminary
studies suggest it is feasible (Taveras et al.
2011a)—because all parents want more sleep!
Funding Funded by grants from NIH R00 HD068506
to Dr. Hawkins, K24 HD069408 and P30 DK092924 to
Dr. Oken, and K24 HL060841 to Dr. Gillman. The content
is solely the responsibility of the authors and does not necessarily represent the official views of the National
Institutes of Health.
References
Agha, M., Agha, R. A., & Sandell, J. (2014). Interventions
to reduce and prevent obesity in pre-conceptual and
pregnant women: A systematic review and metaanalysis. PloS One, 9(5), e95132.
Alfaradhi, M. Z., & Ozanne, S. E. (2011). Developmental
programming in response to maternal overnutrition.
Frontiers in Genetics, 2, 27.
Ali, M. M., Amialchuk, A., & Heiland, F. W. (2011).
Weight-related behavior among adolescents: The role
of peer effects. PloS One, 6(6), e21179.
American Academy of Pediatrics, American Public Health
Association, and National Resource Center for Health
and Safety in Child Care and Early Education. (2012).
Preventing childhood obesity in early care and education: Selected standards from caring for our children;
National health and safety performance standards;
Guidelines for early care and education programs,
3rd edition. http://nrckids.org/CFOC3/PDFVersion/
preventing_obesity.pdf. Accessed 8 Feb 2013.
Anderson, S. E., Gooze, R. A., Lemeshow, S., & Whitaker,
R. C. (2012). Quality of early maternal–child relationship and risk of adolescent obesity. Pediatrics, 129(1),
132–140.
Anderson, S. E., & Whitaker, R. C. (2011). Attachment
security and obesity in US preschool-aged children. Archives of Pediatrics & Adolescent Medicine,
165(3), 235–242.
S.S. Hawkins et al.
Andreyeva, T., Chaloupka, F. J., & Brownell, K. D.
(2011). Estimating the potential of taxes on sugarsweetened beverages to reduce consumption and generate revenue. Preventive Medicine, 52(6), 413–416.
Angell, S. Y., Cobb, L. K., Curtis, C. J., Konty, K. J., &
Silver, L. D. (2012). Change in trans fatty acid content of fast-food purchases associated with New York
City’s restaurant regulation: A pre-post study. Annals
of Internal Medicine, 157(2), 81–86.
Arenz, S., Ruckerl, R., Koletzko, B., & von Kries, R.
(2004). Breast-feeding and childhood obesity–a systematic review. International Journal of Obesity and
Related Metabolic Disorders, 28(10), 1247–1256.
Baird, J., Fisher, D., Lucas, P., Kleijnen, J., Roberts, H., &
Law, C. (2005). Being big or growing fast: Systematic
review of size and growth in infancy and later obesity.
BMJ, 331(7522), 929.
Bartok, C. J., & Ventura, A. K. (2009). Mechanisms
underlying the association between breastfeeding and
obesity. International Journal of Pediatric Obesity,
4(4), 196–204.
Battista, M. C., Hivert, M. F., Duval, K., & Baillargeon,
J. P. (2011). Intergenerational cycle of obesity and diabetes: How can we reduce the burdens of these conditions on the health of future generations? Experimental
Diabetes Research, 2011, 596060.
Belfort, M. B., & Gillman, M. W. (2011). Healthy
infant growth: What are the trade-offs in the developed world? In M. W. Gillman, P. D. Gluckman, &
R. G. Rosenfeld (eds): Recent Advances in Growth
Research: Nutritional, Molecular and Endocrine
Perspectives. Nestlé Nutr Inst Workshop Ser, vol 71,
pp 171–184
Belfort, M. B., Rifas-Shiman, S. L., Rich-Edwards, J. W.,
Kleinman, K. P., Oken, E., & Gillman, M. W. (2008).
Infant growth and child cognition at 3 years of age.
Pediatrics, 122(3), e689–e695.
Ben-Shlomo, Y., & Kuh, D. (2002). A life course approach
to chronic disease epidemiology: Conceptual models,
empirical challenges and interdisciplinary perspectives.
International Journal of Epidemiology, 31(2), 285–293.
Benjamin, S. E., Cradock, A., Walker, E. M., Slining, M.,
& Gillman, M. W. (2008). Obesity prevention in child
care: A review of U.S. state regulations. BMC Public
Health, 8, 188.
Benjamin, S. E., Rifas-Shiman, S. L., Taveras, E. M.,
Haines, J., Finkelstein, J., Kleinman, K., & Gillman,
M. W. (2009). Early child care and adiposity at ages 1
and 3 years. Pediatrics, 124(2), 555–562.
Birch, L. L., & Davison, K. K. (2001). Family environmental factors influencing the developing behavioral
controls of food intake and childhood overweight.
Pediatric Clinics of North America, 48(4), 893–907.
Boeke, C. E., Mantzoros, C. S., Hughes, M. D., L RifasShiman, S., Villamor, E., Zera, C. A., & Gillman,
M. W. (2013). Differential associations of leptin with
adiposity across early childhood. Obesity, 21(7),
1430–1437.
Early in the Life Course: Time for Obesity Prevention
Boney, C. M., Verma, A., Tucker, R., & Vohr, B. R. (2005).
Metabolic syndrome in childhood: Association with
birth weight, maternal obesity, and gestational diabetes mellitus. Pediatrics, 115(3), e290–e296.
Bouret, S. G., Draper, S. J., & Simerly, R. B. (2004).
Trophic action of leptin on hypothalamic neurons that
regulate feeding. Science, 304(5667), 108–110.
Branum, A. M., Parker, J. D., Keim, S. A., & Schempf,
A. H. (2011). Prepregnancy body mass index and
gestational weight gain in relation to child body
mass index among siblings. American Journal of
Epidemiology, 174(10), 1159–1165.
Brion, M. J. (2010). Commentary: Assessing the impact
of breastfeeding on child health: Where conventional
methods alone fall short for reliably establishing causal
inference. International Journal of Epidemiology,
39(1), 306–307.
Brion, M. J., Lawlor, D. A., Matijasevich, A., Horta, B.,
Anselmi, L., Araujo, C. L., Menezes, A. M., Victora,
C. G., & Smith, G. D. (2011). What are the causal
effects of breastfeeding on IQ, obesity and blood
pressure? Evidence from comparing high-income
with middle-income cohorts. International Journal of
Epidemiology, 40(3), 670–680.
Brownell, K. D., Farley, T., Willett, W. C., Popkin, B. M.,
Chaloupka, F. J., Thompson, J. W., & Ludwig, D. S.
(2009). The public health and economic benefits of
taxing sugar-sweetened beverages. The New England
Journal of Medicine, 361(16), 1599–1605.
Casazza, K., Fontaine, K. R., Astrup, A., Birch, L. L.,
Brown, A. W., Bohan Brown, M. M., Durant, N.,
Dutton, G., Foster, E. M., Heymsfield, S. B., McIver,
K., Mehta, T., Menachemi, N., Newby, P. K., Pate,
R., Rolls, B. J., Sen, B., Smith, D. L., Jr., Thomas,
D. M., & Allison, D. B. (2013). Myths, presumptions,
and facts about obesity. The New England Journal of
Medicine, 368(5), 446–454.
Catalano, P. M., Presley, L., Minium, J., & Hauguel-de
Mouzon, S. (2009). Fetuses of obese mothers develop
insulin resistance in utero. Diabetes Care, 32(6),
1076–1080.
Centers for Disease Control and Prevention. (2010).
Racial and ethnic differences in breastfeeding initiation and duration, by state – National Immunization
Survey, United States, 2004–2008. MMWR. Morbidity
and Mortality Weekly Report, 59(11), 327–334.
Centers for Disease Control and Prevention.
(2014).
Breastfeeding
report
card:
United
States, 2014. http://www.cdc.gov/breastfeeding/
pdf/2014breastfeedingreportcard.pdf. Accessed 2 Dec
2014.
Cespedes, E. M., Rifas-Shiman, S. L., Redline, S.,
Gillman, M. W., Pena, M. M., & Taveras, E. M.
(2014). Longitudinal associations of sleep curtailment
with metabolic risk in mid-childhood. Obesity, 22(12),
2586–2592.
Chandler-Laney, P. C., Bush, N. C., Granger, W. M.,
Rouse, D. J., Mancuso, M. S., & Gower, B. A. (2012).
Overweight status and intrauterine exposure to ges-
189
tational diabetes are associated with children’s metabolic health. Pediatr Obes, 7(1), 44–52.
Chen, X., Beydoun, M. A., & Wang, Y. (2008). Is sleep
duration associated with childhood obesity? A systematic review and meta-analysis. Obesity, 16(2), 265–274.
Chriqui, J. F., Eidson, S. S., & Chaloupka, F. J. (2014).
State sales taxes on regular soda (as of January
2014) – BTG Fact Sheet. http://www.bridgingthegapresearch.org/_asset/s2b5pb/BTG_soda_tax_fact_
sheet_April2014.pdf. Accessed 3 Dec 2014.
Christakis, N. A., & Fowler, J. H. (2007). The spread of
obesity in a large social network over 32 years. The
New England Journal of Medicine, 357(4), 370–379.
Coleman-Jensen, A., Gregory, C., & Singh, A. (September
2014). Household food security in the United States in
2013, ERR-173. Washington, DC: U.S. Department of
Agriculture, Economic Research Service.
Crowther, C. A., Hiller, J. E., Moss, J. R., McPhee, A. J.,
Jeffries, W. S., & Robinson, J. S. (2005). Effect of
treatment of gestational diabetes mellitus on pregnancy outcomes. The New England Journal of
Medicine, 352(24), 2477–2486.
Dalenius, K., Brindley, P., Smith, B. L., Reinold, C. M., &
Grummer-Strawn, L. (2012). Pregnancy nutrition surveillance 2010 report. Atlanta, GA: US Department
of health and human services; Centers for Disease
Control and Prevention.
Danese, A., & Tan, M. (2014). Childhood maltreatment
and obesity: Systematic review and meta-analysis.
Molecular Psychiatry, 19(5), 544–554.
DiBaise, J. K., Zhang, H., Crowell, M. D., KrajmalnikBrown, R., Decker, G. A., & Rittmann, B. E. (2008).
Gut microbiota and its possible relationship with obesity. Mayo Clinic Proceedings, 83(4), 460–469.
Ding, E. L., & Hu, F. B. (2008). Determining origins and
causes of childhood obesity via Mendelian randomization analysis. PLoS Medicine, 5(3), e65.
Dinour, L. M., Bergen, D., & Yeh, M. C. (2007). The food
insecurity-obesity paradox: A review of the literature
and the role food stamps may play. Journal of the
American Dietetic Association, 107(11), 1952–1961.
Dixon, B., Pena, M. M., & Taveras, E. M. (2012).
Lifecourse approach to racial/ethnic disparities in
childhood obesity. Advances in Nutrition, 3(1), 73–82.
Dodd, J. M., Turnbull, D. A., McPhee, A. J., Wittert, G.,
Crowther, C. A., & Robinson, J. S. (2011). Limiting
weight gain in overweight and obese women during
pregnancy to improve health outcomes: The LIMIT
randomised controlled trial. BMC Pregnancy and
Childbirth, 11, 79.
Dominguez-Bello, M. G., Costello, E. K., Contreras,
M., Magris, M., Hidalgo, G., Fierer, N., & Knight,
R. (2010). Delivery mode shapes the acquisition and
structure of the initial microbiota across multiple body
habitats in newborns. Proceedings of the National
Academy of Sciences of the United States of America,
107(26), 11971–11975.
Duffey, K. J., & Popkin, B. M. (2013). Causes of increased
energy intake among children in the U.S., 1977–2010.
190
American Journal of Preventive Medicine, 44(2),
e1–e8.
Dunton, G. F., Kaplan, J., Wolch, J., Jerrett, M., &
Reynolds, K. D. (2009). Physical environmental correlates of childhood obesity: A systematic review.
Obesity Reviews, 10(4), 393–402.
Ehrenkranz, R. A., Dusick, A. M., Vohr, B. R., Wright,
L. L., Wrage, L. A., & Poole, W. K. (2006). Growth in
the neonatal intensive care unit influences neurodevelopmental and growth outcomes of extremely low birth
weight infants. Pediatrics, 117(4), 1253–1261.
Eisenmann, J. C., Gundersen, C., Lohman, B. J., Garasky,
S., & Stewart, S. D. (2011). Is food insecurity related
to overweight and obesity in children and adolescents?
A summary of studies, 1995–2009. Obesity Reviews,
12(5), e73–e83.
Elbel, B., Gyamfi, J., & Kersh, R. (2011). Child and adolescent fast-food choice and the influence of calorie
labeling: A natural experiment. International Journal
of Obesity, 35(4), 493–500.
Elobeid, M. A., & Allison, D. B. (2008). Putative
environmental-endocrine disruptors and obesity: A
review. Current Opinion in Endocrinology, Diabetes,
and Obesity, 15(5), 403–408.
Finkelstein, E. A., Trogdon, J. G., Cohen, J. W., & Dietz,
W. (2009). Annual medical spending attributable to
obesity: Payer-and service-specific estimates. Health
Affairs, 28(5), 822–831.
Fisher, S. C., Kim, S. Y., Sharma, A. J., Rochat, R., &
Morrow, B. (2013). Is obesity still increasing among
pregnant women? Prepregnancy obesity trends in 20
states, 2003–2009. Preventive Medicine, 56(6), 372–378.
Fleten, C., Nystad, W., Stigum, H., Skjaerven, R.,
Lawlor, D. A., Davey Smith, G., & Naess, O. (2012).
Parent-offspring body mass index associations in the
Norwegian Mother and Child Cohort Study: A familybased approach to studying the role of the intrauterine environment in childhood adiposity. American
Journal of Epidemiology, 176(2), 83–92.
Foster, G. D., Linder, B., Baranowski, T., Cooper, D. M.,
Goldberg, L., Harrell, J. S., Kaufman, F., Marcus,
M. D., Treviño, R. P., & Hirst, K. (2010). A schoolbased intervention for diabetes risk reduction. The
New England Journal of Medicine, 363(5), 443–453.
Franklin, B., Jones, A., Love, D., Puckett, S., Macklin, J.,
& White-Means, S. (2012). Exploring mediators of
food insecurity and obesity: A review of recent literature. Journal of Community Health, 37(1), 253–264.
Freinkel, N. (1980). Banting lecture 1980. Of pregnancy
and progeny. Diabetes, 29(12), 1023–1035.
Gao, Y. J., Holloway, A. C., Zeng, Z. H., Lim, G. E.,
Petrik, J. J., Foster, W. G., & Lee, R. M. (2005).
Prenatal exposure to nicotine causes postnatal obesity and altered perivascular adipose tissue function.
Obesity Research, 13(4), 687–692.
Getahun, D., Nath, C., Ananth, C. V., Chavez, M. R.,
& Smulian, J. C. (2008). Gestational diabetes in the
United States: Temporal trends 1989 through 2004.
American Journal of Obstetrics and Gynecology,
198(5), e521–e525.
S.S. Hawkins et al.
Gillman, M. W. (2004). A life course approach to obesity.
In D. Kuh (Ed.), A life course approach to chronic disease epidemiology. Oxford: Oxford University Press.
Gillman, M. W. (2005). Developmental origins of health
and disease. The New England Journal of Medicine,
353(17), 1848–1850.
Gillman, M. W. (2011). Commentary: Breastfeeding and
obesity–the 2011 Scorecard. International Journal of
Epidemiology, 40(3), 681–684.
Gillman, M. W. (2016). Interrupting intergenerational
cycles of maternal obesity. In M. S. Fewtrell, F.
Haschke, & S. L. Prescott (eds): Preventive Aspects
of Early Nutrition. Nestlé Nutr Inst Workshop Ser, vol
85, pp 59–69.
Gillman, M. W., Oakey, H., Baghurst, P. A., Volkmer, R. E.,
Robinson, J. S., & Crowther, C. A. (2010). Effect of
treatment of gestational diabetes mellitus on obesity in
the next generation. Diabetes Care, 33(5), 964–968.
Gillman, M. W., & Poston, L. (2012). Maternal obesity.
Cambridge, UK: Cambridge University Press.
Gillman, M. W., Rifas-Shiman, S. L., Berkey, C. S.,
Frazier, A. L., Rockett, H. R., Camargo, C. A., Jr.,
Field, A. E., & Colditz, G. A. (2006). Breast-feeding
and overweight in adolescence: Within-family analysis [corrected]. Epidemiology, 17(1), 112–114.
Gilman, S. E., Gardener, H., & Buka, S. L. (2008). Maternal
smoking during pregnancy and children’s cognitive and
physical development: A causal risk factor? American
Journal of Epidemiology, 168(5), 522–531.
Glaser, R., & Kiecolt-Glaser, J. K. (2005). Stress-induced
immune dysfunction: Implications for health. Nature
Reviews Immunology, 5(3), 243–251.
Glass, T. A., & McAtee, M. J. (2006). Behavioral science
at the crossroads in public health: Extending horizons,
envisioning the future. Social Science & Medicine,
62(7), 1650–1671.
Gonzalez-Suarez, C., Worley, A., Grimmer-Somers, K.,
& Dones, V. (2009). School-based interventions on
childhood obesity: A meta-analysis. American Journal
of Preventive Medicine, 37(5), 418–427.
Grayson, B. E., & Seeley, R. J. (2012). Deconstructing
obesity: The face of fatness before and after the discovery of leptin. Diabetologia, 55(1), 3–6.
Grummer-Strawn, L. M., Reinold, C., & Krebs, N. F.
(2010). Use of World Health Organization and CDC
growth charts for children aged 0–59 months in the
United States. MMWR – Recommendations and
Reports, 59(RR-9), 1–15.
Grynbaum, M. M. (2012). Will soda restrictions help
New York win the war on obesity? BMJ, 345, e6768.
Halfon, N., & Forrest, C. B. (2017). The emerging theoretical framework of life course health development. In
N. Halfon, C. B. Forrest, R. M. Lerner, & E. Faustman
(Eds.), Handbook of life course health-development science. Cham: Springer.
Hammond, R. A. (2009). Complex systems modeling for
obesity research. Preventing Chronic Disease, 6(3),
A97.
Han, J. C., Lawlor, D. A., & Kimm, S. Y. (2010).
Childhood obesity. Lancet, 375(9727), 1737–1748.
Early in the Life Course: Time for Obesity Prevention
Harder, T., Bergmann, R., Kallischnigg, G., & Plagemann,
A. (2005). Duration of breastfeeding and risk of
overweight: A meta-analysis. American Journal of
Epidemiology, 162(5), 397–403.
Harlev, A., & Wiznitzer, A. (2010). New insights on
glucose pathophysiology in gestational diabetes and
insulin resistance. Current Diabetes Reports, 10(3),
242–247.
Hauguel-de Mouzon, S., Lepercq, J., & Catalano,
P. (2006). The known and unknown of leptin in
pregnancy. American Journal of Obstetrics and
Gynecology, 194(6), 1537–1545.
Hawkins, S. S., & Baum, C. F. (2014). Impact of state
cigarette taxes on disparities in maternal smoking during pregnancy. American Journal of Public Health,
104(8), 1464–1470.
Hedderson, M. M., Gunderson, E. P., & Ferrara, A.
(2010). Gestational weight gain and risk of gestational
diabetes mellitus. Obstetrics and Gynecology, 115(3),
597–604.
Hemmingsson, E., Johansson, K., & Reynisdottir, S.
(2014). Effects of childhood abuse on adult obesity:
A systematic review and meta-analysis. Obesity
Reviews, 15(11), 882–893.
Herring, S. J., & Oken, E. (2011). Obesity and diabetes
in mothers and their children: Can we stop the intergenerational cycle? Current Diabetes Reports, 11(1),
20–27.
Herring, S. J., Rose, M. Z., Skouteris, H., & Oken, E.
(2012). Optimizing weight gain in pregnancy to prevent obesity in women and children. Diabetes, Obesity
& Metabolism, 14(3), 195–203.
Huang, T. T., Drewnosksi, A., Kumanyika, S., & Glass,
T. A. (2009). A systems-oriented multilevel framework
for addressing obesity in the 21st century. Preventing
Chronic Disease, 6(3), A82.
Huang, T. T., & Glass, T. A. (2008). Transforming
research strategies for understanding and preventing
obesity. JAMA, 300(15), 1811–1813.
Huh, S. Y., Rifas-Shiman, S. L., Taveras, E. M., Oken, E.,
& Gillman, M. W. (2011). Timing of solid food introduction and risk of obesity in preschool-aged children.
Pediatrics, 127(3), e544–e551.
Iliadou, A. N., Koupil, I., Villamor, E., Altman, D.,
Hultman, C., Langstrom, N., & Cnattingius, S. (2010).
Familial factors confound the association between
maternal smoking during pregnancy and young
adult offspring overweight. International Journal of
Epidemiology, 39(5), 1193–1202.
Institute of Medicine. (1990). Nutrition during pregnancy. Part I, weight gain. Washington, DC: National
Academies Press.
Institute of Medicine. (2007). Nutrition standards for
foods in schools: Leading the way toward healtheir youth. Washington, DC: National Academies
Press.
Institute of Medicine. (2009). Weight gain during pregnancy: Reexamining the guidelines. Washington, DC:
National Academies Press.
191
Institute of Medicine. (2011). Early childhood obesity
prevention policies. Washington DC: National
Academies Press.
Kahn, J. A., Huang, B., Gillman, M. W., Field, A. E.,
Austin, S. B., Colditz, G. A., & Frazier, A. L. (2008).
Patterns and determinants of physical activity in U.S.
adolescents. The Journal of Adolescent Health, 42(4),
369–377.
Kann, L., Kinchen, S., Shanklin, S. L., Flint, K. H.,
Kawkins, J., Harris, W. A., Lowry, R., Olsen, E. O.,
McManus, T., Chyen, D., Whittle, L., Taylor, E.,
Demissie, Z., Brener, N., Thornton, J., Moore, J., Zaza,
S., & Centers for Disease Control and Prevention
(CDC). (2014). Youth risk behavior surveillance–
United States, 2013. MMWR Surveillance Summaries,
63(4), 1–168.
Katz, D. L., O’Connell, M., Njike, V. Y., Yeh, M. C., &
Nawaz, H. (2008). Strategies for the prevention and
control of obesity in the school setting: Systematic
review and meta-analysis. International Journal of
Obesity, 32(12), 1780–1789.
Kim, C., Berger, D. K., & Chamany, S. (2007). Recurrence
of gestational diabetes mellitus: A systematic review.
Diabetes Care, 30(5), 1314–1319.
Kim, C., Newton, K. M., & Knopp, R. H. (2002).
Gestational diabetes and the incidence of type 2 diabetes: A systematic review. Diabetes Care, 25(10),
1862–1868.
Kim, S. Y., England, J. L., Sharma, J. A., & Njoroge, T.
(2011). Gestational diabetes mellitus and risk of childhood overweight and obesity in offspring: A systematic review. Experimental Diabetes Research, 2011,
541308.
Kitano, H. (2002). Systems biology: A brief overview.
Science, 295(5560), 1662–1664.
Kramer, M. S., Chalmers, B., Hodnett, E. D., Sevkovskaya,
Z., Dzikovich, I., Shapiro, S., Collet, J. P., Vanilovich,
I., Mezen, I., Ducruet, T., Shishko, G., Zubovich,
V., Mknuik, D., Gluchanina, E., Dombrovsky, V.,
Ustinovitch, A., Ko, T., Bogdanovich, N., Ovchinikova,
L., & Helsing, E. (2001). Promotion of Breastfeeding
Intervention Trial (PROBIT): A randomized trial in
the Republic of Belarus. JAMA, 285(4), 413–420.
Kramer, M. S., Guo, T., Platt, R. W., Vanilovich, I.,
Sevkovskaya, Z., Dzikovich, I., Michaelsen, K. F.,
Dewey, K., & Promotion of Breastfeeding Interven
tion Trials Study Group. (2004). Feeding effects on
growth during infancy. The Journal of Pediatrics,
145(5), 600–605.
Kramer, M. S., Matush, L., Vanilovich, I., Platt, R. W.,
Bogdanovich, N., Sevkovskaya, Z., Dzikovich, I.,
Shishko, G., Collet, J. P., Martin, R. M., Davey
Smith, G., Gillman, M. W., Chalmers, B., Hodnett,
E., Shapiro, S., & PROBIT Study Group. (2007).
Effects of prolonged and exclusive breastfeeding on
child height, weight, adiposity, and blood pressure
at age 6.5 y: Evidence from a large randomized trial.
The American Journal of Clinical Nutrition, 86(6),
1717–1721.
192
Kropski, J. A., Keckley, P. H., & Jensen, G. L. (2008).
School-based obesity prevention programs: An
evidence-based review. Obesity, 16(5), 1009–1018.
Kuczmarski, R. J., Ogden, C. L., Guo, S. S., GrummerStrawn, L. M., Flegal, K. M., Mei, Z., Curtin, L. R.,
Roche, A. F., & Johnson, C. L. (2002). 2000 CDC
growth charts for the United States: Methods and development. Vital and Health Statistics, 11(246), 1–190.
Kwok, M. K., Schooling, C. M., Lam, T. H., & Leung,
G. M. (2010). Does breastfeeding protect against childhood overweight? Hong Kong’s ‘Children of 1997’
birth cohort. International Journal of Epidemiology,
39(1), 297–305.
Landon, M. B., Spong, C. Y., Thom, E., Carpenter, M. W.,
Ramin, S. M., Casey, B., Wapner, R. J., Varner, M. W.,
Rouse, D. J., Thorp, J. M., Jr., Sciscione, A., Catalano,
P., Harper, M., Saade, G., Lain, K. Y., Sorokin, Y.,
Peaceman, A. M., Tolosa, J. E., Anderson, G. B., &
Eunice Kennedy Shriver National Institute of Child
Health and Human Development Maternal-Fetal
Medicine Units Network. (2009). A multicenter, randomized trial of treatment for mild gestational diabetes. The New England Journal of Medicine, 361(14),
1339–1348.
Larson, N., Ward, D., Neelon, S. B., & Story, M. (2011a).
Preventing obesity among preschool children: How
can child-care settings promote healthy eating and
physical activity? Research synthesis. Princeton, NJ:
Robert Wood Johnson Foundation.
Larson, N., Ward, D. S., Neelon, S. B., & Story, M.
(2011b). What role can child-care settings play in
obesity prevention? A review of the evidence and call
for research efforts. Journal of the American Dietetic
Association, 111(9), 1343–1362.
Lasater, G., Piernas, C., & Popkin, B. M. (2011). Beverage
patterns and trends among school-aged children in the
US, 1989–2008. Nutrition Journal, 10, 103.
Lau, E. Y., Liu, J., Archer, E., McDonald, S. M., & Liu,
J. (2014). Maternal weight gain in pregnancy and
risk of obesity among offspring: A systematic review.
Journal of Obesity, 2014, 524939.
Lawlor, D. A., Lichtenstein, P., Fraser, A., & Langstrom,
N. (2011a). Does maternal weight gain in pregnancy
have long-term effects on offspring adiposity? A sibling study in a prospective cohort of 146,894 men from
136,050 families. The American Journal of Clinical
Nutrition, 94(1), 142–148.
Lawlor, D. A., Lichtenstein, P., & Langstrom, N. (2011b).
Association of maternal diabetes mellitus in pregnancy with offspring adiposity into early adulthood:
Sibling study in a prospective cohort of 280,866 men
from 248,293 families. Circulation, 123(3), 258–265.
Lawlor, D. A., Timpson, N. J., Harbord, R. M., Leary, S.,
Ness, A., McCarthy, M. I., Frayling, T. M., Hattersley,
A. T., & Smith, G. D. (2008). Exploring the developmental overnutrition hypothesis using parentaloffspring associations and FTO as an instrumental
variable. PLoS Medicine, 5(3), e33.
Lehto, R., Mäki, P., Ray, C., Laatikainen, T., & Roos,
E. (2015). Childcare use and overweight in Finland:
S.S. Hawkins et al.
Cross-sectional and retrospective associations among
3- and 5-year-old children. Pediatric Obesity, 11(2),
136–143. doi:10.1111/ijpo.12036.
Li, H. T., Zhou, Y. B., & Liu, J. M. (2013). The impact of
cesarean section on offspring overweight and obesity:
A systematic review and meta-analysis. International
Journal of Obesity, 37(7), 893–899.
Li, R., Fein, S. B., & Grummer-Strawn, L. M. (2010).
Do infants fed from bottles lack self-regulation of
milk intake compared with directly breastfed infants?
Pediatrics, 125(6), e1386–e1393.
Lobstein, T., Baur, L., & Uauy, R. (2004). Obesity in
children and young people: A crisis in public health.
Obesity Reviews, 5(1), 4–104.
Lovasi, G. S., Hutson, M. A., Guerra, M., & Neckerman,
K. M. (2009). Built environments and obesity in disadvantaged populations. Epidemiologic Reviews, 31,
7–20.
Lubans, D. R., Morgan, P. J., Dewar, D., Collins, C. E.,
Plotnikoff, R. C., Okely, A. D., Batterham, M. J.,
Finn, T., & Callister, R. (2010). The Nutrition and
Enjoyable Activity for Teen Girls (NEAT girls) randomized controlled trial for adolescent girls from
disadvantaged secondary schools: Rationale, study
protocol, and baseline results. BMC Public Health,
10, 652.
Ludwig, D. S., Blumenthal, S. J., & Willett, W. C. (2012).
Opportunities to reduce childhood hunger and obesity:
Restructuring the supplemental nutrition assistance
program (the food stamp program). JAMA, 308(24),
2567–2568.
Ludwig, J., Sanbonmatsu, L., Gennetian, L., Adam, E.,
Duncan, G. J., Katz, L. F., Kessler, R. C., Kling, J. R.,
Lindau, S. T., Whitaker, R. C., & McDade, T. W.
(2011). Neighborhoods, obesity, and diabetes–a randomized social experiment. The New England Journal
of Medicine, 365(16), 1509–1519.
Lumley, J., Chamberlain, C., Dowswell, T., Oliver, S.,
Oakley, L., & Watson, L. (2009). Interventions for promoting smoking cessation during pregnancy. Cochrane
Database of Systematic Reviews, 3, CD001055.
Mamun, A. A., Mannan, M., & Doi, S. A. (2014).
Gestational weight gain in relation to offspring obesity over the life course: A systematic review and
bias-adjusted meta-analysis. Obesity Reviews, 15(4),
338–347.
Mani, M., Mandre, S. B., & M. (2010). Events before
droplet splashing on a solid surface. Journal of Fluid
Mechanics, 647, 163.
Mantzoros, C. S., Rifas-Shiman, S. L., Williams, C. J.,
Fargnoli, J. L., Kelesidis, T., & Gillman, M. W. (2009).
Cord blood leptin and adiponectin as predictors of
adiposity in children at 3 years of age: A prospective
cohort study. Pediatrics, 123(2), 682–689.
Martin, R. M., Patel, R., Kramer, M. S., Guthrie, L.,
Vilchuck, K., Bogdanovich, N., Sergeichick, N.,
Gusina, N., Foo, Y., Palmer, T., Rifas-Shiman, S. L.,
Gillman, M. W., Smith, G. D., & Oken, E. (2013).
Effects of promoting longer-term and exclusive breastfeeding on adiposity and insulin-like growth factor-I
Early in the Life Course: Time for Obesity Prevention
at age 11.5 years: A randomized trial. JAMA, 309(10),
1005–1013.
Masse, L. C., Perna, F., Agurs-Collins, T., & Chriqui,
J. F. (2013). Change in school nutrition-related
laws from 2003 to 2008: Evidence from the school
nutrition-environment state policy classification system. American Journal of Public Health, 103(9),
1597–1603.
McGinnis, J. M., Gootman, J. A., & Kraak, V. I. (2006).
Food marketing to children and youth: Threat or
opportunity? Washington, DC: National Academies
Press.
Mello, M. M. (2009). New York City’s war on fat. The New
England Journal of Medicine, 360(19), 2015–2020.
Metzger, M. W., & McDade, T. W. (2010). Breastfeeding
as obesity prevention in the United States: A sibling difference model. American Journal of Human
Biology, 22(3), 291–296.
Michopoulos, V., Powers, A., Moore, C., Villarreal, S.,
Ressler, K. J., & Bradley, B. (2015). The mediating
role of emotion dysregulation and depression on the
relationship between childhood trauma exposure and
emotional eating. Appetite, 91, 129–136.
Mihrshahi, S., Battistutta, D., Magarey, A., & Daniels,
L. A. (2011). Determinants of rapid weight gain during infancy: Baseline results from the NOURISH randomised controlled trial. BMC Pediatrics, 11, 99.
Moholdt, T. T., Salvesen, K., Ingul, C. B., Vik, T., Oken,
E., & Morkved, S. (2011). Exercise Training in
Pregnancy for obese women (ETIP): Study protocol
for a randomised controlled trial. Trials, 12, 154.
Monteiro, P. O., & Victora, C. G. (2005). Rapid growth in
infancy and childhood and obesity in later life–a systematic review. Obesity Reviews, 6(2), 143–154.
National Association for Sport and Physical Education.
(2004). Moving into the future: National standards for physical education (2nd ed.). Reston, VA:
McGraw-Hill.
National Prevention Council. (2011). National prevention
strategy. Washington, DC: U.S. Department of Health
and Human Services, Office of the Surgeon General.
Neelon, S. B., Andersen, C. S., Morgen, C. S., KamperJørgensen, M., Oken, E., Gillman, M. W., & Sørensen,
T. I. A. (2015). Early child care and obesity at 12
months of age in the Danish National Birth Cohort.
International Journal of Obesity, 39(1), 33–38.
Nehring, I., Lehmann, S., & von Kries, R. (2013).
Gestational weight gain in accordance to the IOM/
NRC criteria and the risk for childhood overweight: A
meta-analysis. Pediatric Obesity, 8(3), 218–224.
Nehring, I., Schmoll, S., Beyerlein, A., Hauner, H., & von
Kries, R. (2011). Gestational weight gain and longterm postpartum weight retention: A meta-analysis.
The American Journal of Clinical Nutrition, 94(5),
1225–1231.
Nelson, M. C., Gordon-Larsen, P., & Adair, L. S. (2005).
Are adolescents who were breast-fed less likely to be
overweight? Analyses of sibling pairs to reduce confounding. Epidemiology, 16(2), 247–253.
193
Neu, J., & Rushing, J. (2011). Cesarean versus vaginal
delivery: Long-term infant outcomes and the hygiene
hypothesis. Clinics in Perinatology, 38(2), 321–331.
Newbold, R. R., Padilla-Banks, E., Snyder, R. J., Phillips,
T. M., & Jefferson, W. N. (2007). Developmental
exposure to endocrine disruptors and the obesity epidemic. Reproductive Toxicology, 23(3), 290–296.
Nielsen, L. S., Danielsen, K. V., & Sorensen, T. I. (2011).
Short sleep duration as a possible cause of obesity:
Critical analysis of the epidemiological evidence.
Obesity Reviews, 12(2), 78–92.
Office of the Mayor. (2012). Reversing the epidemic:
The New York City obesity task force plan to prevent
and control obesity. http://www.nyc.gov/html/om/
pdf/2012/otf_report.pdf. Accessed 5 Feb 2013.
O’Tierney, P. F., Barker, D. J., Osmond, C., Kajantie, E.,
& Eriksson, J. G. (2009). Duration of breast-feeding
and adiposity in adult life. The Journal of Nutrition,
139(2), 422S–425S.
Ogden, C. L., Carroll, M. D., Kit, B. K., & Flegal, K. M.
(2014). Prevalence of childhood and adult obesity
in the United States, 2011–2012. JAMA, 311(8),
806–814.
Ogden, C. L., & Flegal, K. M. (2010). Changes in terminology for childhood overweight and obesity. National
Health Statistics Reports, 25, 1–5.
Oken, E., & Gillman, M. W. (2003). Fetal origins of obesity. Obesity Research, 11(4), 496–506.
Oken, E., Levitan, E. B., & Gillman, M. W. (2008).
Maternal smoking during pregnancy and child
overweight: Systematic review and meta-analysis.
International Journal of Obesity, 32(2), 201–210.
Okereke, N. C., Uvena-Celebrezze, J., Hutson-Presley, L.,
Amini, S. B., & Catalano, P. M. (2002). The effect of
gender and gestational diabetes mellitus on cord leptin
concentration. American Journal of Obstetrics and
Gynecology, 187(3), 798–803.
Oteng-Ntim, E., Varma, R., Croker, H., Poston, L., &
Doyle, P. (2012). Lifestyle interventions for overweight and obese pregnant women to improve pregnancy outcome: Systematic review and meta-analysis.
BMC Medicine, 10, 47.
Owen, C. G., Martin, R. M., Whincup, P. H., DaveySmith, G., Gillman, M. W., & Cook, D. G. (2005a).
The effect of breastfeeding on mean body mass
index throughout life: A quantitative review of published and unpublished observational evidence.
The American Journal of Clinical Nutrition, 82(6),
1298–1307.
Owen, C. G., Martin, R. M., Whincup, P. H., Smith, G. D.,
& Cook, D. G. (2005b). Effect of infant feeding on
the risk of obesity across the life course: A quantitative review of published evidence. Pediatrics, 115(5),
1367–1377.
Pan, L., Blanck, H. M., Sherry, B., Dalenius, K., &
Grummer-Strawn, L. M. (2012). Trends in the prevalence of extreme obesity among US preschool-aged
children living in low-income families, 1998–2010.
JAMA, 308(24), 2563–2565.
194
Papas, M. A., Alberg, A. J., Ewing, R., Helzlsouer, K. J.,
Gary, T. L., & Klassen, A. C. (2007). The built environment and obesity. Epidemiologic Reviews, 29,
129–143.
Parker, M., Rifas-Shiman, S. L., Belfort, M. B., Taveras,
E. M., Oken, E., Mantzoros, C., & Gillman, M. W.
(2011). Gestational glucose tolerance and cord blood
leptin levels predict slower weight gain in early
infancy. The Journal of Pediatrics, 158(2), 227–233.
Parsons, T. J., Power, C., Logan, S., & Summerbell, C. D.
(1999). Childhood predictors of adult obesity: A systematic review. International Journal of Obesity and
Related Metabolic Disorders, 23(8), S1–107.
Patel, R., Martin, R. M., Kramer, M. S., Oken, E.,
Bogdanovich, N., Matush, L., Smith, G. D., &
Lawlor, D. A. (2011). Familial associations of adiposity: Findings from a cross-sectional study of 12,181
parental-offspring trios from Belarus. PloS One, 6(1),
e14607.
Pearce, J., Taylor, M. A., & Langley-Evans, S. C. (2013).
Timing of the introduction of complementary feeding
and risk of childhood obesity: A systematic review.
International Journal of Obesity, 37(10), 1295–1306.
Perna, F. M., Oh, A., Chriqui, J. F., Masse, L. C., Atienza,
A. A., Nebeling, L., Agurs-Collins, T., Moser, R. P.,
& Dodd, K. W. (2012). The association of state law
to physical education time allocation in US public
schools. American Journal of Public Health, 102(8),
1594–1599.
Perng, W., Gillman, M. W., Mantzoros, C. S., & Oken,
E. (2014). A prospective study of maternal prenatal
weight and offspring cardiometabolic health in midchildhood. Annals of Epidemiology, 24(11), 793–800
e791.
Piernas, C., & Popkin, B. M. (2011). Increased portion
sizes from energy-dense foods affect total energy
intake at eating occasions in US children and adolescents: Patterns and trends by age group and sociodemographic characteristics, 1977–2006. The American
Journal of Clinical Nutrition, 94(5), 1324–1332.
Powell, L. M., Chriqui, J. F., Khan, T., Wada, R., &
Chaloupka, F. J. (2013). Assessing the potential effectiveness of food and beverage taxes and subsidies
for improving public health: A systematic review of
prices, demand and body weight outcomes. Obesity
Reviews, 14(2), 110–128.
Rideout, V. J., & Hamel, E. (2006). The media family:
Electronic media in the lives of infants, toddlers, preschoolers, and their parents. Menlo Park, CA: Kaiser
Family Foundation.
Rudd Center for Food Policy & Obesity. (2013). Industry
self-regulation of food marketing.. http://www.yaleruddcenter.org/what_we_do.aspx?id=25. Accessed 5
Feb 2013.
Sanbonmatsu, L., Ludwig, J., Katz, L. F., Gennetian, L. A.,
Duncan, G. J., Kessler, R. C., Adam, E., TW, M. D., &
Lindau, S. T. (2011). Moving to opportunity for fair
housing demonstration program – Final impacts evaluation. Washington, DC: U.S. Department of Housing
and Urban Development.
S.S. Hawkins et al.
Savino, F., & Liguori, S. A. (2008). Update on breast milk
hormones: Leptin, ghrelin and adiponectin. Clinical
Nutrition, 27(1), 42–47.
Schmidt, M. E., Haines, J., O’Brien, A., McDonald, J., Price,
S., Sherry, B., & Taveras, E. M. (2012). Systematic
review of effective strategies for reducing screen time
among young children. Obesity, 20(7), 1338–1354.
Section on Breastfeeding. (2012). Breastfeeding and the
use of human milk. Pediatrics, 129(3), e827–e841.
Siega-Riz, A. M., Viswanathan, M., Moos, M. K.,
Deierlein, A., Mumford, S., Knaack, J., Thieda, P.,
Lux, L. J., & Lohr, K. N. (2009). A systematic review
of outcomes of maternal weight gain according to the
Institute of Medicine recommendations: Birthweight,
fetal growth, and postpartum weight retention.
American Journal of Obstetrics and Gynecology,
201(4), 339.e331–339.e314.
Slining, M. M., Benjamin Neelon, S. E., & Duffey, K. J.
(2014). A review of state regulations to promote infant
physical activity in child care. International Journal
of Behavioral Nutrition and Physical Activity, 11(1),
139.
Smith, G. D., & Ebrahim, S. (2003). ‘Mendelian randomization’: Can genetic epidemiology contribute
to understanding environmental determinants of disease? International Journal of Epidemiology, 32(1),
1–22.
National Center for Education Statistics. (2005). Number
of children under 6 years old and not yet enrolled in
kindergarten, percentage in center-based programs,
average weekly hours in nonparental care, and percentage in various types of primary care arrangements, by selected child and family characteristics:
2005. http://nces.ed.gov/programs/digest/d09/tables/
dt09_044.asp. Accessed 5 Feb 2013.
Story, M., Nanney, M. S., & Schwartz, M. B. (2009).
Schools and obesity prevention: Creating school environments and policies to promote healthy eating and
physical activity. The Milbank Quarterly, 87(1), 71–100.
Strasburger, V. C. (2011). Children, adolescents, obesity,
and the media. Pediatrics, 128(1), 201–208.
Taheri, S. (2006). The link between short sleep duration
and obesity: We should recommend more sleep to
prevent obesity. Archives of Disease in Childhood,
91(11), 881–884.
Taveras, E. M., Blackburn, K., Gillman, M. W., Haines,
J., McDonald, J., Price, S., & Oken, E. (2011a). First
steps for mommy and me: A pilot intervention to
improve nutrition and physical activity behaviors of
postpartum mothers and their infants. Maternal and
Child Health Journal, 15(8), 1217–1227.
Taveras, E. M., Gillman, M. W., Kleinman, K., RichEdwards, J. W., & Rifas-Shiman, S. L. (2010). Racial/
ethnic differences in early-life risk factors for childhood obesity. Pediatrics, 125(4), 686–695.
Taveras, E. M., Gillman, M. W., Kleinman, K. P., RichEdwards, J. W., & Rifas-Shiman, S. L. (2013).
Reducing racial/ethnic disparities in childhood
obesity: The role of early life risk factors. JAMA
Pediatrics, 167(8), 731–738.
Early in the Life Course: Time for Obesity Prevention
Taveras, E. M., Gillman, M. W., Pena, M. M., Redline, S.,
& Rifas-Shiman, S. L. (2014). Chronic sleep curtailment and adiposity. Pediatrics, 133(6), 1013–1022.
Taveras, E. M., Gortmaker, S. L., Hohman, K. H., Horan,
C. M., Kleinman, K. P., Mitchell, K., Price, S.,
Prosser, L. A., Rifas-Shiman, S. L., & Gillman, M. W.
(2011b). Randomized controlled trial to improve primary care to prevent and manage childhood obesity:
The high five for kids study. Archives of Pediatrics &
Adolescent Medicine, 165(8), 714–722.
Taveras, E. M., McDonald, J., O’Brien, A., Haines, J.,
Sherry, B., Bottino, C. J., Troncoso, K., Schmidt,
M. E., & Koziol, R. (2012). Healthy habits, happy
homes: Methods and baseline data of a randomized
controlled trial to improve household routines for obesity prevention. Preventive Medicine, 55(5), 418–426.
Taveras, E. M., Rifas-Shiman, S. L., Sherry, B., Oken,
E., Haines, J., Kleinman, K., Rich-Edwards, J. W., &
Gillman, M. W. (2011c). Crossing growth percentiles
in infancy and risk of obesity in childhood. Archives of
Pediatrics & Adolescent Medicine, 165(11), 993–998.
Te Morenga, L., Mallard, S., & Mann, J. (2012). Dietary
sugars and body weight: Systematic review and metaanalyses of randomised controlled trials and cohort
studies. BMJ, 346, e7492.
Thangaratinam, S., Rogozinska, E., Jolly, K., Glinkowski,
S., Roseboom, T., Tomlinson, J. W., Kunz, R., Mol,
B. W., Coomarasamy, A., & Khan, K. S. (2012).
Effects of interventions in pregnancy on maternal
weight and obstetric outcomes: Meta-analysis of randomised evidence. BMJ, 344, e2088.
Torloni, M. R., Betran, A. P., Horta, B. L., Nakamura,
M. U., Atallah, A. N., Moron, A. F., & Valente, O.
(2009). Prepregnancy BMI and the risk of gestational
diabetes: A systematic review of the literature with
meta-analysis. Obesity Reviews, 10(2), 194–203.
Torres, S. J., & Nowson, C. A. (2007). Relationship
between stress, eating behavior, and obesity. Nutrition,
23(11–12), 887–894.
Trasande, L., Attina, T. M., & Blustein, J. (2012).
Association between urinary bisphenol a concentration and obesity prevalence in children and adolescents. JAMA, 308(11), 1113–1121.
Trogdon, J. G., Nonnemaker, J., & Pais, J. (2008). Peer
effects in adolescent overweight. Journal of Health
Economics, 27(5), 1388–1399.
U.S. Department of Agriculture. (2013a). National
school lunch program and school breakfast program:
Nutrition standards for all foods sold in school as
required by the Healthy, Hunger-Free Kids Act of
2010; Interim Final Rule. Federal Register, 78(125),
39067–39120.
U.S. Department of Agriculture. (2013b). Supplemental
Nutrition Assistance Program (SNAP). http://www.
fns.usda.gov/snap. Accessed 5 Feb 2013.
U.S. Department of Health and Human Services. (2004).
The health consequences of smoking: A report of
the surgeon general. Atlanta, GA: U.S. Department
of Health and Human Services; Centers for Disease
Control and Prevention; National Center for Chronic
195
Disease Prevention and Health Promotion; Office on
Smoking and Health.
U.S. Department of Health and Human Services. (2008).
2008 physical activity guidelines for Americans.
Washington, DC: U.S. Department of Health and
Human Services.
Van Der Horst, K., Paw, M. J., Twisk, J. W., & Van
Mechelen, W. (2007). A brief review on correlates of
physical activity and sedentariness in youth. Medicine
and Science in Sports and Exercise, 39(8), 1241–1250.
Ventura, A. K., & Mennella, J. A. (2011). Innate and
learned preferences for sweet taste during childhood.
Current Opinion in Clinical Nutrition and Metabolic
Care, 14(4), 379–384.
Vesco, K. K., Karanja, N., King, J. C., Gillman, M. W.,
Leo, M. C., Perrin, N., CT, M. E., Eckhardt, C. L.,
Smith, K. S., & Stevens, V. J. (2014). Efficacy of a
group-based dietary intervention for limiting gestational weight gain among obese women: A randomized trial. Obesity, 22(9), 1989–1996.
Vesco, K. K., Karanja, N., King, J. C., Gillman, M. W.,
Perrin, N., McEvoy, C., Eckhardt, C., Smith, K. S.,
& Stevens, V. J. (2012). Healthy moms, a randomized trial to promote and evaluate weight maintenance
among obese pregnant women: Study design and rationale. Contemporary Clinical Trials, 33(4), 777–785.
Vgontzas, A. N., Lin, H. M., Papaliaga, M., Calhoun,
S., Vela-Bueno, A., Chrousos, G. P., & Bixler, E. O.
(2008). Short sleep duration and obesity: The role of
emotional stress and sleep disturbances. International
Journal of Obesity, 32(5), 801–809.
Viner, R. M., Ozer, E. M., Denny, S., Marmot, M., Resnick,
M., Fatusi, A., & Currie, C. (2012). Adolescence and
the social determinants of health. Lancet, 379(9826),
1641–1652.
von Kries, R., Bolte, G., Baghi, L., & Toschke, A. M.
(2008). Parental smoking and childhood obesity–is
maternal smoking in pregnancy the critical exposure? International Journal of Epidemiology, 37(1),
210–216.
Wang, Y., & Beydoun, M. A. (2007). The obesity epidemic in the United States–gender, age, socioeconomic, racial/ethnic, and geographic characteristics:
A systematic review and meta-regression analysis.
Epidemiologic Reviews, 29, 6–28.
Wang, Y. C., Gortmaker, S. L., & Taveras, E. M. (2011).
Trends and racial/ethnic disparities in severe obesity among US children and adolescents, 1976–2006.
International Journal of Pediatric Obesity, 6(1), 12–20.
Waterland, R. A., & Jirtle, R. L. (2004). Early nutrition,
epigenetic changes at transposons and imprinted
genes, and enhanced susceptibility to adult chronic
diseases. Nutrition, 20(1), 63–68.
Waters, E., de Silva-Sanigorski, A., Hall, B. J., Brown,
T., Campbell, K. J., Gao, Y., Armstrong, R., Prosser,
L., & Summerbell, C. D. (2011). Interventions for
preventing obesity in children. Cochrane Database of
Systematic Reviews, 12, CD001871.
Wen, X., Gillman, M. W., Rifas-Shiman, S. L., Sherry, B.,
Kleinman, K., & Taveras, E. M. (2012). Decreasing
196
prevalence of obesity among young children in
Massachusetts from 2004 to 2008. Pediatrics, 129(5),
823–831.
Weng, S. F., Redsell, S. A., Swift, J. A., Yang, M., &
Glazebrook, C. P. (2012). Systematic review and
meta-analyses of risk factors for childhood overweight
identifiable during infancy. Archives of Disease in
Childhood, 97(12), 1019–1026.
White House Task Force on Childhood Obesity. (2011).
One year progress report. http://www.letsmove.gov/
sites/letsmove.gov/files/Obesity_update_report.pdf.
Accessed 8 Feb 2013.
S.S. Hawkins et al.
White House Task Force on Childhood Obesity. (2010).
Solving the problem of childhood obesity within a generation: The White House task force on childhood obesity report to the president http://www.letsmove.gov/
sites/letsmove.gov/files/TaskForce_on_Childhood_
Obesity_May2010_FullReport.pdf. Accessed 19 July
2012.
Yu, Z. B., Han, S. P., Zhu, G. Z., Zhu, C., Wang, X. J.,
Cao, X. G., & Guo, X. R. (2011). Birth weight and
subsequent risk of obesity: A systematic review and
meta-analysis. Obesity Reviews, 12(7), 525–542.
Open Access This chapter is licensed under the terms of the Creative Commons Attribution 4.0 International License
(http://creativecommons.org/licenses/by/4.0/), which permits use, sharing, adaptation, distribution and reproduction in
any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license and indicate if changes were made.
The images or other third party material in this chapter are included in the chapter’s Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the chapter’s Creative Commons license
and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain
permission directly from the copyright holder.
Pediatric Type 2 Diabetes:
Prevention and Treatment
Through a Life Course Health
Development Framework
Pamela Salsberry, Rika Tanda, Sarah E. Anderson,
and Manmohan K. Kamboj
1
Introduction
Estimates project that one in three US adults may
have diabetes in 2050 (Boyle et al. 2010). The
rates are even higher in some subgroups; for
example, in Hispanic females it is 1 in 2 (Venkat
Narayan et al. 2003). This problem is not limited
to the USA. Shaw et al. 2010 estimate that 7.7%
P. Salsberry, PhD, RN, FAAN (*)
College of Public Health, Division of Health
Behavior, Health Promotion, Institute for Population
Health, The Ohio State University, 1841 Neil Avenue,
353 Cunz Hall, Columbus, OH 43210, USA
e-mail: Salsberry.1@osu.edu
R. Tanda, PhD, RN
College of Health Science and Professions,
Ohio University, Grover Center W133B, Athens,
OH 45701, USA
e-mail: tandar@ohio.edu
S.E. Anderson, PhD
The Ohio State University, College of Public Health,
336 Cunz Hall, 1841 Neil Avenue, Columbus, OH
43210, USA
e-mail: sanderson@cph.osu.edu
M.K. Kamboj, MD
The Ohio State University, College of Medicine,
Endocrinology, Metabolism and Diabetes,
Nationwide Children’s Hospital,
700 Children’s Drive (ED425), Columbus, OH
43205, USA
e-mail: Manmohan.Kamboj@Nationwidechildrens.org
or 434 million adults will have diabetes by 2030
worldwide (Shaw et al. 2010). Until very recently,
type 2 diabetes mellitus (T2DM) was a disease
diagnosed in adults, but as the childhood obesity
epidemic has spread in both magnitude and
severity, the diagnosis of T2DM in adolescents
and young adults has become more common. The
prevalence of prediabetes in 12–19 years, estimated using National Health and Nutrition
Examination Survey (NHANES) data, increased
from 9% in 1999–2000 to 23% in 2007–2008
(May et al. 2012). Analyses using data from the
SEARCH for Diabetes in Youth Study (SEARCH)
indicate a 30.5% overall increase in the prevalence of T2DM in children/adolescents from
2001 to 2009 (Dabelea et al. 2014). Because of
the newness of this diagnosis in children and adolescents, very little is known about the natural
history of the disease, and it will likely take
decades to fully understand the determinants and
consequences of this new epidemic. Early indicators suggest that the disease may be more severe
(Elder et al. 2012) and more difficult to manage
when diagnosed in adolescents (Zeitler et al. 2012).
A recent analysis estimates that adolescents
with T2DM will lose approximately 15 years of
life and will experience severe and chronic complications by their 40s as a result of the disease
(Rhodes et al. 2012). The mean direct cost for
medical care for a man diagnosed with T2DM for
less than 15 years is estimated at $2465. The economic burden of T2DM rises significantly as the
© The Author(s) 2018
N. Halfon et al. (eds.), Handbook of Life Course Health Development,
DOI 10.1007/978-3-319-47143-3_10
197
P. Salsberry et al.
198
years with disease increase. For those with
disease 15 years or more, the costs are estimated
to be 10–50% higher. This grim forecast highlights that not only will the disease burden be significant but the economic burden will be high in
youth diagnosed with T2DM (Li et al. 2013).
Metabolic comorbidities occur with alarming frequency (West et al. 2009). Within the SEARCH
study, a significant portion of adolescents with
T2DM had hypertension, high LDL cholesterol,
or high triglycerides (Lawrence et al. 2009a;
Mayer-Davis et al. 2009). It is clear that there is
much to do to understand the epidemic, to determine effective prevention strategies, and to
develop interventions for those diagnosed with
the disease. It is also clear from what we are
learning that preventive and treatment strategies
need to begin early.
The risk for a child developing T2DM begins in
utero. Individual health development trajectories
emerge from this starting point and are the result of
factors that accumulate across time and/or are the
result of biological conditioning during sensitive
periods of development. Understanding how these
trajectories emerge, and which factors and events
result in disease-causing pathways, is key to not
only understanding the onset and variable natural
history of the disease but in determining ways to
prevent and treat it. There is an extensive and emergent body of literature that examines the mechanisms, pathways, and determinants of T2DM in
children and adolescents. Genetic and epigenetic
mechanisms are clearly a part of the story, but several studies also demonstrate the influence and
importance of the social context on developing
biological processes (Kempf et al. 2008). These
include behaviors and psychological stressors
nested within families, neighborhoods, and communities. Consistent with the principles of life
course health development that are presented by
Halfon and Forrest (2017), there is a growing consensus that T2DM is the result of the multilevel
interaction of genetic, behavioral, social, and economic factors with the timing, sequence, and dose
of exposure central to long-term outcomes.
A life course health development approach that
details risks and protective factors, as well as preventive and treatment strategies, contextualized
within the developmental stage of the child, is
required. While one’s genetic code may establish
susceptibility toward T2DM development, its
development is the result of a complex process of
person-environment interactions that are multidimensional, multidirectional, and multilevel. A
major aim of this chapter is to demonstrate that a
life course health development approach to pediatric T2DM is critical to the development of a sound
national strategy to prevent and treat T2DM in
children. We review major factors known to influence the development of pediatric T2DM and
track these across childhood. This set of factors
will likely change over the next decade as our
understanding of disease mechanisms deepens,
but what will not change is the importance of placing these factors into context and recognizing that
T2DM development is highly sensitive to the timing and social structuring of multiple environmental exposures. Research recommendations relevant
to the field of maternal and child health regarding
T2DM will be discussed through a life course lens.
This chapter is organized around four key developmental stages: preconception and intrauterine
life, infancy, childhood (early and middle), and
adolescence. These health development stages
unfold against the “backdrop” of each child’s
genetics, race/ethnicity, and family economic
status.
2
The Backdrop
2.1 Genetics
Having a relative with T2DM is an established
risk factor for the disease (Rosenbloom et al.
1999). Through a series of twin studies, heritability of fasting glucose levels has been estimated to
range from 38% to 51% (Katoh et al. 2005;
Snieder et al. 1999). Heritability is a population
parameter that measures the fraction of variation,
i.e., fasting glucose levels, among individuals in a
population that is attributable to their genotypes
(Visscher et al. 2008). At the individual level, a
positive family history of T2DM has been associated with reduced insulin sensitivity at younger
ages in children and adolescents when compared
Pediatric Type 2 Diabetes: Prevention and Treatment Through a Life Course Health Development…
with children with no family history (Arslanian
et al. 2005). There is a 40% increased lifetime
risk for T2DM for children if one parent has
T2DM; this increases to 70% if both parents have
T2DM (Groop and Tuomi 1997). Studies have
found that as many as 80% of youth with T2DM
have a close relative with diabetes (Arslanian
et al. 2005; Copeland et al. 2011).
Most of the genetic studies of T2DM have
been carried out with adults; genetic studies of
T2DM in children and youth are limited because
of low number of subjects (Morgan 2012). An
exception to this is a large meta-analysis that
combined data from 6 European cohorts to test
whether associations of common genetic variants
identified for fasting glucose or insulin levels in
nondiabetic adults were detectable in healthy
children and adolescents. They found that the
majority of novel fasting glucose loci identified
in genome-wide associations studies (GWAS) in
adults were detectable in childhood and at similar
effect sizes (Barker et al. 2011). Work is
progressing to examine whether associations
between adult-identified susceptibility genes with
T2DM hold in pediatric T2DM. For example,
transcription factor 7-like 2 (TCF7L2) has been
found to be associated with an increased risk of
T2DM in African-American youth but not in
non-Hispanic whites (Dabelea et al. 2011).
There is clear evidence that genetics plays a role
in childhood susceptibility to T2DM, but significant research is needed. This research will require
substantial collaboration across sites to achieve
sample sizes large enough to test hypotheses. While
progress is being made in elucidating underlying
gene variants in adults, testing to determine if the
same variants are operating in children and adolescents is required (Morgan 2012).
2.2
Prevalence and Incidence
of T2DM Related to Age, Race,
and Ethnicity
199
rates are noted for adolescents aged 15–19 years,
while incidence in children below 10 years in age
is low. These results come from the SEARCH
study, a multiethnic, population-based study
designed to estimate rates of diabetes (both type
1 and type 2) based upon health-care provider
assessment and in a subpopulation with further
blood analyses. Data from SEARCH indicated a
prevalence of diagnosed T2DM among 10–14year-olds of 0.23 cases per 1000 children in 2009,
up from 0.15 per 1000 in 2001 (Dabelea et al.
2014). Comparable prevalence estimates for adolescents (15–19 years) in 2001 and 2009 were
respectively 0.54 and 0.68 per 1000 (Dabelea
et al. 2014). Data from 1999 to 2010 NHANES,
representative of US youth 12–19 years, indicated a total prevalence of T2DM of 0.36 per
1000 (Demmer et al. 2013). This estimate combines diagnosed and undiagnosed cases of
T2DM, with approximately 1/3 of the total being
undiagnosed but having a fasting glucose level of
≥126 mg/dL (Demmer et al. 2013). The prevalence of pediatric T2DM is higher for females
and among minorities in the USA (Dabelea et al.
2014). SEARCH researchers modeling T2DM
incidence by age and race/ethnicity report increasing incidence during the elementary school
years with a peak in incidence at approximately
age 14 years for all racial-ethnic groups
(Imperatore et al. 2012). At any age, the incidence of T2DM for African-American, Hispanic,
and Asian/Pacific Islander youth is higher than
for non-Hispanic whites (Imperatore et al. 2012;
Dabelea et al. 2007), and the incidence for children under 10 years old is very low (annual incidence <1 per 100,000) (Dabelea et al. 2007).
However, because T2DM develops gradually, can
be asymptomatic, and may be misdiagnosed as
type 1 diabetes mellitus (T1DM) in children,
population estimates of prevalence and incidence
must be interpreted cautiously (Amed et al. 2010a).
2.3 Economic Status
The prevalence of T2DM among children has
risen with population increases in obesity, but
compared to adolescents, T2DM in preadolescent children occurs less frequently. The highest
Data on the relationship between economic status
(of parents) and children and adolescents
diagnosed with T2DM are limited, but there are
P. Salsberry et al.
200
indications of a positive correlation. The treatment
options for T2DM in adolescents and youth study
(TODAY Study) found that 41.5% of their subjects came from households with an annual
income of less than $25,000, and 26.3% were in
families in which the highest education level of
the parent/guardian was less than a high school
degree (Copeland et al. 2011). The association of
economic status measured by educational level,
occupation, and income with T2DM in adults has
been studied extensively and likely offers clues to
risk in children and adolescents. A systematic
review and meta-analysis that was global in scope
found associations with low levels of education
(relative risk (RR) = 1.41, 95% CI: 1.28–1.51),
with occupation (RR = 1.31, 95% CI: 1.09–1.57),
and with low income (RR = 1.31, 95% CI:
1.09–1.57). While the data were limited from
middle- and low-income countries in this study,
the increased risks were independent of the
income levels of countries (Agardh et al. 2011).
Another systematic review examining childhood
socioeconomic status (SES) as a risk for T2DM
found a strong relationship; although this relationship was attenuated by adult SES, a significant relationship remained (Tamayo et al. 2010).
Parental education has been found to be associated with insulin resistance in the CARDIA study
participants, and the association differed by race
and gender. Low parental education was associated with greater insulin resistance in AfricanAmerican and white females and in
African-American males, but not in white males
(Tamayo et al. 2012). Smith et al. 2011 found
similar results in the Framingham Offspring
Study where an inverse relation between cumulative economic status and T2DM in women, but
not men, was noted (Smith et al. 2011). These
results highlight why untangling the pathways
for risk will be difficult.
2.4
Social (Psychological) Stressors
There is strong evidence that acute or chronic
stress during critical periods in early life, childhood, and/or adolescence has long-term risks for
altered metabolic function including the develop-
ment of T2DM (Pervanidou and Chrousos 2012).
Obesity likely mediates this relation brought on
by unhealthy behaviors related to eating and
activity patterns as coping mechanisms for
chronic stress. Furthermore, chronic stress leads
to the dysregulation of the hypothalamicpituitary-adrenal (HPA) axis, with increased cortisol, catecholamine, and elevated insulin
concentrations (Pervanidou and Chrousos 2011).
Key stressors in children are recognized as those
related to the creation of a safe, stable, and nurturing environment, and when these are disrupted, the child’s long-term health suffers
(Shonkoff and Garner 2012).
2.5 Summary
These “backdrop” factors are the platform upon
which health develops and emerges over time,
influenced by dynamic interactions at multiple
levels (biological, behavioral, social/cultural) at
sensitive periods transacting simultaneously. The
next sections illustrate this principle by highlighting key factors within developmental stage
that have been linked to pediatric T2DM.
3
Preconception
and Intrauterine Life
Over the last 20 years, a significant body of evidence from epidemiological and animal studies
has established a strong link between the intrauterine environment and long-term health (Dyer
and Rosenfeld 2011; Gluckman et al. 2008;
McMillen and Robinson 2005; Sebert et al. 2011;
Sinclair et al. 2007). The notion of biological
embedding is now widely accepted, even though
the mechanisms are not completely understood
(Tarry-Adkins and Ozanne 2011). Maternal
nutritional status, gestational diabetes, high
levels of maternal stress, hypoxia/placental insufficiency, and environmental toxins have been
associated with the development of T2DM in
children and adolescents (Warner and Ozanne
2010; Boekelheide et al. 2012). Furthermore, the
relationship of these high-risk maternal factors to
Pediatric Type 2 Diabetes: Prevention and Treatment Through a Life Course Health Development…
intrauterine growth restriction (IUGR) and
small-for-gestational-age (SGA) newborns with
their predisposition for obesity, insulin resistance, and T2DM later in life has been established as well. These factors are briefly reviewed
below. Several excellent papers review the underlying mechanisms of these relationships in
greater depth (Dyer and Rosenfeld 2011;
Armitage et al. 2004; Berends and Ozanne 2012;
Martin-Gronert and Ozanne 2012; Thayer et al.
2012; Prince et al. 2014).
3.1
Maternal Nutrition: Prior
to and During Pregnancy
3.1.1 Maternal Under Nutrition
A series of studies linking low birth weight,
maternal undernutrition, and the development of
cardiovascular disease has fueled much work on
the impact of caloric restriction on the developing fetus (Hales and Barker 2001; Barker et al.
1993). Animal studies have found endocrine and
metabolic abnormalities associated with food
restrictions (Warner and Ozanne 2010). Data
suggest that undernutrition alters the structure
and/or function of the developing endocrinemetabolic axis, leading to an insulin resistance
and energy conservation in light of reduced calories. An overactivation of the HPA axis leads to
increased cortisol levels in growth-restricted
fetuses, a situation that mimics chronic stress
(Kanaka-Gantenbein 2010; Kapoor et al. 2006).
The effects of the undernutrition seem to depend
on the timing and extent of the deprivation. In a
rodent study, a 50% caloric restriction at the end
of the fetal period resulted in reduced β-cell mass
(Bertin et al. 2002), and if the caloric restriction
continued through early life, the reduction in
β-cell mass was permanent (Garofano et al.
1998). Furthermore, the type of diet seems to be
important as well. Because of the importance of
amino acids in fetal development, studies on the
effects of low-protein diets on fetal growth have
been pursued with high interest (FernandezTwinn et al. 2005). Studies have shown that when
dams were fed a low-protein diet, the results were
growth-restricted pups, and if the maternal diets
201
remained low protein during suckling, the growth
restriction was permanent. Most importantly,
these offspring then went on to develop glucose
intolerance and T2DM (Hoet et al. 1992).
3.1.2 Maternal Overnutrition
Maternal obesity at the time of pregnancy is
known to be associated with a greater risk for
child obesity (Salsberry and Reagan 2005), placing them at increased risk for the development of
T2DM. Fetal overnutrition has also been associated with insulin resistance and adult diabetes
(Armitage et al. 2008). In animal studies, offspring born to obese maternal rats had higher
fat-to-lean ratios at birth, and by 3 months of age,
these pups were insulin resistant (Samuelsson
et al. 2008). When pregnant dams are fed a highfat content, the offspring are noted to develop a
phenotype resembling the human metabolic syndrome (Armitage et al. 2008).
3.2 Maternal Stress
Increased maternal stress is associated with
adverse birth outcomes, in particular low birth
weight and preterm birth (Christian 2012). Studies
demonstrate that maternal exposure to glucocorticoids reduces birth weight in sheep and rats
(Tarry-Adkins and Ozanne 2011). Meaney et al.
(2007) have suggested that these birth outcomes
are mediated by effects on the HPA axis. Stressors
in the environment alter maternal physiology and
behavior, which then programs HPA activity in
the offspring (Meaney et al. 2007). These changes
have been linked to the development of T2DM
and other components of the metabolic syndrome
in the offspring (Brunton 2010).
3.3 Maternal Hypoxia/Placental
Insufficiency
Fetal oxygenation is also an independent risk factor for fetal growth abnormalities and long-term
development of T2DM. Reduction in fetal oxygenation has also been associated with IUGR, a
known risk factor for metabolic syndrome.
P. Salsberry et al.
202
Multiple underlying pathophysiological reasons
for fetal hypoxia include placental insufficiency,
uterine placental ligation, and smoking. These
effects all result in IUGR (Tarry-Adkins and
Ozanne 2011).
3.4
Environmental Exposures
Exposures to environmental toxins during pregnancy are under increased scrutiny as the evidence
builds suggesting significant life course health
affects with early-life exposures to a range of
chemicals (Thayer et al. 2012). In particular, nicotine, arsenic, and endocrine disrupting chemicals
(EDC) are tied to metabolic outcomes. For example, high levels of arsenic exposure have been
linked to T2DM in adults, and there is some suggestion that even low doses may also confer an
increased T2DM risk (Navas-Acien et al. 2008;
Maull et al. 2012). The causal mechanisms include
altered glucose metabolism, insulin resistance, and
obesity as a result of exposure (Seki et al. 2012).
Exposure to EDC, particularly bisphenol A (BPA),
is thought to lead to functional changes in gene
expression and increased risk for development of
obesity and T2DM (Barouki et al. 2012; Liu et al.
2013). This is an area of intense activity that will
likely result in significant new knowledge over the
next decade.
3.5
Microbiome
There is growing recognition of the importance
of the human microbiome in pediatrics (Johnson
and Versalovic 2012). During pregnancy a woman’s microbiome undergoes significant change
from the first to third trimester, changes thought
to be beneficial to the mother and the establishment of the neonatal microbiome (Prince et al.
2014; Koren et al. 2012). The infant’s microbiota
is influenced by the mode of delivery, with colonization in vaginally delivered newborns a function of maternal vaginal and intestinal microbiota,
while cesarean delivered newborns exhibit the
microbiome of the maternal skin microbiota.
The infant’s environment and nutrition further
influence the infant’s developing microbiome
(Johnson and Versalovic 2012). These environmental influences on the developing microbiome
are critical, as the microbiome is thought to play
a role in health and disease (Cenit et al. 2014;
Devaraj et al. 2013). Evidence from research in
adults suggests that the microbiome plays a critical role in T2DM. Ongoing research is beginning
to demonstrate implications for prevention and
treatment of metabolic conditions based upon
knowledge of the microbiome; extending this
work to children is of high importance (Szajewska
2013; Versalovic 2013).
3.6
Gestational Diabetes (GDM)
GDM represents a form of impaired glucose tolerance that is first recognized during pregnancy.
Insulin resistance normally occurs in pregnancy.
This allows the metabolic needs of the fetus to be
met, while an increased maternal insulin secretion
compensates for the insulin resistance. But in
GDM, this increased resistance is coupled with a
failure of increased insulin secretion, resulting in
hyperglycemia (Battista et al. 2011). GDM is
harmful to the mother and to the fetus. GDM has
been shown to alter pancreatic development and
insulin sensitivity in the fetus (Warner and Ozanne
2010). Fetal growth is often affected, resulting in
macrosomia or large-for-gestational-age infants
(Dyer and Rosenfeld 2011). The Hyperglycemia
and Adverse Pregnancy Outcome (HAPO) study
found a strong linear relation between fasting glucose and post challenge glucose with macrosomia
and neonatal adiposity (Metzger et al. 2008).
Macrosomic infants born to diabetic mothers have
been found to be more glucose intolerant between
the ages of 10–16 years, are more likely to be
obese, and are at a greater risk for T2DM (Silverman
et al. 1998). Because of methodological differences
in current studies, the long-term effect of GDM on
child outcomes is not fully understood, but studies
do suggest that increased rates of obesity, T2DM,
and other cardiovascular conditions may result
(Kim et al. 2011; Simeoni and Barker 2009).
Pediatric Type 2 Diabetes: Prevention and Treatment Through a Life Course Health Development…
Screening and Diagnosis Given the convincing
evidence that diabetes and gestational diabetes
are harmful to both the mother and fetus, considerable effort has been directed toward establishing consensus criteria for the diagnosis of
diabetes in pregnancy. There is controversy over
the “correct” diabetes threshold, that is, the
level at which maternal blood glucose conveys a
significant risk to the fetus. The HAPO study, a
multisite and multinational study, was designed
to address some of these questions. HAPO study
results showed a linear relation between increasing levels of fasting and 1 and 2 hour plasma
glucose levels post a 75 g oral glucose tolerance
test (OGTT) on the primary study outcomes,
including birth weight above 90th percentile,
cord blood C-peptide level above the 90th percentile, primary cesarean delivery, and neonatal
hypoglycemia (Hadar and Hod 2010). These
results have led to guidelines issued in 2010 by
the International Association of Diabetes and
Pregnancy Study Groups (IADPSG) for the
detection and diagnosis of hyperglycemic disorders in pregnancy. These guidelines include the
determination of the diabetes status of the pregnant woman at the first prenatal visit. Following
these guidelines is expected to increase the
number of all women with diabetes during pregnancy. These guidelines, however, remain controversial (Moses 2010). There are concerns
over increasing costs associated with the identification of a greater number of women with diabetes resulting in an increased number of women
receiving treatment.
The American College of Obstetricians
and Gynecologists and the National Diabetes
Education program have joined together in a call
for action, specifically recommending follow-up
testing during the postpartum period in women
who experienced GDM. Currently only approximately one-half of these women are tested.
Recommendations include the following: If the
postpartum test is normal, retest every 3 years
and at the first prenatal visit for subsequent pregnancies. If prediabetes is diagnosed at the post-
203
partum check, then the recommendation is to test
annually (Gabbe et al. 2012).
Treatment Intrauterine conditions are being
targeted in the HAPO study. The initial results
showed that close control of glucose improved
birth outcomes, leading to the new recommendations for the diagnosis and classification of
hyperglycemia during pregnancy (Metzger et al.
2010). The HAPO study is also following the
outcomes in children born to women with GDM;
this longitudinal follow-up may yield important
insights for the health of the offspring as well.
Early results, however, on offspring at ages 4
and 5 show no difference in body mass index
(BMI) in the treatment group (Gillman et al.
2010). Further work with longer follow-up is
required to determine if there are latent effects
of treatment that will potentially influence outcomes at a later age.
3.7 Summary
Intrauterine life is a sensitive period for T2DM
risk development. Building upon the backdrop
of genetic susceptibility and within the social
and economic context of the mother, the fetus
develops an underlying propensity toward
T2DM development. Much of this comes
through epigenetic changes influenced by
maternal nutrition, life stressors, chemical exposures, and maternal behaviors. The occurrence
of higher levels of risk is strongly correlated
with maternal economic disadvantage and the
result of molecular, physiological, behavioral,
cultural, and evolutionary processes that interact across time and space. Consistent with the
principles of life course health development
(LCHD), development of a phenotype at high
risk for T2DM development is thus the result of
person-environment interactions that are multidimensional, multidirectional, and multilevel,
which are highly sensitive to the timing and
social structure of environmental exposures.
P. Salsberry et al.
204
4
4.1
Infancy
Feeding
Multiple advantages of breastfeeding are well
accepted (American Academy of Pedicatrics
2012; James and Lessen 2009), but clarifying the
relationship between breastfeeding and future
metabolic function is not simple and requires
consideration of multiple factors that have effects
over one’s life course. Controlling social and
environmental factors between infancy and some
later age is difficult because information for such
a long interval is often missing. Despite the limitations, there are several studies that have examined infant feeding type or breastfeeding duration
and their relation to dietary habits or cardiometabolic risks. For example, a study that used the
Hertfordshire cohort has found an independent
association between exclusive breastfeeding
(vs. mixed or bottle-feeding) and adherence to
healthy dietary recommendations and less intake
of processed meat in middle to late adulthood
(Robinson et al. 2013). Socioeconomic factors
were not associated with the dietary recommendation adherence. The authors speculate breast
milk may have provided significant impact on
dietary flavor learning during infancy.
In a recent study by Martin et al. (2014), the
relationship between cardiometabolic risks and
long-term exclusive breastfeeding was examined.
This study was a follow-up study of a randomized control study where one group of new mothers was encouraged to undertake exclusive
breastfeeding and the other nonexclusive breastfeeding. While the original intervention significantly prolonged breastfeeding duration and
exclusiveness, the intervention effect did not
influence cardiometabolic factors, such as blood
pressure, fasting glucose, insulin, insulin resistance, and adiponectin, when children were at
age 11.5 years. A previous follow-up study of this
cohort also failed to show intervention effects on
anthropometry (height, weight, BMI, and skinfold) and blood pressure at age 6.5 years (Kramer
et al. 2009).
Investigating the effects of breastfeeding is
complex as there are likely multiple biological
and behavioral ways that breastfeeding may
influence the infant’s health development.
Nutrition-focused research studies that examine
the importance of the nutrients within breast milk
are ongoing. But there are also suggestions that
breastfeeding may impact an infant’s ability to
self-regulate behavior. For example, Taveras
et al. (2004), using the US mixed-race cohort,
have found that restrictive feeding style is associated with bottle-feeding and shorter breastfeeding duration, which could disrupt the infant’s
ability to self-regulate feeding behavior. Maternal
restrictive feeding style was also found to be
associated with increased child’s weight for age
at 18 months among an African-American sample (Thompson and Bentley 2013).
There has been particular interest in the effects
of breastfeeding on children who are exposed to
maternal diabetes in utero. The evidence is mixed
regarding the effects of breastfeeding among
offspring of diabetic mothers (ODM) and development of obesity and T2DM during childhood. A
study from Germany found that high consumption
of breastmilk during the first 7 days of life among
ODM may pose deleterious effects on body weight
and glucose tolerance measured at age 2 years
(Plagemann et al. 2002). These authors suggest
that certain substances contained in early milk of
mother’s with diabetes may program for metabolic
dysregulation in their offspring.
In the Nurses’ Health Study II (Mayer-Davis
et al. 2006), researchers found that exclusive
breastfeeding compared to exclusive formula
feeding, or longer duration of breastfeeding
(>9 months) compared to never breastfed, was
associated with lower odds of being overweight
at ages 9–14 years, controlling for child’s age,
gender, and pubertal development. However,
further controlling for other maternal and child
factors resulted in null association, and a separate
analysis for ODM did not reach statistical significance. A case control study using the SEARCH
participants found that longer duration of
breastfeeding resulted in a reduction in T2DM,
controlling for maternal, child’s, and social factors. When the child’s weight (measured using
BMIz) was included in the analysis, the association was attenuated to null, suggesting that the
Pediatric Type 2 Diabetes: Prevention and Treatment Through a Life Course Health Development…
child’s BMIz may mediate the pathway to a
diagnosis of T2DM (Mayer-Davis et al. 2008).
The mechanisms involved in the effects of
breastfeeding on child’s future weight status or
risks for T2DM are unclear. However, insulin and
other metabolic hormones that are present in
breast milk likely play some role. Insulin is present at more than threefold concentration in early
milk collected within the first weeks of the postpartum period than in mature milk collected
3 months postpartum (Ley et al. 2012). The insulin concentration in early milk does not appear to
be affected by the mother’s gestational diabetes
status. While the insulin concentration in mature
milk of nondiabetic women is not different from
that of women with GDM (Ley et al. 2012),
maternal prenatal metabolic indicators, such as
BMI, fasting glucose, insulin resistance, insulin
sensitivity, and adiponectin levels, can predict
insulin concentration in mature milk (Ley et al.
2012; Whitmore et al. 2012).
Other research efforts have been focused on
infant feeding practices, such as timing of solid
food introduction and amount of sugar-sweetened
drink during infancy and early childhood, and
their effects on childhood obesity development
(Gaffney et al. 2012). Another area is investigation of micro- and macronutrient constituents of
breast milk. A randomized clinical trial demonstrated that health modifications made to the diets
of pregnant and lactating women improved infant
metabolic health (Aaltonen et al. 2011). More
research is needed to investigate how infant feeding may influence metabolic health development
over the life course. An excellent review article
summarizes the critical next questions in lactation and neonatal nutrition, providing a road map
for this necessary research (Neville et al. 2012).
4.2
Infant Growth Patterns
Several studies (Fabricius-Bjerre et al. 2011;
Ekelund et al. 2007; Eriksson et al. 2006) highlight significant associations between infant/
childhood growth patterns and development of
T2DM and later cardiovascular disease. Because
growth is orchestrated by a complex series of
205
interactions between growth hormone, sex steroids, and insulin, tracking growth over time is an
important global health measure. Understanding
high-risk growth patterns may assist in earlier
identification of high-risk children with the
potential to begin interventions at an earlier age.
The Helsinki Birth Cohort Study (HBCS) 1934–
1944 has shown that a small body size at birth
with thinness during the first year followed by
higher BMIs later in childhood is associated with
the development of T2DM (Eriksson 2006). Also,
this study found that an early age at adiposity
rebound was associated with a markedly
increased risk of T2DM in adulthood (Eriksson
2011). An important recent finding suggests that
breastfeeding may alter childhood growth even in
children exposed to overnutrition during the
pregnancy. In the Exploring Perinatal Outcomes
among Children (EPOCH) study, breastfeeding
was associated with long-term effects on childhood BMI growth extending into late childhood.
Slower BMI growth velocity was seen among
adequately breastfed (breastfeeding >6 months)
children compared with infants who had low
breastfeeding status, and these benefits lasted
through age 9 years (Crume et al. 2012).
4.3 Sleep Duration
Shortened sleep duration is suggested as a risk factor for obesity, insulin resistance, and Type 2 diabetes among adults. Long-term effects of shortened
sleep during infancy on metabolic function are
unknown. A limited number of studies that examined the relationship between sleep duration
during infancy and subsequent obesity in early
childhood have produced conflicting results. For
example, studies of US children (Taveras et al.
2008, 2014) have found chronic sleep curtailment
from infancy to age 7 years was associated with
increased BMI z-scores. Because short sleep hours
were associated with longer TV viewing time, the
investigators suggested the screen time as one of
the potential pathways for the association.
Similarly, Bell and Zimmerman (2010) using sample from the Panel Survey of Income DynamicsChild Development Supplement, reported that
P. Salsberry et al.
206
shortened night time sleep (less than 9 hours) during early childhood (ages 0–59 months) but not
during school age was associated with increased
likelihood of being overweight or obese at followup at age 5–10 years. Both US studies include relatively large number of minority population
(30–50%) whose nighttime sleep is on average
shorter than white children. However, studies of
Dutch children and Australian children have
shown no such association (Hiscock et al. 2011;
Klingenberg et al. 2013). The latter two studies
utilized objective measures of sleep duration such
as actinography, while the US studies have relied
on parental report. Improvement in measurement
of child’s sleep duration and other behavioral
markers in longitudinal studies are needed to
advance the knowledge in this area.
4.4
Parenting and Postpartum
Depression
The impact of maternal postpartum depression
on parenting is significant during the first year of
life. Mothers who suffer from postpartum depression may lack responsiveness to infant cues for
hunger or satiety. However, the studies examining the association between maternal depressive
symptoms and child weight status have produced
mixed results. Several studies conducted in the
USA have shown the association between maternal postpartum depressive symptoms and inappropriate parenting practices such as shorter or
less intense breastfeeding (McLearn et al. 2006;
Gaffney et al. 2014), inappropriate early feeding
practices (Gaffney et al. 2014), and fewer healthy
interactions with infants (McLearn et al. 2006).
Exposure to chronic maternal depressive symptoms has also been associated with increased risk
of childhood overweight (Wang et al. 2013).
Cumulative exposure to maternal depressive
symptoms in early infancy can lead to disruption
of behavioral and metabolic cues for sensation
of hunger and satiety. A study involving five
European countries, however, has shown no relationship between maternal depressive symptoms
and adiposity measures at age 24 months (Grote
et al. 2010). The investigators of the latter study
noted that prevalence of high depressive symptom
scores differed significantly by the participating
countries, and therefore, the instrument to measure depressive symptoms used in this study may
not be comparably used by the participants across
different study sites. More prospective cohort
studies focused on the relationship between
infant/child development and maternal lifestyles
as well as infant’s surrounding environment are
needed. Cohort studies such as the Amsterdam
Born Children and their Development (ABCD)
study (van Eijsden et al. 2011) may provide further knowledge on this area.
4.5 Summary
Infant development occurs within a context of
“backdrop” factors and as an extension of what
has happened during the prior period (fetal life).
Because evolutionary life-history theory suggests
that development during fetal life prepares the
neonate for a particular external environment,
when conditions in utero match the conditions in
infancy, development unfolds as an extension of
pathways begun in utero. When a mismatch
occurs between these environments, aspects of
development may be compromised. This may be
the case when undernutrition in fetal life is followed by an abundance of nutrients postnatally, a
particularly risky circumstance for metabolic
health. The important point is that infant health
development must be seen as conditioned by the
interaction of processes and conditions during
intrauterine life, and that this wider lens brings
into sharper focus the multiple interacting risks
contributing to the development of T2DM. Our
examinations of how experiences in infancy feed
forward into this continuously evolving health
developmental process suggest that long-term
health behaviors are beginning to be set through
complex transactions between maternal behaviors
and preferences regarding feeding and sleep (both
of which are influenced by the mother’s own environment and psychological state) and the infant’s
developing behavioral response patterns.
Pediatric Type 2 Diabetes: Prevention and Treatment Through a Life Course Health Development…
5
5.1
Childhood
Childhood Overweight
and Obesity
Almost all children and adolescents with T2DM
are also overweight or obese, and excess adiposity, particularly visceral adiposity, is associated
with insulin resistance (Dabelea et al. 2007;
Haines et al. 2007; Amed et al. 2010b). Studies
with US and Canadian youth have found that
75–95% of those with T2DM are obese (BMI
percentile > 95th for age and sex) (Mayer-Davis
et al. 2009; Amed et al. 2010b; Liu et al. 2009;
Lawrence et al. 2009b; Bell et al. 2009). Another
study in obese children found that 25% of
obese children 4–10 years of age and 21% of
11–18 years of age were found to have an
impaired glucose tolerance. Silent T2DM was
identified in 4% of the obese adolescents (Sinha
et al. 2002). Researchers in the UK using the
International Obesity Task Force (IOTF) cut
point for defining obesity (which is higher than
the US reference (Rolland-Cachera 2011)) found
that 83% of youth with T2DM were obese
(Haines et al. 2007). These results clearly point to
the importance of understanding the underlying
factors that contribute to childhood obesity, and
also point toward the complex path that underlies
the development of T2DM. Further understanding of how multiple factors interact, with a key
emphasis toward the timing of these factors in
reference to the life course is needed (Ong 2010;
Tounian 2011).
5.2
Impaired Glucose Tolerance
and Progression to T2DM
Obese children may have deficits in glucose regulation that predict future T2DM, as well as less
favorable risk factor profiles for cardiovascular
disease (West et al. 2009; Burns et al. 2014).
Evidence of impaired glucose tolerance, elevated
fasting glucose, insulin resistance, and/or previously undiagnosed “silent” T2DM has been documented in multiple samples of obese youth
207
(Wabitsch et al. 2004; Sinha et al. 2002; Morrison
et al. 2012). Morrison et al. (2012) studied 259
overweight and obese children and adolescents
(aged 5–17 years) who were seeking weight-loss
treatment and found that >20% had prediabetes—impaired glucose tolerance as measured by
an oral glucose tolerance test or elevated fasting
glucose (Morrison et al. 2012). Among these
youth with prediabetes, a majority with impaired
glucose tolerance did not have elevated fasting
glucose levels (Morrison et al. 2012), which is
consistent with findings from an Italian study
(Morandi et al. 2014). The current American Diabetes Association (ADA) guidelines (American
Diabetes Association 2010) recommend that
overweight children ≥10 years or pubertal with
two or more additional risk factors for T2DM
have their fasting glucose measured (see Table 1).
ADA guidelines do not recommend screening
youth with an oral glucose tolerance test.
Morrison et al. found that the sensitivity and
specificity of the ADA criteria to identify youth
with prediabetes were respectively 39% and
70%. Furthermore, in their sample the prevalence
of prediabetes in children 5–9 years old was not
significantly different from that of youth
≥10 years old (Morrison et al. 2012). Some evidence indicates that elevated fasting triglycerides
may help to identify obese children and adolescents who would benefit from an oral glucose
tolerance test (Morrison et al. 2012; Morandi
et al. 2014). More longitudinal studies of children at risk for T2DM are needed to elucidate the
natural history and progression of the disorder. In
a sample of 117 obese children and adolescents
(4–18 years) in which two oral glucose tolerance tests were administered between 18 and
24 months apart, Weiss et al. 2005 found that of
38 youth with impaired glucose tolerance at the
first time point, 8 developed T2DM, 10 still had
impaired glucose tolerance, and 15 (46%) had
normal glucose tolerance at the second time point
(Weiss et al. 2005). Among the 84 youth with
normal glucose tolerance initially, 8 had impaired
glucose tolerance at the follow-up assessment
(Weiss et al. 2005). Progression from prediabetes
to T2DM has been studied among adults (Morris
P. Salsberry et al.
208
Table 1 Summary of screening guidelines for T2DM: Childhood overweight and obesity
Year
Recommendations
Other risk factors
2007
BMI > 85th% and aged ≥ 10 years
with ≥2 other risk factors FGT or 2 h.
GTT; BMI ≥ 95th% and
aged ≥ 10 years FGT every 2 years
ADA (Amercian Diabetes
Association 2000)
2012
BMI > 85th% or weight > % of ideal
for height with ≥2 other risk factors
screened every 3 years beginning at
age 10 years or onset of puberty,
whichever is earlier. Testing done
every 2 years
ISPADa (International
Society for Pediatric and
Adolescent Diabetes)
2011
BMI > 85th% with other risk factors.
In Asian children, screen with low or
high birth weight with 1 risk factor,
regardless of BMI
Parental obesity
Family history
Current lifestyle
BMI trajectory CVD risk factors
(HBP, cholesterol)
Family history of T2DM in first or
second-degree relatives
Certain race/ethnic groupsb
Signs of insulin resistance or
conditions associated with insulin
resistance (AN, HTN, dyslipidemia,
PCOS, SGA)
Maternal diabetes or GDM
Immediate family history of T2DM,
early family history of CVD, or
Signs of insulin resistance (AN,
dyslipidemia, HBP, PCOS)
Diabetes screening
Expert Committee
(Barlow 2007)
Overweight/Obesity screening
2010
USPSTF (Barton 2010)
Expert Committee
2007
(Barlow 2007)
Screen at 6 year and older for obesity
Every well child visit: height and
weight, calculate and plot BMI
a
The ISPAD guidelines are also sensitive to country resources, so that screening decisions will likely be tied directly to
the health-care resources with a country
b
Native American, African-American, Latino, Asian American, Pacific Islander
et al. 2013), and although less is currently known
about how the disease develops in children
and adolescents, this is an area of active
investigation.
5.3
Diabetes Prevention
A number of lifestyle interventions to prevent
T2DM in youth at high risk have been evaluated
in randomized trials. The Yale Bright Bodies
Healthy Lifestyle program is an intensive
6-month behavioral intervention for obese youth
and their families that has been evaluated in multiple randomized controlled trials (Savoye et al.
2007, 2011, 2014). Among 174 ethnically diverse
children and adolescents aged 8–16 years—69
assigned to the control condition—the youth in
the intervention group showed reductions in adiposity at 6 months that were maintained at 1 year
as well as reductions in fasting insulin levels,
which were seen at 6 months and also maintained
for 1 year (Savoye et al. 2007). These youth were
also assessed 24 months after the start of the trial
and the favorable changes in body composition
and insulin sensitivity seen at 6 and 12 months
were sustained at similar levels at 24 months
(Savoye et al. 2011). A subsequent trial among
10–16-year-olds with prediabetes (n = 38 receiving the Bright Bodies intervention and n = 37
standard clinical care) also demonstrated
improvements in glucose metabolism and insulin
sensitivity as well as reductions in adiposity and
improved fitness (Savoye et al. 2014). Such
results are encouraging, and although the intervention is intensive, it uses a standardized curriculum and is likely to be cost-effective long
term (Savoye et al. 2014).
The HEALTHY Study was a national 3-year
middle-school intervention program designed to
reduce risk factors for type 2 diabetes. It used a
randomized trial designed to determine whether
Pediatric Type 2 Diabetes: Prevention and Treatment Through a Life Course Health Development…
changes to the school environment of middleschool students over a 3-year period could impact
their risk for T2DM (Foster et al. 2010; Hirst
et al. 2009). Prevention of T2DM in youth
involves encouraging changes in eating and activity behaviors that should also reduce risk for obesity. Although many large and well-conducted
randomized trials of school-based interventions
to prevent obesity have had limited impact on
obesity prevalence (Luepker et al. 1996;
Caballero et al. 2003; Brown and Summerbell
2009), such interventions may improve glucose
homeostasis and other risk factors for T2DM
(Trevino et al. 2004; Rosenbaum et al. 2007). The
Healthy Study was a group randomized trial conducted in 42 middle schools (21 intervention, 21
control) in 7 areas of the USA between fall 2006
and spring 2009 (Hirst et al. 2009). Students were
in 6th grade when enrolled and in 8th grade when
outcomes were assessed. The interventions in the
Healthy Study included changes to the school
environment with regard to nutrition (Gillis et al.
2009), physical activity (McMurray et al. 2009),
and health messaging (Venditti et al. 2009). The
primary outcome was prevalence of BMI ≥85th
percentile (overweight and obesity), and at
baseline about 50% of students in intervention
and control schools were overweight or obese
(Kaufman et al. 2009). Over the course of the
study, this prevalence decreased in intervention
schools to 45.8% and in control schools to 45.2%,
and thus there was not an effect of the intervention on the prevalence of overweight and obesity
(Foster et al. 2010). However, the intervention
did have a modest and statistically impact on the
secondary outcomes of obesity, mean BMI
z-score, high waist circumference, and fasting
insulin levels (Foster et al. 2010). These results
suggest that changes to the school environment
regarding nutrition, physical activity, and health
messaging may reduce the risk of childhood
onset of T2DM (Foster et al. 2010)
Some evidence suggests that interventions targeting physical fitness may be as effective in
reducing risk for T2DM in youth as multicomponent interventions. Among obese 7–11-year-old
children, a program of 40 min of aerobic exercise
5 days per week for 4 months reduced adiposity
209
and insulin levels, but these effects were not
sustained in the absence of the program (Ferguson
et al. 1999). A dose-response effect for the benefit of time spent in exercise on fitness and
cardiometabolic measures of T2DM risk was
demonstrated in a subsequent study of overweight and obese 7–11-year-olds randomized to
a 10–15-week program of 20 or 40 min per school
day of aerobic activity or to a control group
(Davis et al. 2012). Similar findings were noted
in a pilot randomized study in which physical
activity was added to a family-based weight control treatment. The exercise group had lower visceral abdominal fat posttreatment, a result that
may be key in future work to reduce diabetes risk
(Saelens et al. 2011). In a YMCA-based program
targeted to prevent the development of T2DM in
high-risk inner-city African-American children
(TAT: Taking Action Together), initial results
were encouraging with improvement in glucoregulation in boys (but not girls) and a decrease
in BMI z-scores in the treatment group (Ritchie
et al. 2010; Raman et al. 2010).
5.4 Obesity Prevention
Because of the strong association between obesity
and T2DM, much of the literature that discusses
prevention of T2DM focuses on weight reduction
or healthy weight maintenance throughout childhood. In a systematic review of the effectiveness of
weight management interventions in children, the
US Preventative Services Task Force (USPSTF)
results support at least in the short term benefits of
comprehensive medium-to-high intensity behavioral interventions in children 6 years and older
(Whitlock et al. 2010). The outcomes studied in
the USPSTF review were limited to childhood
weight and not the progression to T2DM (see
Table 1).
Recent evidence points to the importance and
benefit of intervening early in children’s lives to
reduce obesity (Knowlden and Sharma 2012),
and a number of randomized controlled treatment
trials in preschool-aged children have demonstrated success. An intensive 16-week multidisciplinary program for overweight or obese Dutch
210
children aged 3–5 years and their parent was
compared to a “usual-maintained care” control,
and results showed reductions in BMI and waist
circumference that were for 12 months (Bocca
et al. 2012a). Among 33 obese preschool-aged
(2–5 years) children in the Midwest USA, a pilot
randomized trial compared two 6-month multidisciplinary treatment programs (one in which all
sessions were held at a clinic and another program that alternated between clinic sessions and
individual home-based visits by a pediatric psychology fellow); both interventions were consistent with Expert Committee (Barlow 2007) stage
3 recommendations and were compared to a control condition of pediatrician counseling as recommended by the Expert Committee as stage 1
treatment (Stark et al. 2014). Compared to pediatrician counseling, children in the intervention
with home visits group had greater reductions in
BMI z-score at 6 months (end of intervention)
and at 12 months than did children in the clinic
only intervention (Stark et al. 2014). These
results suggest that parents of obese preschoolaged children may benefit from in-home guidance regarding implementation and maintenance
of parenting behaviors related to weight reduction. In an observational study of children aged
6–16 years seeking treatment for obesity in
Sweden, researchers evaluated whether age and
severity of obesity impacted the likelihood of
treatment success over a 3-year period (Danielsson
et al. 2012). They found that younger children
(6–9 years) were more likely to have clinically
meaningful reductions in weight status than older
children and adolescents, and that treatment success was rare for adolescents particularly if they
were severely obese (Danielsson et al. 2012). The
difficulties associated with treating obesity once
it is established underscore the need to prevent
excess weight gain in young children before they
become obese.
It is well established that parents play a key
role in obesity treatment efforts directed at youth
(Kitzmann et al. 2010), and for obvious reasons
parents are the main audience for obesity prevention efforts in early childhood (Skouteris et al.
2011, 2012; Hesketh and Campbell 2010).
P. Salsberry et al.
Interventions directed at parents of infants have
shown promise in reducing children’s risk for
obesity (Campbell et al. 2013; Wen et al. 2012),
and parents of infants may be receptive to messaging aimed at increasing adoption of healthy
eating and activity behaviors. Emerging evidence
also suggests that interventions to promote
positive parenting in general may help to reduce
risk for childhood obesity (Harvey-Berino and
Rourke 2003; Brotman et al. 2012). Maternal
warmth and sensitivity are associated in prospective cohort studies with lower risk for obesity
(Anderson et al. 2012, 2014). An explanatory
mechanism may involve the impact of responsive
parenting on the child’s developing stress response system and regulatory pathways in the limbic areas of the brain (McEwen 2008; Schore
2005). In a national sample of preschool-aged
children, the household routines of having family
meals, ensuring that children get adequate sleep,
and limiting time spent watching television were
associated with lower prevalence of obesity
(Anderson and Whitaker 2010). A randomized
trial of a 6-month home-based intervention to
promote these household routines in a sample of
low-income families of 2–5-year-old children
reduced children’s BMI (Haines et al. 2013).
Recent evidence points to the importance of
intervening early. In a randomized clinical trial targeting 3–5-year-old overweight or obese children,
a multidisciplinary program reduced BMI, BMI
z-scores, waist circumference, and waist circumference z-scores compared to a usual treatment
control group. The treatment results were still
present 12 months after the start of the intervention (Bocca et al. 2012b). A 6-month intensive
home-based behavioral intervention program
aimed at obese preschoolers (ages 2–5 years) has
also shown promise in reducing BMI z-scores,
both during the treatment period and at 12 months
follow-up (Saelens et al. 2011). In an observational study of obese children ages 6–16 years, all
were exposed to a behavioral treatment; changes
in BMIs across a 3-year treatment window showed
that younger aged children (6–9 compared with
14–16 years) were more successful in improving
outcomes (Danielsson et al. 2012).
Pediatric Type 2 Diabetes: Prevention and Treatment Through a Life Course Health Development…
5.5
Screening Guidelines
Given what we know about factors associated
with diabetes, screening for T2DM in children
and adolescents is linked to weight status of the
child, age of the child, and pubertal status.
Several groups have published guidelines for
T2DM screening in children. These guidelines
are summarized in Table 1.
5.5.1 Growth in Childhood
Birth weight and growth trajectories have both
been recognized as factors that are associated
with the life course risk of T2DM. Given this, it
seems key that individuals from the start understand their risk and that primary-care providers
collect these data for all children. Surprisingly
few guidelines currently recommend including
birth weight as a component of a child’s risk
assessment for T2DM. An exception to this is the
ISPAD screening guidelines that include birth
size in their guidelines for Asian children. No
recommendations were found that considered the
actual growth of the child with respect to possible
high-risk trajectories. Including birth size and
growth as screeners for high-risk children has yet
to be fully examined. A better understanding of
the growth trajectory may be useful in identifying
high-risk children earlier, but further work is
needed in this area.
5.6
Summary
Childhood is the period where health behaviors
are formed. While there is evidence that these
behaviors are influenced by experience in the
prior developmental periods, there is also evidence that significant change is still possible,
especially in children under 5 years. Increasing
evidence from both observational and intervention studies suggests that behavioral change is
possible early on, but by middle childhood it
becomes increasingly more difficult to affect real
behavior change. These findings are pushing the
research community to design studies that include
young children, and the evidence is accumulating
that interventions at the youngest ages achieve
the best outcomes. During this period the child’s
211
world widens as he/she transitions into preschool
and kindergarten, raising further the need to
widen the focus of intervention beyond the
family.
6
6.1
Adolescence
Transition into Adolescence
Puberty is an important phase in the life course
that needs to be considered in studying the risks
for T2DM. During puberty there is a known
physiological insulin resistance and hyperinsulinemia that is thought to facilitate pubertal
weight gain and growth (Burt Solorzano and
McCartney 2010). The timing of the start of this
insulin resistance has been reported to begin
in mid-childhood, some years before puberty
(Jeffery et al. 2012). Understanding this insulin
resistance as it relates to the pubertal process is of
high importance, as the rates of childhood diabetes continue to rise. Obesity exaggerates this
pubertal insulin resistance, especially in girls,
and may partially explain advanced maturation in
obese females (Burt Solorzano and McCartney
2010).
6.2 Diagnosis of T2DM
The rising prevalence of both type 1 and type 2
among children and adolescents, in conjunction
with increasing obesity throughout the population, has complicated the diagnosis of diabetes
type. Past reliance on phenotype to distinguish
and diagnose type of diabetes is not reliable
(Amed et al. 2010a; Rosenbloom et al. 2008). In
obese adolescents who present with new-onset
diabetes, measurement of pancreatic autoantibodies is recommended as necessary to differentiate the type of diabetes (Zeitler 2010).
6.3 Treatment
Overall, there are few published evidence-based
treatment studies for pediatric T2DM. The
ADA and the ISPAD have published treatment
212
recommendations, but these are primarily
consensus based and built on current guidelines
for adult T2DM (Amed et al. 2010a; Herbst et al.
2014). There are a handful of studies that have
either been conducted or are ongoing specifically to address pediatric T2DM, studies which
address both questions of prevention and
treatment.
Treatment for T2DM has been addressed in a
large multicenter randomized clinical trial, the
Treatment Options for T2DM in Adolescents and
Youth (known as the TODAY Study) (Copeland
et al. 2011). Subjects were between the ages of
10 and 17, diagnosed with T2DM for less than
2 years, and had a BMI percentile at or above the
85th percentile for age and sex. Subjects were
randomized into one of 3 comparison groups:
metformin monotherapy, metformin plus rosiglitazone, or metformin plus an intensive family
lifestyle program. Rates of failure (defined as
poor glycemic control) were 52% in metformin
alone, 39% in metformin plus rosiglitazone, and
47% in metformin plus the lifestyle program.
Greatest weight loss was seen in the metformin
plus the lifestyle program (Zeitler et al. 2012).
These results highlight the difficulty that the
majority of adolescents have in maintaining good
glycemic control and suggest that many will
require insulin within a few years of diagnosis.
Ongoing research is needed to determine optimal
management strategies in adolescents with
T2DM.
The goal of therapy is to achieve euglycemia.
The blood glucose levels should be monitored
based on individual needs. Self- monitored blood
glucose testing may be done at premeal times, or
limited to before breakfast and before dinner
only. Postprandial blood glucose levels, obtained
2 h after dinner, also offer helpful data for dose
titration. The hemoglobin A1c is monitored at the
time of clinic follow-up every 3 months. This
reflects the average blood glucose over the past
3-month period. The aim of treatment is to
achieve a hemoglobin A1c of less than 6.5% and
the target blood glucose range = 70–120 mg/dl.
The first intervention for treatment of T2DM
remains healthy lifestyle modifications including
dietary modifications (including decreasing
P. Salsberry et al.
caloric intake) and increasing physical activity
and exercise regimens to achieve weight stabilization and ultimately weight loss. Data from a
recent study suggests that regular exercise was
associated with lowering of the hemoglobin A1c
(Herbst et al. 2014). Initiating changes in portion
control as well as food choices and implementing
modest amounts of exercise often prove difficult.
Often adolescents have long-standing overweight
and obesity concerns. Lifestyle changes tend to
be more successful if initiated by the whole family instead of the particular individual being
targeted.
If the diagnostic parameters are consistent
with T2DM, pharmacologic treatment will need
to be initiated. The particular therapeutic regimen
initiated is determined by the initial presentation
of T2 DM. If the adolescent presents in diabetic
ketoacidosis, insulin therapy is instituted, initially intravenously, with subsequent transition to
subcutaneous insulin therapy after the resolution
of the acute diabetes ketoacidosis phase. Oral
medication, specifically metformin, is initiated as
well once the ketosis has resolved. If the presentation of T2DM is less acute, with the diagnosis
being made on routine office testing, and hemoglobin A1c is less than 9%, medication therapy
may be initiated first with metformin. In patients
with symptomatology in between these 2 scenarios, where hemoglobin A1c is more than 9% in
the presence of typical symptomatology, but no
diabetes ketoacidosis, therapy may be initiated
with metformin along with basal insulin (Zeitler
et al. 2014). Other medications used for treatment of T2DM in adults are not approved for
adolescents below 16 years of age.
Adherence to therapeutic interventions is key
for achieving adequate diabetes control and is
strongly emphasized. Modifications to regimen
are based on hemoglobin A1c and review of
blood glucose data. Adolescents who are on metformin therapy as well as basal insulin may need
additional rapid acting insulin to cover their
meals. Adolescents who were initiated on insulin
and metformin, and achieve goal A1c, are feasible to gradually decrease the insulin dose, after
they are on maximal metformin dosing. If adolescents are able to modify lifestyle factors as well
Pediatric Type 2 Diabetes: Prevention and Treatment Through a Life Course Health Development…
as manage medication therapy adequately, it may
be feasible to discontinue insulin altogether,
while continuing on metformin therapy, albeit
done very gradually with close monitoring.
6.3.1 Bariatric Surgery
Bariatric surgery for weight loss or uncontrolled
T2DM is not commonly undertaken in the adolescent population. Results obtained in studies
with adults that report weight loss and improvement in T2DM after bariatric surgery procedures
(Courcoulas et al. 2014) has caused increased
interest in its use in adolescents with morbid obesity. When bariatric surgery has been used as a
treatment strategy for extreme obesity in adolescents, it has been found to be effective in improving or reversing abnormal glucose metabolism,
but long-term outcome data in this age group is
not yet available. This treatment option is limited
to a very specific population because of potential
complications (Brandt et al. 2010).
6.4
213
screening in youth diagnosed with T2DM. These
guidelines are summarized in Table 2.
In a study examining adherence to ADA recommendations for lipid levels, blood pressure,
hemoglobin A1c, microalbuminuria, and eye
examinations, 95% of the participants reported
having their blood pressure checked at all or most
visits, 88% had lipid levels measured, 83% had
kidney function tested, 68% underwent HbA1c
testing, and 66% underwent an eye examination
(Waitzfelder et al. 2011). Participants aged
18 years or older, particularly those with T2DM,
tended to have fewer tests of all kinds performed.
Age and family income emerged as important
correlates of overall quality of care in multivariate
models: older age and lower income were associated with not meeting the guidelines. Overall,
while there was good compliance with the
ADA-recommended guidelines, efforts to improve HbA1c testing and eye examinations were
needed, especially among older adolescents and
young adults (Waitzfelder et al. 2011).
Surveillance
6.5 Comorbidities
Comorbidities associated with T2DM should be
investigated and treated as needed based on individual clinical presentation. The ADA and the
ISPAD have published guidelines for comorbidity
The TODAY study has demonstrated that a
significant portion of adolescents with T2DM
develop comorbidities. Careful attention to these
Table 2 Summary of surveillance guidelines for comorbidities
ADA (Amercian Diabetes
Association 2000)
ISPAD (ISPAD 2011)
Year
2012
2011
Recommendations
Blood pressure measurement annually
Fasting lipid profile
Microalbuminuria assessment
Dilated eye examination performed at the time of diagnosis
Evaluation of polycystic ovarian disease and other comorbidities such
as sleep apnea, hepatic steatosis, orthopedic complications, and
psychosocial concerns
Blood pressure measured at least annually
Initial eye examination within 3 months of diagnosis and then annually
from 11 years on after 2 years of diabetes duration
Annual screening for microalbuminuria from age 11 and after 2 years
of diabetes duration
Fasting blood lipids after initial diagnosis and stabilization of diabetes
≥12 years; if family history of hypercholesterolemia, early CVD, or
family history is unknown, screening should start at 2 years
Peripheral and autonomic neuropathy assessed by history and physical
exam from age 11 years with 2 years of diabetes duration
P. Salsberry et al.
214
must be part of any management plan for these
youths. Common comorbidities include hypertension and other metabolic conditions. Table 2
summarizes guidelines for monitoring the development of comorbidities in children/adolescents
with T2DM.
6.6
Care Transition into Adult
Practice
The emerging adult period (ages 18–30 years) is
a high-risk time for youth with T2DM. These
transitions from the pediatric to adult care setting
often result in loss to follow-up at a time when
the emergence of complications is occurring and
the youth are engaging in high-risk health
behaviors (Peters and Laffel 2011). In the majority of health-care institutions, there are no wellstructured, seamless processes in place to ensure
smooth transitions of care for any chronic care
condition. Furthermore, there is little evidencebased research to guide practice on the management of this care transition in youth with T2DM.
In the absence of evidenced-based programs and
because of concerns about this transition, many
programs are in the process of designing and/or
implementing transition of care initiatives (Lee
2013). In 2011, a consensus-based document outlining recommendations for transition from pediatric and to adult diabetes care systems was
published. Part of this document specifically
highlighted the need for research to determine
the best strategies for transition to diabetic adult
care to assure optimal outcomes for the youth
(Peters and Laffel 2011).
6.7
Summary
Because T2DM is most likely diagnosed during
or shortly after puberty, the emphasis in adolescence is focused on understanding the components of care for the disease, including the
surveillance for and management of comorbidities. In adolescence the consequences of the
child’s early environments are becoming clear
and the window for prevention has narrowed.
7
Summary
and Recommendations
7.1 Summary
• Pediatric T2DM is a life course problem—this
chapter demonstrates that no one factor and no
one time period provides the explanation;
prevention and treatment is not simple. To
optimize childhood metabolic health, we must
begin prior to conception and at every age
seize the opportunities to shape the child’s
environment to enhance metabolic health
development.
• Significant progress has been made in describing the growing pediatric T2DM epidemic and
in identifying individual risk factors that
contribute to its development. Work is ongoing to understand the underlying mechanisms
of pediatric T2DM. This knowledge base will
likely grow rapidly over the next decade.
• A major limitation of much of this work is that
the studies tend to be singularly focused on
exploring and understanding a particular risk,
and are completed using a specific disciplinary approach. What is missing from most of
this literature is the recognition that these risks
exist within a dynamic developmental context
with implications from micro- to macrosystems. The next generation of studies needs to
consider how risks play out differently
depending upon prior or current exposures
and environments. What are the risk modifiers
and mediators?
• Accounting for the influence of time-specific
exposures during sensitive developmental
periods in conjunction with exposures across
time is limited in the current research, and
must be addressed more thoroughly in future
studies.
• Another major task facing pediatric T2DM
researchers is to understand the developmental similarities and differences in T2DM
between children and adults. When T2DM
“threatens” or occurs in children/adolescents,
the challenges of understanding and intervening are different from those in the adult population, both biologically and behaviorally.
Pediatric Type 2 Diabetes: Prevention and Treatment Through a Life Course Health Development…
• Pediatric T2DM must be understood within
the dynamic context of the child’s overall
health development. Research on the factors
and developmental processes occurring early
in life has shown how the emergence of T2DM
children and adolescents is conditioned by
prior experiences, exposures, and response
patterns earlier in the life course.
• Translating the current knowledge into
practice-relevant evidence is lagging and must
be accelerated.
7.2
7.2.1
Recommendations
Recommendation 1:
To Continue to Elucidate
Mechanisms that Contributes
to Pediatric T2DM by Focusing
the Research in Key Areas
There are several parts to this recommendation
including the identification of substantive
research areas to guide further work, as well as
recommendations about the process for doing the
work. Life course health development research
will require transdisciplinary teams of scientists
that examine phenomenon across different levels
(genetic, cellular, biological systems, families,
neighborhoods, and large and complex macrosystems) at different developmental stage of the
child. No one individual, and no one discipline,
can achieve this knowledge by working in a disciplinary silo.
There are several areas that will require further investigation, and in particular we need to
increase our understanding of factors that may
mitigate the effects of adverse early-life programming. Childhood adversity, including the
type of adversity (e.g., resource lack compared
with family chaos), and chronic stress are noted
as likely to be involved in the biological programming of several chronic health conditions
including pediatric T2DM, but this is poorly
accounted for in most of the current studies.
Inclusion of poverty and family stress get at some
of these factors, but better conceptual and empirical models are needed. Furthermore, the timing
of the exposure for these risks is also not well
215
documented, and their time-specific impacts are
not well understood (i.e., is it poverty during
pregnancy, during early childhood, or both?). On
the positive side, there is a small but growing
literature on the importance of parenting style,
particularly nurturing behaviors, as possibly
ways of mitigating the negative effects of various
types of adverse environmental contexts.
Research priorities include:
• Understanding the health capital and health
development capacity that mothers (and
fathers) bring to conception and determine
which of these factors are most important for
child health development
• Determining how adult-recognized T2DM genetic susceptibility applies in pediatric T2DM
• Increased work to identify the epigenetic
changes that occur in response to risk exposures (e.g., environmental toxins, stressors)
• Nutrition research, at every stage of
childhood
• Understanding how health behaviors develop
across time
But none of these are factors will provide a
satisfactory explanation in isolation; each must
be conditioned on prior and co-occurring factors.
A life course health development perspective
suggests that research must be embedded within
the life stage of the child, taking into account the
child’s full developmental history, including the
intrauterine period. Exploring contemporaneous
factors, e.g., physical activity, and their association with T2DM should be done conditional on
prior life course exposures, not simply on current
factors; for example, knowing that the child was
exposed to diabetes in utero and the birth size of
the infant may be key markers for understanding
differential effects of contemporaneous factors.
Figure 1 integrates these ideas to display the life
stages along the horizontal axis and prevention,
screening, and treatment/surveillance along the y
axis. Embedded within this figure but not displayed because of the complexity is that at each
point multiple factors from multiple levels are
transacting simultaneously to bring about an
outcome.
P. Salsberry et al.
216
Rx of DM –
T2DM or GDM
(HAPO Study)
Diet quality
Prevenon,
Treatment &
Surveillance
Longitudinal follow-up of children born to mothers with DM,
GDM, with targeted intervenons aimed at high risk groups
Preschool
obesity Tx
DM Screening @
1st prenatal
visit, then risk
stratified
Set optimal wt
gain target
Smoking
cessation
Screening
Determinants,
Risk Factors for
T2DM
NOTE: Green indicates
recommendaons based upon
consensus documents (the what
we know).
Red indicates areas for further
study (that is, areas where we
need more knowledge).
Maternal GDM,
Family DM
history. BMI,
smoking,
acvity level,
diet
Preconception
GDM, Family
DM history,
BMI,
Smoking, High
level of stress,
Diet quality of
mother;
Race/
ethnicity
Pregnancy
TODAY STUDY
(Treatment)
HEALTHY Study
(prevenon)
Community-Based
Programs (YMCA)
TAT
Transioning to
adult care
Surveillance for DM
& Met Syndrome
complicaons:
renopathy, HBP,
Lipids, triglycerides
Exercise Trials
Bariatric surgery
“Stress” Assessment of Family as well as individual child
High risk groups: BMI %le>85th , starng at 1 0
or earlier if reached puberty screen for T2DM
Growth Trajectories: Height and Weight
Diet/feeding –
nutrient
composion
Lifestyle factors: Diet, acvity condioned on
family history, individual risk for diabetes
Feeding-processes
First Year
Years 2 & 3
Preschool
School – age
Adolescent
Fig. 1 A life course approach for understanding the risks, prevention and treatment of type 2 diabetes
7.2.2
Recommendation 2:
To Strengthen Data
and Methods Used to Address
Identified Research Priorities
and Explore the Feasibility
of an International Effort
to Harmonize Ongoing Birth
Cohorts for the Purpose
of Developing a Better
Understanding of the Timing,
Dose, and Interaction of Factors
that Contribute to Pediatric
T2DM Worldwide
Few data sets used to study the onset and impacts
of T2DM include significant information on the
broader context of the family, school, or neighborhood. In studies with good social data, biological data are often not included. A wider
(micro to macro contexts) and longer (birth
through adolescence) lens is needed to study
T2DM. The need for longitudinal studies that
can provide this lens is widely noted in the
research literature, but because of the costs and
subject burden, these long-term studies are difficult to carry out. However, there are a large
number of longitudinal birth cohort studies
ongoing worldwide that may have the potential
for improving the understanding of pediatric
T2DM. The recommendation here is to explore
the feasibility of an international effort to harmonize ongoing birth cohorts to develop a better
understanding of the timing, dose, and interactions of factors that contribute to T2DM worldwide. An added benefit of the development of
such a data set is that it would provide the data to
generate and test hypotheses, not just about
T2DM but data directly relevant to the testing of
the life course health development theoretical
framework.
Table 3 presents an overview of international
birth cohort studies. Of these studies, one dates to
the 1950s, one to the 1960s, two during the
1970s, and another two in the 1980s. Twelve of
the birth cohorts began data collection in the
1990s and 21 began data collection between 2000
and 2011. From this list, there are a handful of
studies with data elements (birth data, multiple
data collection points throughout childhood,
including adolescent health and height and
weight) pertinent to informing a life course picture of key pathways in this epidemic. Six birth
cohorts were identified as having collected
data beyond 1995 and included at least one data
collection point in adolescence (defined as data
Table 3 Birth cohort longitudinal studies
Cohort/country
Early Childhood
Longitudinal
Study—Birth
cohort/US
(ECLS)
National Child
Development
Study (NCDS):
UK 1958 Cohort
1970 British
Cohort Study
Years
Births in 2001
Births in 1958
Births in 1970
Ages/data collection
points
Birth through
kindergarten entry:
9 months, 2 years,
4 years, and
kindergarten
1958, 7, 11, 16, 23,
33, 42, 46, 50; in
2003 @45, 9000 in
biomedical screens
1970, 5, 10, 16, 30,
34, and 42 (in 2012)
Sample characteristics
14,000 diverse nationally rep.
sample; oversampling of API,
AI, and Alaskan natives, twins,
LBW
Measures
Cognitive, social, emotional, physical
development—assessments, parents, child care
settings, and KGT
Comments: PI/Managing entity//
website
National Center for Education
Statistics
http://nces.ed.gov/ecls
17,000 born during 1 week in
1958 in England, Scotland,
Wales
Physical, educational, economic, employment,
family life, health behaviors, well-being, social
participation, attitudes
Centre for Longitudinal Studies,
Institute of Education: Jane Elliot, PI
http://www.cls.ioe.ac.uk/
17,000 people born in England,
Scotland, Wales in single week
in 1970
Health, physical educational and social
development, economic circumstances
Sibling and parent info; parenting, childcare,
school choice, child behavior and cognitive
development, child and parent health, parent’s
employment and education, income, poverty,
neighborhood, and residential mobility; social
capital and ethnicity
Education, social work/support for parents,
health, childcare, social inclusion
Managed by Centre for Longitudinal
Studies, Institute of Education; Alice
Sullivan, PI
http://www.cls.ioe.ac.uk/
Managed by Centre for Longitudinal
Studies, Institute of Education
Lucinda Platt, PI
http://www.cls.ioe.ac.uk/
Millennium
Cohort Study
Births in
2000–2001
9 months, 3, 5, 7, 11
(in 2012) plus health
visitor survey report
and fertility survey
report
19,000 people in the UK
Growing up in
Scotland (GUS)
Births in
Scotland:
2003/2004
birth cohort 1
and
2010/2011
birth cohort 2
Data collection yearly
on the early cohorts;
for the 2010
cohorts—only at key
transitions
Growing up in
Ireland
Births in
1999/2000 for
older cohort;
2007/2008 for
infant cohort
Two rounds of data
collections with each
group
8000 children were enrolled into
the study in 2005–20065000
were babies (~10 months) born
between 6/ 2004 and 5/
20053000 were toddlers
(~34 months) born between
6/2002 and 5/ 2003. 6000
children born between 3/2010
and 2/ 2011 will be enrolled
during 2010–2011; data
collection at 10 months old.
8500 9-year-olds and
11,000 9-month-olds interviewed
during September 2008 to April
2009. Second round data
collection took place between
January and August 2011.
The main aim of the study is to paint a full
picture of children in Ireland and how they are
developing in the current social, economic, and
cultural environment
Centre for Research onFamilies and
Relationships—University of
Edinburghhttp://growingupinscotland.
org.uk/
PI: Professor James Williams at
Trinity College Dublin. http://www.
growingup.ie/
(continued)
Table 3 (continued)
Cohort/country
NLSY: Children
of the ‘79
Years
US
Panel study of
income
dynamics
(PSID)
US Birth 1968
and ongoing
National
Longitudinal
Survey of
Children and
Youth
Canada 1994
and ongoing
Ages/data collection
points
The NLSY child
sample is comprised
of all children born to
the NLSY79 mothers;
mothers have been
interviewed
biennially; data on
child begins at birth
through adulthood
Birth through
adulthood. Annually
from 1968 to 1997,
biennially 1998. Child
development
supplement 1n 1997
from birth −12 and
2002/2003 for
5–18 years.
2007/2008 at
10–18 years. 18 years
transition to
adulthood at ‘05,‘07,
‘09, ‘11
Birth to early
adulthood—eight
cycles with varying
age ranges
(0–25 years)
Comments: PI/Managing entity//
website
Access to in data available at Bureau
of Labor Statistics web site www.bls.
gov/nls/nlsy79.htm
Sample characteristics
More than 11,000 children have
been identified; birth years range
from 1979 through 2010
Measures
Cognitive, behavioral, health status, home
environment, psychosocial, parental involvement,
economic conditions of the family, risky
behaviors
70,000 individuals over four
decades. Includes initial
participants from 1968 and their
biological/adopted descendants.
1997 and 1999 included 511
immigrants to the USA
Cognitive, behavioral and health status.
Afterschool physical activities, home
environment, psychosocial, parental involvement,
schooling and school environment
http://psidonline.isr.umich.edu/Guide/
default.aspx
Cycle 8: 35,795 children
Child health, family psychosocial, emotional,
development, teenage risk behaviors
http://www.statcan.gc.ca/start-debuteng.html
Danish
Longitudinal
Survey of
Children
Denmark
1995 and
2003
Birth to adulthood:
Birth 1995, 1996,
1999, 2003, 2007 (11
years old)
6011 children (born Sept 15-Oct
31, 1995 and mothers are Danish
citizens); 611 children (born
April 1-Dec 31, 1995 and
mothers had been residing in
Demark for 3 years); 6000
children born in 1995 who were
placed in group homes/foster
care—data collection began in
2003
Growing up in
France (ELFE)
France 2011
(children born
on specific
days of year
2009 [4 days
in each of the
four different
quarters])
Birth to adulthood:
birth, 2 months, 3, 6,
11
1000 children (goal is 20,000)
Effect of differences in the socioeconomic
situation, way of life, ethnicity, upbringing, and
organization of home life within the family and
its effects on childhood development.
Consequences of the institutionalized childhood.
Effectiveness of societal support of families and
children. Effect of leave-of-absence schemes on
the parents’ perception of the father/mother role,
or their perception of the role of the family/”the
public sector.” Effect of parents’ mutual relations
(conflict/harmony) on their children’s daily life.
Effect of parental education, way of life, cultural
background and working conditions of the
parents on the way they prioritize family life.
Conditions in childhood/adolescence that effect
social integrate. What social mechanisms
influence child resiliency
To study the various factors interacting all over
the life course up to adulthood (family structure,
social and physical environment, schooling,
health, and nutritional behavior) and to clarify the
impact of the experience lived during infancy on
the individual’s physical, psychological, social,
and professional development. Measure the
cumulative exposures to specific environmental
conditions or behaviors year after year and to
analyze their consequences on social and health
inequalities
http://www.sfi.dk/the_danish_
longitudinal_survey_of_
children-3214.aspx
http://www.cls.ioe.ac.uk/page.aspx?&
sitesectionid=326&sitesectiontitle=EL
FE+%28Growin%20g+up+in+France
(continued)
Table 3 (continued)
Cohort/country
European
Longitudinal
Study of
Pregnancy and
Childhood
(ELSPAC)
Years
Europe born
1991 to 1995:
Great Britain,
Isle of Man,
Czech
Republic,
Slovakia,
Russia,
Ukraine,
Croatia and
Estonia;
recently
added: Spain
and Greece.
Growing up in
Australia
Australia 2004
Ages/data collection
points
Pregnancy, birth to
19 years: 6 and
18 months; 3, 5, 7, 11,
15, 18 and 19 years
Two cohorts—
families with
4–5-year-old children
(4–19 years) and
families with
0–1-year-old infants
(birth to 16 years)
Interviewed biennially
Sample characteristics
7589 children
10,000 children – infant cohort
(Dabelea et al. 2014; Grote et al.
2010) and child cohort (4991)
Measures
Detect biological, psychological, and social
factors as well as factors of external environment
associated with survival and health of the fetus,
infant and a child. Biopsychosocial determinants
of the child’s development from conception until
the end of school age, while taking into
consideration a lifestyle of the family,
environment, care, or relationships among family
members as well as personalities of the mother
and the father. Furthermore, we are aiming to find
the links between biopsychosocial determinants
as well as origin and development of health
disorders of a fetus, newborn, and a child.
Medical issues during pregnancy—bleeding,
eclampsia, infections, adolescent mothers.
Specific termination of pregnancy—preterm
delivery, intrauterine growth retardation of the
fetus, abortions, child of diabetic mother.
Growth— weight; height; length of long bones;
circumference of head, waist, and arm; skin
folders; and other anthropometric data.
Development—developmental landmarks,
motoric coordination, intellectual abilities
Children’s social, economic, and cultural
environments to their adjustment and well-being
Comments: PI/Managing entity//
website
Research Institute of Preventive and
Social Paediatrics at the Faculty of
Medicine, Masaryk University Brno,
Czech Republic: Lubomír Kukla, PI;
Website: http://www.aloe-app.com/
index-en.php
Daryl Higgins, PI Website: http://
www.aifs.gov.au/growingup/index.
html
Birth to twenty
South Africa
1990
Birth through
adulthood Birth to
14 years annually and
14 through adulthood
biennially;
multigenerational
3273 children (1594 recruited
antenatal); 3273 women
(singleton births)
The Avon
Longitudinal
Study of Parents
and Children
1992
(ALSPAC)—
Children of the
‘90s study
UK
Birth to early
adulthood
14,000 mothers in the former
county of Avon enrolled during
pregnancy in 1991 and 1992.
The health and development of
their children has been followed
in great detail ever since
Lifecycle approach and attempts to integrate
historical, personal, social, and economic
information with data collected during all major
developmental stages. In the early years, the
emphasis was on physical growth, health and
health service usage; in the preschool years
attention turned to social behavior, and home and
center support for learning; during primary
school, data has been collected on educational
progress; in the teen years the study is
concentrating on precursors to adult health and
adjustment, including sexual behavior, school
achievement, substance use, diet, activity, and
conflict with the law. Regularly assessments are
made of children’s residential circumstances,
household composition, and socioeconomic
conditions. Growth measurements are taken and
blood and urine samples subjected to a number of
biochemical tests. Body composition, blood
pressure and physical activity are also assessed
During pregnancy: medical history and physical
health of mother and grandparents, exposure to
environmental pollutants, sleeping patterns, diet,
caffeine and alcohol consumption, smoking,
illegal drug taking, psychological well-being of
parents, their expectations and attitudes, social
support networks. Early childhood: health,
lifestyle, and development. At focus clinics,
physical measurements and psychological tests,
regular biological samples, DNA extraction,
school performance on SATS and GCSEs.
ALSPAC links to additional sources of data on
participants include national pupil database and
school census data, and information on death and
cancer
Child Youth Family and Social
Development, Human Sciences
Research Council: Linda Richter, PI
http://www.wits.ac.za/academic/
health/research/birthto20/10274/
home.html
http://www.bristol.ac.uk/alspac/
(continued)
Table 3 (continued)
Cohort/country
Childhood
Obesity – Early
Programming
by Infant
Nutrition
(CHOPIN)
Years
Germany:
2002 to 2004
enrollment
All Babies in
Southeast
Sweden
Sweden: Oct
1st 1997 to
Oct 1st 1999
Aarhus Birth
Cohort
Births
beginning in
1990
Denmark
Amsterdam
Born Children
and their
Development
(ABCD study)
Babies After
SCOPE:
Evaluating the
Longitudinal
Impact using
Neurological
and Nutritional
Endpoints
Ages/data collection
points
Follow through to age
11; data collection
yearly through 8 and
then again at 11
Sample characteristics
1678 infants in Belgium,
Germany, Italy, Poland, Spain
Measures
Birth outcomes (wt, length, gestational age,
Apgar, sex); child exposures infancy through age
11; development; child health, biological
samples. Also included a 1 year multicenter
intervention trial on newborn infants, to test
whether feeding infant formulae (differing in
protein content), can influence growth trajectories
during the first year of life
Multiple—beginning
during fetal life,
including bio-samples
during pregnancy
through age 14 years
Pre-pregnancy,
second and third
trimester
17,000 infants
Main exposure: nutrition, physical activity, sleep
pattern, infections, psychosocial factors, child
care, stress, drugs, vaccinations, genetics,
Outcomes: autoimmunity (Type 1 DM, celiac
disease, RA), allergy, obesity, Type 2 DM
Fetal programming hypothesis and on various
aspects of pregnancy, birth, and neonatal care.
Birth outcomes (wt, lt, GA at birth, Apgar,
congenital malformations, stillbirth, sex)
2003–2004
Pre-pregnancy,
pregnancy (1st, 2nd,
3rd trimester), birth, 5
and 6 years to
adulthood
6161 infants and 8266 mothers
Ireland: Aug
1st 2008 to
Oct 31 2011
Birth to 5 years, birth
to 1, 2, and 5 years
2185 children born in Ireland
100,000 children and 100,000
mothers who gave birth at the
Dept of Obst and Gyn, Aarhus
Univ Hospital, Skejby
Association between lifestyle, psychosocial
conditions and nutritional status during
pregnancy, and the child’s health at birth (birth
weight, pregnancy duration, perinatal mortality)
and in later life (growth, physical development,
behavior and cognitive functioning)
Maternal health, fetal growth, and childhood
nutrition, growth and development in the first
2 years of life. Birth outcomes (wt, lt, GA at
birth, sex)
Comments: PI/Managing entity//
website
PI: Berthold Koletzko, Div. Metabolic
and Nutritional Medicine, Dr. von
Hauner Children’s Hospital,
Ludwig-Maximilians-University of
Munich Medical
http://www.ist-world.org/
ProjectDetails.aspx?ProjectId=c63efc
2bdd954069bed34a6d65d33dd4&Sou
rceDatabaseId=e4fcfde0182a45898e8
741a1abae3984
PI Johnny Ludvigsson; Dept of Clin
Exp Medicine, Linköping university,
Sweden
http://www.researchweb.org/is/lio/
ansokan/56531
Dept. of Pediatrics and Perinatal
Epidemiology Research Unit, Aarhus
University Hospital, Skejby,
Denmark:Tine Brink Henriksen, PI
http://fetotox.au.dk/the-fetotox-birthcohorts/
aarhus-birth-cohort-biobank-abcb/
Academic Medical Centre/ University
of Amsterdam: Manon van Eijsden;
Tanja G.M. Vrijkotte, PI
http://www.abcd-study.nl/
University College Cork: PI Deirdre
Murray, PI
http://www.
nationalchildrensresearchcentre.ie/
research/
baseline-study-babies-after-scopeevaluating-longitundinal-impactusing-neurological-and-nutritionalendpoints/
Bologna Birth
Cohort
Italy
2004–2005
Birth to adulthood:
birth, 1, 2, 3, and
7 years
654 children, 651 mothers, 593
fathers delivering at the
University Hospital of Bolonga
Born in
Bradford
UK Jan 1st
2007 to Dec
24th 2010
Birth to adulthood: 1,
2, and 3 years
14,000 children, 14,000 mothers,
and 3000 fathers
Copenhagen
Child Cohort
2000
(CCC2000)
Denmark
Births in 2000
Pregnancy, birth to
adulthood: birth, 1, 6,
and 11 years
6090 children, 6090 mothers
Danish National
Birth Cohort
(DNBC)
Denmark
Births
1996–2002
Pregnancy, birth to
adulthood:
birth—7 years,
11 years
95,000 children, 100,418
mothers
Dortmund
Nutritional and
Anthropometric
Longitudinally
Designed Study
(DONALD)
Germany
Births from
1985 and
ongoing
Infants 3–6 months
old to adulthood:
birth—18 years
1300 each children, mothers, and
fathers; open cohort study.
Annual recruitment of 30–40
infants
Exposure: passive smoking, breastfeeding,
nutrition, allergens, indoor pollution, motor
vehicles pollution Outcome: wheezing, asthma,
eczema, atopic dermatitis, sleep, cognitive
development. Birth outcomes (wt, lt, GA at birth,
Apgar, stillbirth, sex)
Describe health and ill-health within a largely
bi-ethnic population; identify modifiable causal
pathways that promote well-being, or contribute
to ill-health; develop, design and evaluate
interventions to promote health and well-being;
Develop a model for integrating operational,
epidemiological, and evaluative research into
practice within the NHS and other health-related
systems; build and strengthen the local research
capacity
Developmental trajectories of psychopathology
and mental disorders from birth in a general
population birth cohort and to determine at risk
states of mental illness in childhood and
adolescence. Birth outcomes (wt, lt, GA at birth,
Apgar, congenital malformations, stillbirth, sex)
Pregnancy complications and diseases in
offspring as a function of factors operating in
pregnancy/fetal and early life. Fetal growth as an
outcome and as a determinant of later health.
Main exposures: infection, alcohol, tobacco,
environments, medication, psychosocial factors.
Nutrition in pregnancy, in general but with focus
upon fish intake Outcome measures: life-long
follow-up of children—mortality and morbidity.
Birth outcomes (wt, lt, GA at birth, Apgar,
congenital malformations, stillbirth, sex)
Environmental exposure is not the main focus of
the birth outcomes (wt, lt, GA at birth, apgar,
sex). Child exposures: child care, second hand
smoke, smoking, alcohol, breastfeeding, diet, PA,
medications, head circumference, sexual
maturation, allergies, infections, cardiac, cancer,
DM, BP, sleep, urine samples
Maria Pia Fantini, PI
Bradford Institute for Health
Research: John Wright, PI
http://www.borninbradford.nhs.uk/
Child and Adolescent psychiatric
Centre, University Hospital of
Copenhagen: Anne Mette Skovgaard,
PI
Statens Serum Institut, Copenhagen:
Jørn Olsen, PI
http://www.ssi.dk/English/RandD/
Research%20areas/Epidemiology/
DNBC/
Research Insitute of Child Nutrition:
Anette Buyken, Thomas Remer, Ute
Alexy, Mathilde Kersting – Pis
(continued)
Table 3 (continued)
Ages/data collection
points
Birth to adulthood:
birth, 1, 7, 9, and
12 years
Cohort/country
Gateshead
Millennium
Study
Years
UK births
from June 1st
1999 to May
31st 2000
Sample characteristics
1029 children and 1011 mothers
GECKO
Drenthe cohort
Netherlands
born April 1st
2006 to April
1st 2007
Pregnancy, birth to
adulthood:
birth—5 years
2997 children born in Drenthe,
Netherlands. Pregnant mothers
recruited in the third trimester
Generation R
Netherlands
birth Jan 21st
2002 to Feb
1st 2006
Pregnancy, birth to
adulthood: birth—5,
7, and 9 years
10,000 children, 97,780 mothers,
and 6500 fathers
Measures
Feeding, weaning, and growth, including
characteristics of maternal feeding style, eating
attitudes, and depression. Childhood aspects of
developmental psychopathology, behavioral and
emotional development, and repetitive behaviors.
Origins of childhood obesity, with particular
reference to food intake, physical activity and
sedentary behavior. Original purpose was
investigation of weight faltering
The overall aim of the GECKO Drenthe cohort is
to study the prevalence and early risk factors for
the development of childhood overweight and fat
distribution at very young age. Within this birth
cohort the growth of the children is closely
followed, starting at birth until adulthood. The
children are followed using questionnaires that
the parents have to answer and the regular visits
to the Well Baby Clinics, where length, weight,
head circumference, and hip and waist
circumference are measured. Furthermore,
physical activity is measured using
accelerometers. At birth, cord blood was sampled
and stored for (epi)genetic analysis and
biomarkers. During the first 9 months of life, the
children were measured every month (incl.
questionnaire); thereafter this frequency
decreased until once every 2 years from 3 years
onward. The children are now 5 years old
Identification of early critical periods and causal
pathways for growth, development, and health.
Birth outcomes (wt, length, GA at birth, Apgar,
congenital malformation, still birth, sex)
Comments: PI/Managing entity//
website
Newcastle University: Ashley
Adamson, PI
http://www.cls.ioe.ac.uk/page.aspx?&
sitesectionid=332&sitesectiontitle=Ga
teshead+Millennium+Study
University Medical Center Groningen:
E. Corpeleijn, PI
Erasmus Medical Center Rotterdam:
Vincent Jaddoe, PI
http://research.lunenfeld.ca/
eagle/?page=841
Italy June 1st
2003 to Oct
31st 2004
Birth to 20 years:
birth, 1, 3, and 8 years
708 children, 693 mothers
(>18 years), and 63 fathers
Environmental exposures, diet, air pollution
asthma and allergies, accidents and injuries,
obesity, minor birth defects, neurodevelopment.
Birth outcomes (wt, length, GA at birth, Apgar,
congenital malformation, still birth, sex)
Department of Epidemiology
Regional Health Service lazio Region:
Daniela Porta, PI
Germany
births Jan 1st
1995 to Jan
1st 1998
Birth to adulthood:
birth—4, 6, 10,
and15 years
5991 children, maternal
pregnancy outcomes
HelmholtzZentrum München:
D. Berdel; A.v. Berg; C.P. Bauer;
U. Krämer; S. Koletzko;
J. Heinrich – PIs
Denmark
births April
1st 1984 to
April 1st 1987
Pregnancy, birth
annually through 18
to 20 years
KOALA Birth
Cohort Study
(KOALA)
Netherlands
Birth Jan 1st
2000 to Oct
1st 2002
Pregnancy, Birth to
adulthood: birth—2,
5–9 years
11,144 children of live birth
singleton pregnancy, 11,980
mothers who attended 2 midwife
in Aalborg or Odense during
week 35 to 38 of pregnancy
2843 children, 2900 mothers
>14 weeks in a pregnancy pelvic
pain or from an alternate
lifestyle practices, 2900 fathers
Intervention substudy; double -blinded
randomized trial for hypoallergenic formulae,
observational substudy: natural course of atopic
diseases and metabolic disorders; mental health,
nutrition, body weight and physical activity; Birth
outcomes (wt, lt, GA at birth)
Birth outcomes (wt, lt, GA at birth, Apgar,
congenital malformations, sex). Converted from a
trial to a birth cohort
Maastricht University: Carel Thijs, PI
http://www.koala-study.nl/
Lifeways
CrossGeneration
Cohort Study
Ireland 2001
to 2003
Pregnancy, birth:
birth—3, 5, and
10 years
Main outcomes: 1. allergy and asthma
development, 2. growth and overweight
development (inflammatory, immunological,
metabolic, cardiorespiratory and cardiovascular
aspects) Main exposures: microbial (infections,
gut microbiota), breastfeeding and breast milk
composition, nutrition, physical activity,
parenting practices and parenting styles; home,
neighborhood, and childcare environment. Birth
outcomes (wt, lt, GA at birth, Apgar, congenital
malformations, sex)
Physical and psychological health status,
socioeconomic circumstances, birth, childhood,
early adulthood and early middle age. Birth
outcomes (wt, GA at birth, Apgar, congenital
malformations, stillbirth, sex)
Genetic and
Environment:
Prospective
Study on
Infancy in Italy
(GASPII)
German Infant
Study on the
influence of
Nutrition
Intervention
(GINIplus)
Healthy Habits
for two (HHf2)
1074 children, 1061 mothers,
325 fathers
The Danish Epidemiology Science
Centre: Jørn Olsen, PI
University College Dublin: Cecily
Kelleher, PI
http://www.ucd.ie/phpps/research/
clinicalepidemiologygroup/
lifewayscross-generationcohortstudy/
(continued)
Table 3 (continued)
Ages/data collection
points
Pregnancy, birth to
16 years:
birth—4 years
Sample characteristics
1590 children; 1610 mothers
>16 years and 9–13 wks
gestation; 37 fathers
Norway 2003
to 2009
Birth to adulthood:
Birth −2 years,
8 years
2500 children breastfeeding at 2
wks of age, 2500 mothers, 2400
fathers
Norway Jan
1st 1999 to
Dec 31 2008
Pregnancy, birth to
adulthood: birth, 1, 3,
5, 7, 8 years
10,500 children, 90,700 mothers
17–20 wks pregnant, 72,100
fathers
Cohort/country
Mother Child
Cohort in Crete
(RHEA)
Years
Greece Feb
1st 2007 to
Feb 29th 2008
Norwegian
Human Milk
Study (HUMIS)
Norwegian
Mother and
Child Cohort
Study (MoBa)
Measures
1. The evaluation of nutritional, environmental,
biological, and psychosocial exposures in the
prenatal period and in early childhood. 2. The
evaluation of the association of these exposures
with the development of the fetus and the child,
reproductive outcomes, endocrine-related
outcomes, neurodevelopment, obesity, and the
occurrence of chronic diseases including asthma
and allergies, metabolic syndrome, and endocrine
disorders that appear first in childhood but that
may perpetuate to adulthood. 3. The evaluation of
mother’s health during and after pregnancy. 4.
The evaluation of genetic susceptibility and the
interactions between genetic and environmental
factors affecting child health. Birth outcomes (wt,
lt, GA at birth, Apgar, stillbirth, sex)
Exposures: environmental toxicants, gut
microbiota, pops, Pfos, organotins, gut
microbiota, Bifidobacterium, Lactobacillus, early
nutrition, human milk composition, duration of
breastfeeding. Main outcomes: child health,
reproductive health, asthma and allergy, ADHD,
neuropsychological development, depression,
overweight. Birth outcomes (wt, lt, GA at birth,
Apgar, congenital malformations, sex)
Causes of diseases. Additional aims are to detect
early signs of disease and to describe the
development of diseases. Birth outcomes (wt, lt,
GA at birth, apgar, congenital malformations,
stillbirth)
Comments: PI/Managing entity//
website
Dept of Social Medicine, Medical
School, Heraklion, University of
Crete, Greece and CREAL,
Barcelona, Spain: Manolis Kogevinas
and Leda Chatzi – PIs
Norwegian Institute of Public Health:
Merete Eggesbø, PI
Norwegian Institute of Public Health:
Per Magnus, PI
http://www.fhi.no/eway/default.aspx?
pid=240&trg=Main_6664&Main_666
4=6894:0:25,7372:1:0:0:::0:0&MainC
ontent_6898=6671:0:25,7899:1:0:0:::
0:0&List_6673=6674:0:25,7905:1:0:0
:::0:0
Odense Child
Cohort
Denmark
Births
beginning in
2010
Infants 0–5 months to
18 years
1650 children, 2249 mothers,
2102 fathers—living in Odense,
Denmark
Interaction between the fetus/infant/child during
pregnancy, birth, and childhood and the social
and environmental determinants for health and
disease. Birth outcomes (wt, lt, GA at birth,
Apgar, stillbirth, sex)
Study on the
pre- and early
postnatal
determinants of
child health and
development
(EDEN –
France)
France Births
2003 to 2005
Pregnancy, birth to
8 years: birth to
5 years and 8 years
1900 children, 2000 mothers
>18 years and <24 wks pregnant,
1800 fathers
Pre- and postnatal determinant of the child’s
development and health. Birth outcomes (wt, lt,
GA at birth, Apgar, congenital malformations,
stillbirth, sex)
Odense University Hospital, Hans
Christian Andersen Children’s
Hospital, Paediatric Research Unit:
Steffen Husby, PI
http://www.sdu.dk/en/Om_SDU/
Institutter_centre/Klinisk_institut/
Forskning/Forskningsenheder/
Paediatri/Odense+Børnekohorte
CESP INSERM: Heude Barbara, PI
The cohorts outlined here met the following criteria: Enrollment of the mother/child into the cohort was either prior to the pregnancy, during the pregnancy, or at birth; there was data collected
with some frequency in childhood; the studies were not single/narrow focused)
Key source is http://www.birthcohorts.net; this site is an inventory of birth cohort longitudinal studies maintained by Department of Public Health, University of Copenhagen, Professor Anne-Marie
Nybo Andersen
P. Salsberry et al.
228
collection at 15 years or beyond). These cohorts
also gathered data for childhood weight, height,
health assessment, and various social parameters.
The cohorts were the Canadian National Longitudinal Survey of Children and Youth (NLSCY,
data collected at various age points from birth
through age 25 over the course of 8 cycles), the
European Longitudinal Study of Pregnancy and
Childhood (ELSPAC, data collected at nine
points from birth through 19 years and includes
the countries of Great Britain, Isle of Man, Czech
Republic, Slovakia, Russia, Ukraine, Croatia,
Estonia, Spain and Greece), the Growing Up in
Australia survey (data collected through two
separate cohorts with one beginning at birth to
1 year and the second at 4/5 years with follow-up
to age 19 years), the South Africa’s Birth to 20
longitudinal study (data collected through two
separate cohorts that followed children ages birth
to 14 years and age 14 years through adulthood),
the UK’s Avon Longitudinal Study of Parents
and Children (ALSPAC) (data collected on children born between 1990 and 1992 from birth
through adulthood), and finally, the German
Infant Study on the Influence of Nutrition
Intervention (GINIplus), collected data on
children born between 1995 and 1998 annually
from birth through 4 years and then at 6, 10, and
15 years of age.
In addition to the data limitations currently
facing the pediatric T2DM research community,
there are also methodological/analytical
challenges. To make progress in all of these
research priorities, large amounts of data will be
needed. These data will be diverse ranging from
genetic and other biologicals to social structure
factors. Management and analyses of such
diverse data will require new methods and collaborations that will include basic scientists,
mathematicians, data engineers, practitioners,
and translational scientists. Researchers will
need to employ big data analytics and a complex
system perspective that can address multiple
causes and outcomes. The challenges are significant, but the opportunities vast to make a
real difference to the health development of all
children
7.2.3
Recommendation 3:
To Translate the Growing Body
of Knowledge into EvidencedBased Prevention
and Treatment Strategies
There are clear needs for both prevention and
treatment trials in children and adolescents.
There is a dearth of evidence-based studies and
guidelines available to pediatric practitioners
and pediatric endocrinologist to address the multiple management issues faced when treating
patients with T2DM. Areas of clear need include
prevention strategies for children born at high
risk. For example, low birth weight newborns,
offspring of diabetic mothers, could be placed in
programs to test interventions to modify risk.
This is a very important area of pediatric and
diabetes research with implications on health
status and care for large number of children into
adulthood and should be of high priority for
funding agencies.
Prevention trials are needed that can identify
strategies to reduce the risk for developing T2DM,
and as recent work demonstrates, understanding
the age at intervention is key. Prevention strategies may be gleaned from longitudinal data collected for other reasons. While not a prospective
trial, the primary data elements could be supplemented either through new data collection efforts
(e.g., during adolescence collecting insulin, glucose, etc.), or by combining the primary data with
other administrative data—child electronic health
records of heights and weights throughout childhood—or both. With home address information,
child contexts could be tracked over time and
neighborhoods accounted for in the analyses.
Finally, there is much work to be done to translate the knowledge into policy at every level.
Pediatric practitioners need up-to-date guidelines
to identify children at high risk for T2DM at young
ages. Families must be given the information about
the possible long-term consequences of decisions
made in early life. Local, state, and national governments also need policies in place that enhance
child health development throughout childhood.
Only then will we be able to achieve the goal of
optimal health development for every child.
Pediatric Type 2 Diabetes: Prevention and Treatment Through a Life Course Health Development…
References
Aaltonen, J., Ojala, T., Laitinen, K., Poussa, T., Ozanne,
S., & Isolauri, E. (2011). Impact of maternal diet during pregnancy and breastfeeding on infant metabolic
programming: A prospective randomized controlled
study. European Journal of Clinical Nutrition, 65(1),
10–19.
Agardh, E., Allebeck, P., Hallqvist, J., Moradi, T., &
Sidorchuk, A. (2011). Type 2 diabetes incidence and
socio-economic position: A systematic review and
meta-analysis. International Journal of Epidemiology,
40(3), 804–818.
Amed, S., Daneman, D., Mahmud, F. H., & Hamilton,
J. (2010a). Type 2 diabetes in children and adolescents. Expert Review of Cardiovascular Therapy,
8(3), 393–406.
Amed, S., Dean, H. J., Panagiotopoulos, C., Sellers, E. A.,
Hadjiyannakis, S., Laubscher, T. A., et al. (2010b).
Type 2 diabetes, medication-induced diabetes, and
monogenic diabetes in Canadian children: A prospective national surveillance study. Diabetes Care, 33(4),
786–791.
Amercian Diabetes Association. (2000). Type 2 diabetes
in children and adolescents. Pediatrics, 105(3 Pt 1),
671–680.
American Academy of Pedicatrics. (2012). Breastfeeding
and the use of human milk. Pediatrics, 129(3),
e827–e841.
American Diabetes Association. (2010). Diagnosis and
classification of diabetes mellitus. Diabetes Care,
33(Suppl 1), S62–S69.
Anderson, S. E., & Whitaker, R. C. (2010). Household
routines and obesity in US preschool-aged children.
Pediatrics, 125(1), 420–428.
Anderson, S. E., Gooze, R. A., Lemeshow, S., & Whitaker,
R. C. (2012). Quality of early maternal-child relationship and risk of adolescent obesity. Pediatrics, 129(1),
132–140.
Anderson, S. E., Lemeshow, S., & Whitaker, R. C. (2014).
Maternal-infant relationship quality and risk of obesity at age 5.5 years in a national US cohort. BMC
Pediatrics, 14(1), 54.
Armitage, J. A., Khan, I. Y., Taylor, P. D., Nathanielsz,
P. W., & Poston, L. (2004). Developmental programming of the metabolic syndrome by maternal nutritional imbalance: How strong is the evidence from
experimental models in mammals? The Journal of
Physiology, 561(Pt 2), 355–377.
Armitage, J. A., Poston, L., Taylor, P., & D. (2008).
Developmental origins of obesity and the metabolic
syndrome: The role of maternal obesity. Frontiers of
Hormone Research, 36, 73–84.
Arslanian, S. A., Bacha, F., Saad, R., & Gungor, N.
(2005). Family history of type 2 diabetes is associated
with decreased insulin sensitivity and an impaired balance between insulin sensitivity and insulin secretion
in white youth. Diabetes Care, 28910, 115–119.
Barker, D. J., Hales, C. N., Fall, C. H., Osmond, C.,
Phipps, K., & Clark, P. M. (1993). Type 2 (non-insulin-
229
dependent) diabetes mellitus, hypertension and hyperlipidaemia (syndrome X): Relation to reduced fetal
growth. Diabetologia, 36(1), 62–67.
Barker, A., Sharp, S. J., Timpson, N. J., Bouatia-Naji,
N., Warrington, N. M., Kanoni, S., et al. (2011).
Association of genetic loci with glucose levels in
childhood and adolescence: A meta-analysis of over
6,000 children. Diabetes, 60(6), 1805–1812.
Barlow, S. E. (2007). Expert committee recommendations
regarding the prevention, assessment, and treatment of
child and adolescent overweight and obesity: Summary
report. Pediatrics, 120(Suppl 4), S164–S192.
Barouki, R., Gluckman, P. D., Grandjean, P., Hanson,
M., & Heindel, J. J. (2012). Developmental origins of
non-communicable disease: Implications for research
and public health. Environmental Health : A Global
Access Science Source, 11, 42.
Barton, M. (2010). Screening for obesity in children and
adolescents: US preventive services task Force recommendation statement. Pediatrics, 125(2), 361–367.
Battista, M. C., Hivert, M. F., Duval, K., & Baillargeon, J. P.
(2011). Intergenerational cycle of obesity and diabetes:
How can we reduce the burdens of these conditions on
the health of future generations? Experimental Diabetes
Research, 2011, 596060. doi:10.1155/2011/596060.
Bell, J. F., & Zimmerman, F. J. (2010). Shortened nighttime sleep duration in early life and subsequent childhood obesity. Archives of Pediatrics & Adolescent
Medicine, 164(9), 840–845.
Bell, R. A., Mayer-Davis, E. J., Beyer, J. W., D’Agostino,
R. B., Jr., Lawrence, J. M., Linder, B., et al. (2009).
Diabetes in non-Hispanic white youth: Prevalence,
incidence, and clinical characteristics: The SEARCH
for diabetes in youth study. Diabetes Care, 32(Suppl
2), S102–S111.
Berends, L. M., & Ozanne, S. E. (2012). Early determinants
of type-2 diabetes. Best Practice & Research. Clinical
Endocrinology & Metabolism, 26(5), 569–580.
Bertin, E., Gangnerau, M. N., Bellon, G., Bailbe, D.,
Arbelot De Vacqueur, A., & Portha, B. (2002).
Development of beta-cell mass in fetuses of rats
deprived of protein and/or energy in last trimester of pregnancy. American Journal of Physiology.
Regulatory, Integrative and Comparative Physiology,
283(3), R623–R630.
Bocca, G., Corpeleijn, E., Stolk, R. P., & Sauer, P. J.
J. (2012a). Results of a multidisciplinary treatment
program in 3-year-old to 5-year-old overweight or
obese children: A randomized controlled clinical
trial. Archives of Pediatrics & Adolescent Medicine,
166(12), 1109–1115.
Bocca, G., Corpeleijn, E., Stolk, R. P., & Sauer, P. J.
(2012b). Results of a multidisciplinary treatment
program in 3-year-old to 5-year-old overweight or
obese children: A randomized controlled clinical
trial. Archives of Pediatrics & Adolescent Medicine,
166(12), 1109–1115.
Boekelheide, K., Blumberg, B., Chapin, R. E., Cote,
I., Graziano, J. H., Janesick, A., et al. (2012).
Predicting later-life outcomes of early-life expo-
230
sures. Environmental Health Perspectives, 120(10),
1353–1361.
Boyle, J. P., Thompson, T. J., Gregg, E. W., Barker, L. E.,
& Williamson, D. F. (2010). Projection of the year
2050 burden of diabetes in the US adult population:
Dynamic modeling of incidence, mortality, and prediabetes prevalence. Population Health Metrics, 8, 29.
doi:10.1186/1478-7954-8-29.
Brandt, M. L., Harmon, C. M., Helmrath, M. A., Inge,
T. H., McKay, S. V., & Michalsky, M. P. (2010). Morbid
obesity in pediatric diabetes mellitus: Surgical options
and outcomes. Nature reviews. Endocrinology, 6(11),
637–645.
Brotman, L. M., Dawson-McClure, S., Huang, K. Y.,
Theise, R., Kamboukos, D., Wang, J., et al. (2012).
Early childhood family intervention and long-term
obesity prevention among high-risk minority youth.
Pediatrics, 129(3), e621–e628.
Brown, T., & Summerbell, C. (2009). Systematic review
of school-based interventions that focus on changing
dietary intake and physical activity levels to prevent
childhood obesity: An update to the obesity guidance
produced by the National Institute for health and clinical excellence. Obesity Reviews, 10(1), 110–141.
Brunton, P. J. (2010). Resetting the dynamic range of
hypothalamic-pituitary-adrenal axis stress responses
through pregnancy. Journal of Neuroendocrinology,
22(11), 1198–1213.
Burns, S. F., Lee, S., Bacha, F., Tfayli, H., Hannon, T. S.,
& Arslanian, S. A. (2014). Pre-diabetes in overweight
youth and early atherogenic risk. Metabolism, 63(12),
1528–1235.
Burt Solorzano, C. M., & McCartney, C. R. (2010).
Obesity and the pubertal transition in girls and boys.
Reproduction, 140(3), 399–410.
Caballero, B., Clay, T., Davis, S. M., Ethelbah, B., Rock,
B. H., Lohman, T., et al. (2003). Pathways: A schoolbased, randomized controlled trial for the prevention of
obesity in American Indian schoolchildren. American
Journal of Clinical Nutrition, 78(5), 1030–1038.
Campbell, K. J., Lioret, S., McNaughton, S. A., Crawford,
D. A., Salmon, J., Ball, K., et al. (2013). A parentfocused intervention to reduce infant obesity risk
behaviors: A randomized trial. Pediatrics, 131(4),
652–660. doi:10.1542/peds.2012-2576.
Cenit, M. C., Matzaraki, V., Tigchelaar, E. F., &
Zhernakova, A. (2014). Rapidly expanding knowledge
on the role of the gut microbiome in health and disease.
Biochimica et Biophysica Acta, 1842(10), 1981–1992.
Christian, L. M. (2012). Physiological reactivity to
psychological stress in human pregnancy: Current knowledge and future directions. Progress in Neurobiology,
99(2), 106–116. doi:10.1016/j.pneurobio.2012.07.003.
Copeland, K. C., Zeitler, P., Geffner, M., Guandalini, C.,
Higgins, J., Hirst, K., et al. (2011). Characteristics of
adolescents and youth with recent-onset type 2 diabetes: The TODAY cohort at baseline. Journal of Clinical
Endocrinology and Metabolism, 96(1), 159–167.
Courcoulas, A., Goodpaster, B., Eagleton, J., Belle, S.,
Kalarchian, M., Lang, W., et al. (2014). Surgical vs
P. Salsberry et al.
medical treatments for type 2 diabetes mellitus: A
randomized clinical trial. JAMA Surgery, 7(149),
707–715.
Crume, T. L., Ogden, L. G., Mayer-Davis, E. J., Hamman,
R. F., Norris, J. M., Bischoff, K. J., et al. (2012). The
impact of neonatal breast-feeding on growth trajectories of youth exposed and unexposed to diabetes
in utero: The EPOCH study. International Journal of
Obesity, 36(4), 529–534.
Dabelea, D., Bell, R. A., D’Agostino, R. B., Jr.,
Imperatore, G., Johansen, J. M., Linder, B., et al.
(2007). Incidence of diabetes in youth in the United
States. JAMA, 297(24), 2716–2724.
Dabelea, D., Dolan, L. M., D’Agostino, R., Jr., Hernandez,
A. M., McAteer, J. B., Hammon, R. F., et al. (2011).
Association testing of TCF7L2 polymorphisms with
type 2 diabetes in multi-ethnic youth. Diabetologia,
54(3), 535–539.
Dabelea, D., Mayer-Davis, E. J., Saydah, S., Imperatore,
G., Linder, B., et al. (2014). Prevalence of type 1 and
type 2 diabetes among children and adolescents from
2001 to 2009. JAMA, 311(17), 1778–1786.
Danielsson, P., Kowalski, J., Ekblom, O., & Marcus, C.
(2012a). Response of severely obese children and adolescents to behavioral treatment. Archives of Pediatrics
& Adolescent Medicine, 166(12), 1103–1108.
Danielsson, P., Kowalski, J., Ekblom, O., & Marcus,
C. (2012b). Response of severely obese children
and adolescents to behavioral treatment. Archives
of Pediatrics & Adolescent Medicine, 166(12),
1103–1108.
Davis, C. L., Pollock, N. K., Waller, J. L., Allison, J. D.,
Dennis, B. A., Bassali, R., et al. (2012). Exercise
dose and diabetes risk in overweight and obese children: A randomized controlled trial. JAMA, 308(11),
1103–1112.
Demmer, R. T., Zuk, A. M., Rosenbaum, M., &
Desvarieux, M. (2013). Prevalence of diagnosed
and undiagnosed type 2 diabetes mellitus among US
adolescents: Results from the continuous NHANES,
1999-2010. American Journal of Epidemiology,
178(7), 1106–1113.
Devaraj, S., Hemarajata, P., & Versalovic, J. (2013).
The human gut microbiome and body metabolism: Implications for obesity and diabetes. Clinical
Chemistry, 59(4), 617–628.
Dyer, J. S., & Rosenfeld, C. R. (2011). Metabolic imprinting by prenatal, perinatal, and postnatal overnutrition:
A review. Seminars in Reproductive Medicine, 29(3),
266–276.
van Eijsden, M., Vrijkotte, T. G., Gemke, R. J., & van der
Wal, M. F. (2011). Cohort profile: The Amsterdam
born children and their development (ABCD) study.
International Journal of Epidemiology, 40(5),
1176–1186.
Ekelund, U., Ong, K. K., Linne, Y., Neovius, M., Brage,
S., Dunger, D. B., et al. (2007). Association of weight
gain in infancy and early childhood with metabolic risk in young adults. The Journal of Clinical
Endocrinology and Metabolism, 92(1), 98–103.
Pediatric Type 2 Diabetes: Prevention and Treatment Through a Life Course Health Development…
Elder, D. A., Herbers, P. M., Weis, T., Standiford, D.,
Woo, J. G., & D’Alessio, D. A. (2012). Beta-cell dysfunction in adolescents and adults with newly diagnosed type 2 diabetes mellitus. Journal of Pediatrics,
160(6), 904–910.
Eriksson, J. G. (2006). Early growth, and coronary heart
disease and type 2 diabetes: Experiences from the
Helsinki birth cohort studies. International Journal of
Obesity, 30(Suppl 4), S18–S22.
Eriksson, J. G. (2011). Early growth and coronary heart
disease and type 2 diabetes: Findings from the Helsinki
birth cohort study (HBCS). The American Journal of
Clinical Nutrition, 94(6 Supplement), 1799S–1802S.
Eriksson, J. G., Osmond, C., Kajantie, E., Forsen, T. J., &
Barker, D. J. (2006). Patterns of growth among children who later develop type 2 diabetes or its risk factors. Diabetologia, 49(12), 2853–2858.
Fabricius-Bjerre, S., Jensen, R. B., Faerch, K., Larsen, T.,
Molgaard, C., Michaelsen, K. F., et al. (2011). Impact
of birth weight and early infant weight gain on insulin
resistance and associated cardiovascular risk factors
in adolescence. PloS One, 6(6), e20595. doi:10.1371/
journal.pone.0020595.
Ferguson, M. A., Gutin, B., Le, N. A., Karp, W., Litaker,
M., Humphries, M., et al. (1999). Effects of exercise
training and its cessation on components of the insulin
resistance syndrome in obese children. International
Journal of Obesity and Related Metabolic Disorders,
23(8), 889–895.
Fernandez-Twinn, D. S., Wayman, A., Ekizoglou, S.,
Martin, M. S., Hales, C. N., & Ozanne, S. E. (2005).
Maternal protein restriction leads to hyperinsulinemia
and reduced insulin-signaling protein expression in
21-mo-old female rat offspring. American Journal of
Physiology. Regulatory, Integrative and Comparative
Physiology, 288(2), R368–R373.
Foster, G. D., Linder, B., Baranowski, T., Cooper, D. M.,
Goldberg, L., Harrell, J. S., et al. (2010). A schoolbased intervention for diabetes risk reduction. New
England Journal of Medicine, 363(5), 443–453.
Gabbe, S. G., Landon, M. B., Warren-Boulton, E., &
Fradkin, J. (2012). Promoting health after gestational
diabetes: A National Diabetes Education Program
call to action. Obstetrics and Gynecology, 119(1),
171–176.
Gaffney, K. F., Kitsantas, P., & Cheema, J. (2012). Clinical practice guidelines for feeding behaviors and
weight-for-age at 12 months: A secondary analysis of
the infant feeding practices study II. Worldviews on
Evidence- Based Nursing, 9(4), 234–242.
Gaffney, K. F., Kitsantas, P., Brito, A., & Swamidoss,
C. S. (2014). Postpartum depression, infant feeding
practices, and infant weight gain at six months of age.
Journal of Pediatric Health Care, 28(1), 43–50.
Garofano, A., Czernichow, P., & Breant, B. (1998). Betacell mass and proliferation following late fetal and
early postnatal malnutrition in the rat. Diabetologia,
41(9), 1114–1120.
Gillis, B., Mobley, C., Stadler, D. D., Hartstein, J., Virus,
A., Volpe, S. L., et al. (2009). Rationale, design and
methods of the HEALTHY study nutrition interven-
231
tion component. International Journal of Obesity,
33(Suppl 4), S29–S36.
Gillman, M. W., Oakey, H., Baghurst, P. A., Volkmer,
R. E., Robinson, J. S., & Crowther, C. A. (2010).
Effect of treatment of gestational diabetes mellitus on
obesity in the next generation. Diabetes Care, 33(5),
964–968.
Gluckman, P. D., Hanson, M. A., Cooper, C., & Thornburg,
K. L. (2008). Effect of in utero and early-life conditions on adult health and disease. New England
Journal of Medicine, 359(1), 61–73.
Groop, L. C., & Tuomi, T. (1997). Non-insulin-dependent
diabetes mellitus–a collision between thrifty genes
and an affluent society. Annals of Medicine, 29, 37–53.
Grote, V., Vik, T., von Kries, R., Luque, V., Socha, J.,
Verducci, E., et al. (2010). Maternal postnatal depression and child growth: A European cohort study. BMC
Pediatrics, 10, 14. doi:10.1186/1471-2431-10-14.
Hadar, E., & Hod, M. (2010). Establishing consensus criteria for the diagnosis of diabetes in pregnancy following the HAPO study. Annals of the New York Academy
of Sciences, 1205, 88–93.
Haines, L., Wan, K. C., Lynn, R., Barrett, T. G., & Shield,
J. P. (2007). Rising incidence of type 2 diabetes in
children in the U.K. Diabetes Care, 30(5), 1097–1101.
Haines, J., McDonald, J., O’Brien, A., Sherry, B., Bottino,
C. J., Schmidt, M. E., et al. (2013). Healthy habits,
happy homes: Randomized trial to improve household
routines for obesity prevention among preschool aged
children. JAMA Pediatrics, 167(11), 1072–1079.
Hales, C. N., & Barker, D. J. (2001). The thrifty phenotype hypothesis. British Medical Bulletin, 60, 5–20.
Halfon, N., & Forrest, C. B. (2017). The emerging theoretical framework of life course health development. In
N. Halfon, C. B. Forrest, R. M. Lerner, & E. Faustman
(Eds.), Handbook of life course health-development
science. Cham: Springer.
Harvey-Berino, J., & Rourke, J. (2003). Obesity prevention in preschool native-American children: A pilot
study using home visiting. Obesity Research, 11(5),
606–611.
Herbst, A., Kapellen, T., Schober, E., Graf, C., Meissner,
T., & Holl, R. (2014). Impact of regular physical activity on blood glucose control and cardiovascular risk
factors in adolescents with type 2 diabetes mellitus –
a multicenter study of 578 patients from 225 centres.
Pediatric Diabetes, 16(3), 204–210.
Hesketh, K. D., & Campbell, K. J. (2010). Interventions
to prevent obesity in 0-5 year olds: An updated systematic review of the literature. Obesity, 18(Supple 1),
S27–S35.
Hirst, K., Baranowski, T., DeBar, L., Foster, G. D.,
Kaufman, F., Kennel, P., et al. (2009). HEALTHY
study rationale, design and methods: Moderating risk
of type 2 diabetes in multi-ethnic middle school students. International Journal of Obesity, 33(Suppl 4),
S4–20.
Hiscock, H., Scalzo, K., Canterford, L., & Wake, M.
(2011). Sleep duration and body mass index in 0-7year olds. Archives of Disease in Childhood, 96(8),
735–739.
232
Hoet, J. J., Dahri, S., Snoeck, A., Reusens-Billen, B., &
Remacle, C. (1992). Importance of diets and their
effect on fetal development: Function and structure
of the endocrine pancreas following protein deficiency during intrauterine life. Bulletin et Mémoires
de l’Académie Royale de Médecine de Belgique, 147
(3–5), 174–181. discussion 81–83.
Imperatore, G., Boyle, J. P., Thompson, T. J., Case, D.,
Dabelea, D., Hamman, R. F., et al. (2012). Projections
of type 1 and type 2 diabetes burden in the U.S.
population aged <20 years through 2050: Dynamic
modeling of incidence, mortality, and population
growth. Diabetes Care, 35(12), 2515–2520.
ISPAD. (2011). Global IDF/ISPAD guideline for childhood and Pediatric diabetes. Brussels, Belgium:
International Diabetes Foundation.
James, D. C., & Lessen, R. (2009). Position of the
American dietetic association: Promoting and supporting breastfeeding. Journal of the American Dietetic
Association, 109(11), 1926–1942.
Jeffery, A. N., Metcalf, B. S., Hosking, J., Streeter, A. J.,
Voss, L. D., & Wilkin, T. J. (2012). Age before stage:
Insulin resistance rises before the onset of puberty:
A 9-year longitudinal study (EarlyBird 26). Diabetes
Care, 35(3), 536–541.
Johnson, C. L., & Versalovic, J. (2012). The human
microbiome and its potential importance to pediatrics.
Pediatrics, 129(5), 950–960.
Kanaka-Gantenbein, C. (2010). Fetal origins of adult diabetes. Annals of the New York Academy of Sciences,
1205, 99–105.
Kapoor, A., Dunn, E., Kostaki, A., Andrews, M. H.,
& Matthews, S. G. (2006). Fetal programming of
hypothalamo-pituitary-adrenal function: Prenatal
stress and glucocorticoids. The Journal of Physiology,
572(Pt 2), 31–44.
Katoh, S., Lehtovirta, M., Kaprio, J., Harjutsalo, V.,
Koskenvuo, M., Eriksson, J., et al. (2005). Genetic
and environmental effects on fasting and postchallenge plasma glucose and serum insulin values in
Finnish twins. Journal of Clinical Endocrinology and
Metabolism, 90(5), 2642–2647.
Kaufman, F. R., Hirst, K., Linder, B., Baranowski, T.,
Cooper, D. M., Foster, G. D., et al. (2009). Risk factors for type 2 diabetes in a sixth-grade multiracial
cohort: The HEALTHY study. Diabetes Care, 32(5),
953–955.
Kempf, K., Rathmann, W., & Herder, C. (2008). Impaired
glucose regulation and type 2 diabetes in children
and adolescents. Diabetes/Metabolism Research and
Reviews, 24, 427–437.
Kim, S. Y., England, J. L., Sharma, J. A., & Njoroge, T.
(2011). Gestational diabetes mellitus and risk of childhood overweight and obesity in offspring: A systematic review. Experimental Diabetes Research, 2011,
541308.
Kitzmann, K. M., Dalton, W. T., 3rd, Stanley, C. M.,
Beech, B. M., Reeves, T. P., et al. (2010). Lifestyle
interventions for youth who are overweight: A metaanalytic review. Health Psychology, 29(1), 91–101.
P. Salsberry et al.
Klingenberg, L., Christensen, L. B., Hjorth, M. F.,
Zangenberg, S., Chaput, J. P., Sjodin, A., et al. (2013).
No relation between sleep duration and adiposity indicators in 9-36 months old children: The SKOT cohort.
Pediatric Obesity, 8(1), e14–e18.
Knowlden, A. P., & Sharma, M. (2012). Systematic review
of family and home-based interventions targeting
paediatric overweight and obesity. Obesity Reviews,
13(6), 499–508.
Koren, O., Goodrich, J. K., Cullender, T. C., Spor, A.,
Laitinen, K., Backhed, H. K., et al. (2012). Host
remodeling of the gut microbiome and metabolic
changes during pregnancy. Cell, 150(3), 470–480.
Kramer, M. S., Matush, L., Vanilovich, I., Platt, R. W.,
Bogdanovich, N., Sevkovskaya, Z., et al. (2009). A
randomized breast-feeding promotion intervention did
not reduce child obesity in Belarus. The Journal of
Nutrition, 139(2), 417S–421S.
Lawrence, J., Mayer-Davis, E., Ryeynolds, K., Beyer, J.,
Pettitt, D., D’Agostino, R. B., et al. (2009a). Diabetes
in Hispanic American youths. Diabetes Care,
32(Supplement 2), S123–S132.
Lawrence, J. M., Mayer-Davis, E. J., Reynolds, K., Beyer,
J., Pettitt, D. J., D’Agostino, R. B., et al. (2009b).
Diabetes in Hispanic American youth: Prevalence,
incidence, demographics, and clinical characteristics:
The SEARCH for diabetes in youth study. Diabetes
Care, 32(Suppl 2), S123–S132.
Lee, Y. (2013). Diabetes care for emerging adults:
Transition from pediatric to adult diabetes care
systems. Annals of Pediatric Endocrinology &
Metabolism, 18, 106–110.
Ley, S. H., Hanley, A. J., Sermer, M., Zinman, B., &
O’Connor, D. L. (2012). Associations of prenatal
metabolic abnormalities with insulin and adiponectin
concentrations in human milk. The American Journal
of Clinical Nutrition, 95(4), 867–874.
Li, R., Bilik, D., Brown, M. B., Zhang, P., Ettner, S. L.,
Ackerman, R. T., et al. (2013). Medical costs associated with type 2 diabetes complications and comorbidities. The American Journal of Managed Care,
19(5), 421–430.
Liu, L. L., Yi, J. P., Beyer, J., Mayer-Davis, E. J., Dolan,
L. M., Dabelea, D. M., et al. (2009). Type 1 and type 2
diabetes in Asian and Pacific Islander U.S. youth: The
SEARCH for diabetes in youth study. Diabetes Care,
32(Suppl 2), S133–S140.
Liu, J., Yu, P., Qian, W., Li, Y., Zhao, J., Huan, F., et al.
(2013). Perinatal bisphenol a exposure and adult glucose homeostasis: Identifying critical windows of
exposure. PloS One, 8(5), e64143.
Luepker, R. V., Perry, C. L., McKinlay, S. M., Nader, P. R.,
Parcel, G. S., Stone, E. J., et al. (1996). Outcomes of
a field trial to improve children’s dietary patterns and
physical activity. The child and adolescent trial for
cardiovascular health. CATCH collaborative group.
The Journal of the American Medical Association,
275(10), 768–776.
Martin, R. M., Patel, R., Kramer, M. S., Vilchuck, K.,
Bogdanovich, N., Sergeichick, N., et al. (2014). Effects
Pediatric Type 2 Diabetes: Prevention and Treatment Through a Life Course Health Development…
of promoting longer-term and exclusive breastfeeding on cardiometabolic risk factors at age 11.5 years:
A cluster-randomized, controlled trial. Circulation,
129(3), 321–329.
Martin-Gronert, M. S., & Ozanne, S. E. (2012). Mechanisms underlying the developmental origins of disease. Reviews in Endocrine & Metabolic Disorders,
13(2), 85–92.
Maull, E. A., Ahsan, H., Edwards, J., Longnecker, M. P.,
Navas-Acien, A., Pi, J., et al. (2012). Evaluation of the
association between arsenic and diabetes: A National
Toxicology Program workshop review. Environmental
Health Perspectives, 120(12), 1658–1670.
May, A. L., Kuklina, E. V., & Yoon, P. W. (2012).
Prevalence of cardiovascular disease risk factors
among US adolescents, 1999–2008. Pediatrics, 129(6),
1035–1041.
Mayer-Davis, E. J., Rifas-Shiman, S. L., Zhou, L., Hu,
F. B., Colditz, G. A., & Gillman, M. W. (2006). Breastfeeding and risk for childhood obesity: Does maternal diabetes or obesity status matter? Diabetes Care,
29(10), 2231–2237.
Mayer-Davis, E. J., Dabelea, D., Lamichhane, A. P.,
D’Agostino, R. B., Jr., Liese, A. D., et al. (2008).
Breast-feeding and type 2 diabetes in the youth of
three ethnic groups: The SEARCh for diabetes in
youth case-control study. Diabetes Care, 31(3),
470–475.
Mayer-Davis, E. J., Beyer, J., Bell, R. A., Dabelea, D.,
D’Agostino, R., Jr., Imperatorek, G., et al. (2009).
Diabetes in African American youth: Prevalence, incidence, and clinical characteristics: The SEARCH for
diabetes in youth study. Diabetes Care, 32(Supplement
2), S112–S122.
McEwen, B. S. (2008). Understanding the potency of
stressful early life experiences on brain and body function. Metabolism Clinical and Experimental, 57(Suppl
2), S11–S15.
McLearn, K. T., Minkovitz, C. S., Strobino, D. M., Marks,
E., & Hou, W. (2006). Maternal depressive symptoms
at 2 to 4 months post partum and early parenting practices. Archives of Pediatrics & Adolescent Medicine,
160(3), 279–284.
McMillen, I. C., & Robinson, J. S. (2005). Developmental
origins of the metabolic syndrome: Prediction, plasticity, and programming. Physiological Reviews, 85(2),
571–633.
McMurray, R. G., Bassin, S., Jago, R., Bruecker, S., Moe,
E. L., Murray, T., et al. (2009). Rationale, design and
methods of the HEALTHY study physical education
intervention component. International Journal of
Obesity, 33(Suppl 4), S37–S43.
Meaney, M. J., Szyf, M., & Seckl, J. R. (2007). Epigenetic mechanisms of perinatal programming of
hypothalamic-pituitary-adrenal function and health.
Trends in Molecular Medicine, 13(7), 269–277.
Metzger, B. E., Lowe, L. P., Dyer, A. R., Trimble,
E. R., Chaovarindr, U., Coustan, D. R., et al. (2008).
Hyperglycemia and adverse pregnancy outcomes.
233
The New England Journal of Medicine, 358(19),
1991–2002.
Metzger, B. E., Gabbe, S. G., Persson, B., Buchanan, T. A.,
Catalano, P. A., Damm, P., et al. (2010). International
association of diabetes and pregnancy study groups
recommendations on the diagnosis and classification
of hyperglycemia in pregnancy. Diabetes Care, 33(3),
676–682.
Morandi, A., Maschio, M., Marigliano, M., Miraglia
Del Giudice, E., Moro, B., Peverelli, P., et al. (2014).
Screening for impaired glucose tolerance in obese
children and adolescents: A validation and implementation study. Pediatric Obesity, 9(1), 17–25.
Morgan, A. R. (2012). Determining genetic risk factors for pediatric type 2 diabetes. Current Diabetes
Reports, 12(1), 88–92.
Morris, D. H., Khunti, K., Achana, F., Srinivasan, B.,
Gray, L. J., Davies, M. J., et al. (2013). Progression
rates from HbA(1c) 6.0-6.4% and other prediabetes definitions to type 2 diabetes: A meta-analysis.
Diabetologia, 56(7), 1489–1493.
Morrison, K. M., Xu, L., Tarnopolsky, M., Yusuf, Z.,
Atkinson, S. A., & Yusuf, S. (2012). Screening for dysglycemia in overweight youth presenting for weight
management. Diabetes Care, 35(4), 711–716.
Moses, R. G. (2010). New consensus criteria for GDM:
Problem solved or a pandora’s box? Diabetes Care,
33(3), 690–691.
Navas-Acien, A., Silbergeld, E. K., Pastor-Barriuso, R., &
Guallar, E. (2008). Arsenic exposure and prevalence of
type 2 diabetes in US adults. JAMA, 300(7), 814–822.
Neville, M. C., Anderson, S. M., McManaman, J. L.,
Badger, T. M., Bunik, M., Contractor, N., et al. (2012).
Lactation and neonatal nutrition: Defining and refining the critical questions. Journal of Mammary Gland
Biology and Neoplasia, 17(2), 167–188. doi:10.1007/
s10911-012-9261-5.
Ong, K. K. (2010). Early determinants of obesity.
Endocrine Development, 19, 53–61.
Pervanidou, P., & Chrousos, G. P. (2011). Stress and
obesity/metabolic syndrome in childhood and adolescence. International Journal of Pediatric Obesity,
6(Supplement 1), 21–28.
Pervanidou, P., & Chrousos, G. P. (2012). Metabolic consequences of stress during childhood and adolescence.
Metabolism, 61(5), 611–619.
Peters, A., & Laffel, L. (2011). Diabetes care for emerging
adults: Recommendations for transition from pediatric
to adult diabetes care systems: A position statement
of the American Diabetes Association. Diabetes Care,
34(11), 2477–2485.
Plagemann, A., Harder, T., Franke, K., & Kohlhoff, R.
(2002). Long-term impact of neonatal breast-feeding
on body weight and glucose tolerance in children of
diabetic mothers. Diabetes Care, 25(1), 16–22.
Prince, A. L., Antony, K. M., Ma, J., & Aagaard, K. M.
(2014). The microbiome and development: A mother’s perspective. Seminars in Reproductive Medicine,
32(1), 14–22.
234
Raman, A., Ritchie, L. D., Lustig, R. H., Fitch, M. D.,
Hudes, M. L., & Fleming, S. E. (2010). Insulin resistance is improved in overweight African American
boys but not in girls following a one-year multidisciplinary community intervention program. Journal
of Pediatric Endocrinology & Metabolism, 23(1–2),
109–120.
Rhodes, E. T., Prosser, L. A., Hoerger, T. J., Lieu, T.,
Ludwig, D. S., & Laffel, L. M. (2012). Estimated
morbidity and mortality in adolescents and young
adults diagnosed with type 2 diabetes mellitus.
Diabetic Medicine, 29(4), 453–463.
Ritchie, L. D., Sharma, S., Ikeda, J. P., Mitchell, R. A.,
Raman, A., Green, B. S., et al. (2010). Taking action
together: A YMCA-based protocol to prevent type-2
diabetes in high-BMI inner-city African American
children. Trials, 11, 60.
Robinson, S., Ntani, G., Simmonds, S., Syddall, H.,
Dennison, E., Sayer, A. A., et al. (2013). Type of milk
feeding in infancy and health behaviours in adult life:
Findings from the Hertfordshire cohort study. The
British Journal of Nutrition, 109(6), 1114–1122.
Rolland-Cachera, M. F. (2011). Childhood obesity:
Current definitions and recommendations for their
use. International Journal of Pediatric Obesity, 6(5–
6), 325–331.
Rosenbaum, M., Nonas, C., Weil, R., Horlick, M.,
Fennoy, I., Vargas, I., et al. (2007). School-based
intervention acutely improves insulin sensitivity and
decreases inflammatory markers and body fatness in
junior high school students. The Journal of Clinical
Endocrinology and Metabolism, 92(2), 504–508.
Rosenbloom, A. L., Joe, J. R., Young, R. S., & Winter,
W. E. (1999). Emerging epidemic of type 2 diabetes in
youth. Diabetes Care, 22(2), 345–354.
Rosenbloom, A. L., Silverstein, J. H., Amemiya, S.,
Zeitler, P., & Klingensmith, G. J. (2008). ISPAD clinical practice consensus guidelines 2006-2007. Type 2
diabetes mellitus in the child and adolescent. Pediatric
Diabetes, 9(5), 512–526.
Saelens, B. E., Grow, H. M., Stark, L. J., Seeley, R. J.,
& Roehrig, H. (2011). Efficacy of increasing physical activity to reduce children’s visceral fat: A pilot
randomized controlled trial. International Journal of
Pediatric Obesity, 6(2), 102–112.
Salsberry, P. J., & Reagan, P. B. (2005). Dynamics of
early childhood overweight. Pediatrics, 116(6),
1329–1338.
Samuelsson, A. M., Matthews, P. A., Argenton, M.,
Christie, M. R., McConnell, J. M., et al. (2008). Dietinduced obesity in female mice leads to offspring
hyperphagia, adiposity, hypertension, and insulin
resistance: A novel murine model of developmental
programming. Hypertension, 51(2), 383–392.
Savoye, M., Shaw, M., Dziura, J., Tamborlane, W. V.,
Rose, P., Guandalini, C., et al. (2007). Effects of
a weight management program on body composition and metabolic parameters in overweight children: A randomized controlled trial. JAMA, 297(24),
2697–2704.
P. Salsberry et al.
Savoye, M., Nowicka, P., Shaw, M., Yu, S., Dziura, J.,
Chavent, G., et al. (2011). Long-term results of an
obesity program in an ethnically diverse pediatric
population. Pediatrics, 127(3), 402–410.
Savoye, M., Caprio, S., Dziura, J., Camp, A., Germain,
G., Summers, C., et al. (2014). Reversal of early
abnormalities in glucose metabolism in obese youth:
Results of an intensive lifestyle randomized controlled
trial. Diabetes Care, 37(2), 317–324.
Schore, A. N. (2005). Back to basics: Attachment, affect
regulation, and the developing right brain: Linking
developmental neuroscience to pediatrics. Pediatrics
in Review, 26(6), 204–217.
Sebert, S., Sharkey, D., Budge, H., & Symonds, M. E.
(2011). The early programming of metabolic health:
Is epigenetic setting the missing link? The American
Journal of Clinical Nutrition, 94(6 Supplement),
1953S–1958S.
Seki, Y., Williams, L., Vuguin, P. M., & Charron, M. J.
(2012). Minireview: Epigenetic programming of diabetes and obesity: Animal models. Endocrinology,
153(3), 1031–1038.
Shaw, J. E., Socree, R. A., & Zimmet, P. Z. (2010). Global
estimates of the prevalence of diabetes for 2010 and
2030. Diabetes Research and Clinical Practice, 87,
4–14.
Shonkoff, J. P., & Garner, A. S. (2012). The lifelong
effects of early childhood adversity and toxic stress.
Pediatrics, 129(1), e232–e246.
Silverman, B. L., Rizzo, T. A., Cho, N. H., & Metzger,
B. E. (1998). Long-term effects of the intrauterine
environment. The Northwestern University diabetes
in pregnancy Center. Diabetes Care, 21(Suppl 2),
B142–B149.
Simeoni, U., & Barker, D. J. (2009). Offspring of diabetic
pregnancy: Long-term outcomes. Seminars in Fetal &
Neonatal Medicine, 14(2), 119–124.
Sinclair, K. D., Lea, R. G., Rees, W. D., & Young, L. E.
(2007). The developmental origins of health and disease: Current theories and epigenetic mechanisms.
Society of Reproduction and Fertility Supplement, 64,
425–443.
Sinha, R., Fisch, G., Teague, B., Tamborlane, W. V.,
Banyas, B., Allen, K., et al. (2002). Prevalence of
impaired glucose tolerance among children and adolescents with marked obesity. The New England
Journal of Medicine, 346(11), 802–810.
Skouteris, H., McCabe, M., Swinburn, B., Newgreen, V.,
Sacher, P., & Chadwick, P. (2011). Parental influence
and obesity prevention in pre-schoolers: A systematic review of interventions. Obesity Reviews, 12(5),
315–328.
Skouteris, H., McCabe, M., Ricciardelli, L., Milgrom, J.,
Baur, L., Aksan, N., et al. (2012). Parent–child interactions and obesity prevention: A systematic review
of the literature. Early Child Development and Care,
182(2), 153–174.
Smith, B. T., Lynch, J. W., Fox, C. S., Harper, S.,
Abrahamowicz, M., Almeida, N. D., et al. (2011).
Life-course socioeconomic position and type 2 dia-
Pediatric Type 2 Diabetes: Prevention and Treatment Through a Life Course Health Development…
betes mellitus: The Framingham offspring study.
American Journal of Epidemiology, 173(4), 438–447.
Snieder, H., Boomsma, D. I., van Doornen, L. J., & Neale,
M. C. (1999). Bivariate genetic analysis of fasting
insulin and glucose levels. Genetic Epidemiology,
16(4), 426–446.
Stark, L. J., Clifford, L. M., Towner, E. K., Filigno,
S. S., Zion, C., Bolling, C., et al. (2014). A pilot
randomized controlled trial of a behavioral familybased intervention with and without home visits to
decrease obesity in preschoolers. Journal of Pediatric
Psychology, 39(9), 1001–1012. doi:10.1093/jpepsy/
jsu059.
Szajewska, H. (2013). Microbiota modulation: Can probiotics prevent/treat disease in pediatrics? Nestle
Nutrition Institute Workshop Series, 77, 99–110.
Tamayo, T., Christian, H., & Rathmann, W. (2010).
Impact of early psychosocial factors (childhood socioeconomic factors and adversities) on future risk of
type 2 diabetes, metabolic disturbances and obesity:
A systematic review. BMC Public Health, 10, 525.
doi:10.1186/1471-2458-10-525.
Tamayo, T., Jacobs, D. R., Jr., Strassburger, K., Giani, G.,
Seeman, T. E., Matthews, K., et al. (2012). Race- and
sex-specific associations of parental education with
insulin resistance in middle-aged participants: The
CARDIA study. European Journal of Epidemiology,
27(5), 349–355.
Tarry-Adkins, J. L., & Ozanne, S. E. (2011). Mechanisms
of early life programming: Current knowledge and
future directions. The American Journal of Clinical
Nutrition, 94(6 Supplement), 1765S–1771S.
Taveras, E. M., Scanlon, K. S., Birch, L., Rifas-Shiman,
S. L., Rich-Edwards, J. W., & Gillman, M. W. (2004).
Association of breastfeeding with maternal control
of infant feeding at age 1 year. Pediatrics, 114(5),
e577–e583.
Taveras, E. M., Rifas-Shiman, S. L., Oken, E., Gunderson,
E. P., & Gillman, M. W. (2008). Short sleep duration in
infancy and risk of childhood overweight. Archives of
Pediatrics & Adolescent Medicine, 162(4), 305–311.
Taveras, E. M., Gillman, M. W., Pena, M. M., Redline, S.,
& Rifas-Shiman, S. L. (2014). Chronic sleep curtailment and adiposity. Pediatrics, 133(6), 1013–1022.
Thayer, K. A., Heindel, J. J., Bucher, J. R., & Gallo, M. A.
(2012). Role of environmental chemicals in diabetes
and obesity: A National Toxicology Program workshop review. Environmental Health Perspectives,
120(6), 779–789.
Thompson, A. L., & Bentley, M. E. (2013). The critical
period of infant feeding for the development of early
disparities in obesity. Social Science & Medicine, 97,
288–296.
Tounian, P. (2011). Programming towards childhood obesity. Annals of Nutrition & Metabolism, 58(Suppl 2),
30–41.
Trevino, R. P., Yin, Z., Hernandez, A., Hale, D. E., Garcia,
O. A., & Mobley, C. (2004). Impact of the Bienestar
school-based diabetes mellitus prevention program
on fasting capillary glucose levels: A randomized
235
controlled trial. Archives of Pediatrics & Adolescent
Medicine, 158(9), 911–917.
Venditti, E. M., Elliot, D. L., Faith, M. S., Firrell, L. S.,
Giles, C. M., Goldberg, L., et al. (2009). Rationale,
design and methods of the HEALTHY study behavior intervention component. International Journal of
Obesity, 33(Suppl 4), S44–S51.
Venkat Narayan, K. M., Boyle, J. P., Thompson, T. J.,
Sorensen, S. W., & Willimason, D. F. (2003). Lifetime
risk for riabetes mellitus in the United States. JAMA,
290(14), 1884–1889.
Versalovic, J. (2013). The human microbiome and probiotics: Implications for pediatrics. Annals of Nutrition
& Metabolism, 63(Suppl 2), 42–52.
Visscher, P. M., Hill, W. G., & Wray, N. R. (2008).
Heritability in the genomics era–concepts and misconceptions. Nat Rev Genet, 9(4), 255–266.
Wabitsch, M., Hauner, H., Hertrampf, M., Muche,
R., Hay, B., Mayer, H., et al. (2004). Type II diabetes mellitus and impaired glucose regulation in
Caucasian children and adolescents with obesity living in Germany. International Journal of Obesity and
Related Metabolic Disorders, 28(2), 307–313.
Waitzfelder, B., Pihoker, C., Klingensmith, G., Case, D.,
Anderson, A., Bell, R. A., et al. (2011). Adherence
to guidelines for youths with diabetes mellitus.
Pediatrics, 128(3), 531–538.
Wang, L., Anderson, J. L., Dalton, W. T., Wu, T., Liu, X.,
& Zheng, S. (2013). Maternal depressive symptoms
and the risk of overweight in their children. Maternal
and Child Health Journal, 17(5), 940–948.
Warner, M. J., & Ozanne, S. E. (2010). Mechanisms
involved in the developmental programming of adulthood disease. The Biochemical Journal, 427(3),
333–347.
Weiss, R., Taksali, S. E., Tamborlane, W. V., Burgert,
T. S., Savoye, M., & Caprio, S. (2005). Predictors of
changes in glucose tolerance status in obese youth.
Diabetes Care, 28(4), 902–909.
Wen, L. M., Baur, L. A., Simpson, J. M., Rissel, C.,
Wardle, K., & Flood, V. M. (2012). Effectiveness of
home based early intervention on children’s BMI at
age 2: Randomised controlled trial. BMJ, 344, e3732.
doi:10.1136/bmj.e3732.
West, N. A., Hamman, R. F., Mayer-Davis, E. J.,
D’Agostino, R. B., Jr., Marcovina, S. M., Liese,
A. S., et al. (2009). Cardiovascular risk factors among
youth with and without type 2 diabetes: Differences
and possible mechanisms. Diabetes Care, 32(1),
175–180.
Whitlock, E. P., O’Connor, E. A., Williams, S. B., Beil,
T. L., & Lutz, K. W. (2010). Effectiveness of weight
management interventions in children: A targeted
systematic review for the US preventive service task
Force. Pediatrics, 125(2), e396–e418.
Whitmore, T. J., Trengove, N. J., Graham, D. F., &
Hartmann, P. E. (2012). Analysis of insulin in human
breast milk in mothers with type 1 and type 2 diabetes
mellitus. International Journal of Endocrinology,
2012, 296368. doi:10.1155/2012/296368.
236
Zeitler, P. (2010). Approach to the obese adolescent with new-onset diabetes. The Journal of
Clinical Endocrinology and Metabolism, 95(12),
5163–5170.
Zeitler, P., Hirst, K., Pyle, L., Linder, B., Copeland, K.,
Arslanian, S., et al. (2012). A clinical trial to maintain
P. Salsberry et al.
glycemic control in youth with type 2 diabetes. New
England Journal of Medicine, 366(24), 2247–2256.
Zeitler, P., Tandon, N., Nadeau, K., Urakami, T., Barrett,
T., & Maahs, D. (2014). Type 2 diabetes in the child
and adolescent. Pediatric Diabetes, 20(Supplement),
20–26.
Open Access This chapter is licensed under the terms of the Creative Commons Attribution 4.0 International License
(http://creativecommons.org/licenses/by/4.0/), which permits use, sharing, adaptation, distribution and reproduction in
any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license and indicate if changes were made.
The images or other third party material in this chapter are included in the chapter’s Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the chapter’s Creative Commons license
and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain
permission directly from the copyright holder.
Life Course Health Development
in Autism Spectrum Disorders
Irene E. Drmic, Peter Szatmari, and Fred Volkmar
1
Introduction
Health development results from dynamic personenvironment interactions that begin before conception and continue throughout the lifespan
(Halfon et al. 2014; Russ et al. 2014). Life course
health development (LCHD) incorporates theories and empirical evidence from the biological,
physical, and social sciences to formulate a framework that explains how health develops over the
life course. The principles of life course health
development (see Halfon and Forrest 2017)
describe health as an emergent set of capacities
that develop continuously over the lifespan in
complex, nonlinear processes that result from
multilevel interactions of individuals and their
environments. Furthermore, health is sensitive to
the timing and social structuring of environmental
exposures, as well as to timing and synchronization
I.E. Drmic (*)
Hospital for Sick Children, Toronto, ON, Canada
e-mail: Irene.drmic@sickkids.ca
P. Szatmari
Centre for Addiction and Mental Health,
Hospital for Sick Children, University of Toronto,
Toronto, ON, Canada
e-mail: Peter.Szatmari@utoronto.ca
F. Volkmar
Child Study Center, Yale University School
of Medicine, New Haven, CT, USA
e-mail: Fred.Volkmar@yale.edu
of molecular, physiological, behavioral, social,
and cultural functions. Finally, evolution enables
and constrains the range of adaptive plastic
responses and health developmental pathways
associated with these phenotypes.
The life course health development conceptual framework can be applied to autism spectrum disorders (ASD) to help prioritize a research
agenda and improve health development across
the lifespan for individuals with ASD, their families, and communities. Novel approaches to
understanding ASD are welcome given that ASD
represents a major public health challenge. The
estimated annual cost of caring for individuals
with ASD in the USA is $137 billion, with a lifetime cost per individual estimated to be $2.4 million for those with co-occurring intellectual
disability and $1.4 million for those without
intellectual disability (Dawson and Bernier
2013). The following chapter is not an exhaustive
review of the literature in ASD; rather, we highlight some issues and findings that are pertinent
to an understanding of ASD using the lens of the
life course health development principles.
1.1
What Is Autism Spectrum
Disorder (ASD): The Clinical
Phenotype
Autism spectrum disorders are a group of
neuropsychiatric conditions all of which share
© The Author(s) 2018
N. Halfon et al. (eds.), Handbook of Life Course Health Development,
DOI 10.1007/978-3-319-47143-3_11
237
238
difficulties in social engagement and social interaction as their major diagnostic feature. As a
result of this basic problem, difficulties arise in
learning, cognitive organization, attention, and
adaptive skills. Autism, the prototypic disorder,
was described by Kanner in 1943 as an “inborn
disturbance” of social contact. Autism was first
included in a new category of pervasive developmental disorders when first recognized officially
in the third edition of Diagnostic and Statistical
Manual of Mental Disorders (DSM;APA 1980).
Autism is a condition of early onset and poses
multiple problems in health development. Many
parents are concerned about affected children’s
development by age 1 with the vast majority of
parents worried by age 2 (Chawarska et al. 2008).
An increasing body of work has focused on
autism as it first develops in infancy (Rogers
2009). In a minority of cases (about 20%), parents report some period of normal development
followed by loss of skills; the clinical significance of regression continues to be debated (Parr
et al. 2011; Shattuck et al. 2009).
Problems in social interaction are a hallmark
of autism and related conditions. Social difficulties are distinctive even considering any associated
developmental
delays.
Similarly,
communication problems are present and persistent even in the more cognitively able children
who lack verbal and nonverbal social communication skills such as gestures, facial expression,
and eye contact. Even when children do speak,
they exhibit a number of difficulties including
echolalia, pronoun use difficulties, abnormal
prosody, and in the social use of language (Paul
2008). Kanner noted the range of unusual behaviors frequent in autism including motor mannerisms (stereotyped movement), difficulties with
change, and insistence on routines. The apparent
over-engagement with the nonsocial world arises
in the general lack of social engagement. This
suggests a potential route both as a mechanism
for learning difficulties and for intervention.
The original impression that autism was not
associated with cognitive problems rested, in
large part, on the unusual abilities of children in
some areas; in some cases, these skills are highly
circumscribed and may arise even in association
with overall intellectual delay (Hermelin 2001).
I.E. Drmic et al.
Even for the most able individuals, scatter in abilities is often present with a relative deficit in verbal as opposed to perceptual abilities. It is also
the case that there is frequently a gap between
abilities in a formal testing situation and “realworld” or adaptive skills – the latter reflecting the
significant difficulties individuals have in generalization and in tasks that involved fast-paced
decision making, executive functioning, and
social interaction (Robinson et al. 2009). In more
classical autism, nonverbal skills tend to be much
stronger than verbal abilities; the opposite profile
may hold true for Asperger’s disorder although
this has been debated (Klin et al. 1996; Meyer
and Minshew 2002; Noterdaeme et al. 2010).
As noted above in addition to classic autism of
the type described by Kanner, a number of other
conditions with some apparent similarity to
autism have been recognized, although their
validity apart from autism continues to be hotly
debated. Asperger’s disorder was described
(Asperger 1944) the year after Kanner’s report
first appeared. Although the children (all boys)
had major problems in social interaction, in contrast to Kanner’s (1943) cases, they demonstrated
good language abilities and were more likely to
exhibit motor difficulties, unusual and allconsuming esoteric special interests, and their
problems seemed to come to clinical attention
rather later (Woodbury-Smith and Volkmar
2009). Although included in DSM-IV and
International Statistical Classification of
Diseases and Related Health Problems (ICD)-10
as a distinctive disorder, Asperger’s disorder has
been the focus of much debate and is not considered a diagnosable disorder in DSM-V.
A subthreshold pervasive developmental disorder category was included in DSM since 1980
to encompass individuals whose difficulties
appear to fall close to autism but do not achieve
thresholds for diagnosis of that, or related, conditions. Interestingly this concept also was
described in the 1940s although not with great
precision. It has as always been recognized to
significantly outnumber more classical autism
(Fombonne 2005a, b) by probably four or five
times although research in this area has, paradoxically, been limited. In recent years, the tendency
to equate pervasive developmental disorder not
Life Course Health Development in Autism Spectrum Disorders
otherwise specified with more classic autism has
led to a broader autism spectrum concept
although it must be emphasized that the DSM-5
autism spectrum disorder concept would likely
exclude many cases that in the past had been said
to have pervasive developmental disorder not
otherwise specified (Volkmar and McPartland
2014).
1.2
Epidemiology
It is clear that ASD has a strong genetic component and is likely related to the interaction of
multiple genes (Devlin and Scherer 2012); ASD
invariably originates in the first years of life.
Poverty and lack of access to adequate resources
and intervention clearly impact identification,
service provision, and likely outcome (Liptak
et al. 2008; Thomas et al. 2007), but this area of
work remains one that is relatively little studied.
Various factors complicate the study of prevalence rates in ASD. There has been a concern that
rates have risen over time although in his review
of the topic Fombonne (2005a, b) suggests that a
median rate of strictly defined autism would be
about 1 per 800 to 1000 people with the much
broader pervasive developmental disorder category being much more common. Several recent
studies have reported higher rates (e.g., Brugha
et al. 2011; Elsabbagh et al. 2012), but studies of
adults in community settings report similar rates
to those estimated by Fombonne and others
(Brugha et al. 2011; Elsabbagh et al. 2012; Rutter
1973) and various factors which might account
for apparent increases in rates have been identified (King and Bearman 2009; Rutter 1982) with
potential overestimation due to methodology
(i.e., case-confirmation strategies) and small
sample sizes resulting in high estimates (Levy
et al. 2009). Such factors include increased public awareness and increased recognition of more
cognitively able individuals, applying the diagnosis to individuals with specific syndromes such
as Down syndrome and widening of diagnostic
criteria, sundry methodological problems, and
“diagnostic substitution” – the latter referring to
an issue, particularly in the USA, where an edu-
239
cational label of autism may be used to obtain
services in preference to other labels such as
mental retardation (Fombonne 2005a, b).
Most studies have reported significantly
higher rates of ASD in boys than in girls although
girls with more strictly defined autism are also
more likely to have intellectual disability
(Fombonne 2005a, b). The three- to fivefold
increase in boys is similar to other neurodevelopmental disorders such as language disorders and
attention deficit hyperactivity disorder (ADHD).
It is possible that milder expression in some girls
is underdiagnosed (Landa et al. 2007). There has
been speculation that greater severity sometimes
observed in girls is a function of a greater genetic
risk for the condition (Fombonne 2005a, b) and
that observed sex differences might relate to fundamental cognitive mechanisms (Baron-Cohen
2011). In reality, sex differences have been
remarkably little studied and our understanding
of this phenomenon remains very limited.
Potential social class correlates of autism first
noted by Kanner (1943) in his original sample
have not generally held up in subsequent research,
presumably reflecting a referral bias in Kanner’s
original cases (Fombonne 2005a, b; Wing 1980).
However, disparities in diagnosis rates are
observed with children from backgrounds of poverty or minority status often going undiagnosed
(LaFramboise et al. 2009). Although diagnostic
presentation is rather similar around the world,
cultural factors may impact both diagnosis and
treatment (Brown and Rogers 2003).
1.3
Classification and Diagnosis
Several approaches were undertaken in the late
1970s to provide more operational definitions of
autism. Rutter’s approach (1978) emphasized
difficulties in social interaction, communication,
and unusual behaviors of early onset that were
not solely attributable to mental retardation. Both
family genetic and longitudinal studies provided
important information on the stability and validity of the diagnosis apart from other conditions
and led to the inclusion of autism in DSM-III
(APA 1980). The DSM-III approach was modified
I.E. Drmic et al.
240
in 1987 when DSM-III-R appeared and yet again
with DSM-IV which appeared in 1994. The
DSM-IV definition was based on results of a
large international field trial and became consistent with the tenth revision of the ICD leading to
two decades of diagnostic stability (Volkmar and
McPartland 2014). For DSM-5, a number of
changes were made and have proven controversial. The adoption of the term autism spectrum
disorder seems well justified; however, the criteria proposed appear to really be more relevant to
classic “Kanner’s autism,” and many individuals,
particularly the more cognitively able, would
likely lose their diagnosis (Volkmar and
McPartland 2014). As a result, cases with “wellestablished” diagnosis of Asperger’s or pervasive
developmental disorder not otherwise specified
(PDD-NOS; in DSM-IV terms) could retain an
ASD diagnosis – thus effectively creating two
diagnostic systems in current use; the advent of
ICD-11 and the Research Domain Criteria
(RDoC) approach suggest that for some time
there may be considerable controversy about definitional issues (see Baker et al. 2012; Brand
et al. 2012; Mattilla et al. 2011; McGrew et al.
2012; Watts 2012; Huerta et al. 2012).
2
Pathogenesis
2.1
Hereditability
ASD is now considered a disorder caused at least
in part by genetic factors (Szatmari et al. 1998).
The evidence for this comes from studies showing that ASD runs within families, from twin
studies that show that this familial aggregation is
due to genetic and possibly to shared environmental risk factors (Sullivan et al. 2012) and to
the recent identification of several different networks of genes responsible for synaptic and axonal development and neuronal migration
(Malhotra and Sebat 2012).
Early family studies looked at the recurrence
risk to a sibling if one child in the family had
ASD. Those sibling risks were around 2% which
were considered to be very much higher than the
general population rate estimated at that time to
be 4 per 10,000 (Szatmari et al. 1998). With better study designs and with a better appreciation of
the breadth of the ASD phenotype, these family
studies now suggest that the risk to sibling is
closer to 9% (Ritvo et al. 1989). This needs to be
seen in the context thought of more recent prevalence estimates of ASD of around 1%. Thus, it
seems the sibling recurrence risk is roughly ten
times the population base rate (Constantino et al.
2010).
The baby sibs paradigm provides a more valid
estimate of sibling recurrence risk. In these studies, the infant sibling of an older child with ASD
is followed from birth to 3 years of age. In this
context, the emergence of autistic signs and
symptoms can be carefully documented prospectively, and the presence or absence of an ASD
diagnosis is determined by 3 years of age. These
studies have shown that signs and symptoms of
the ASD phenotype begin to emerge around
6–12 months of age, and upward of 20% of siblings will have a diagnosis of ASD at 3 years of
age (Chawarska et al. 2008; Zwaigenbaum et al.
2012). Interestingly, despite the fact that these
studies have demonstrated that ASD can be diagnosed as early as 3 years and that best practice in
ASD, such as the 2007 American Academy of
Pediatrics guideline that all children should be
screened for ASD at 18 and 24 months, the average age of diagnosis in the USA is still close to
5 years and higher in those with milder forms of
ASD (CDC 2014).
Twin studies are used to determine the mechanism of familial aggregation. Earlier studies
suggested very sharp discrepancies between
monozygous and dizygous concordance rates
suggesting not only that the mechanism of heritability was genetic but that there was little
room for environmental risk factors in playing
an important role in causation. The twin study
with the largest sample size and the one carried
out with the most methodological rigor was
recently published (Hallmayer et al. 2011). In
that study, monozygous concordance rates were
closer to 60%, and dizygous rates were 20% rate
seen in the recent infant sibling studies. While
the Hallmayer et al. twin study also provides
support for the importance of genetic factors in
Life Course Health Development in Autism Spectrum Disorders
the etiology of the disorder, the higher than
expected dizygous concordance rate also suggests a significant role for shared environmental
risk factors.
2.2
Genetic Contribution
Hundreds of different genes have been described
in ASD that have been disrupted by either deletions or duplications of copy number. These
genes all appear to be related to different aspects
of neuronal development (largely synaptic functioning) or neuronal migration (Li et al. 2012).
This is an important finding as it steers mechanisms of etiology away from neurotransmitters
and monoamines, a theory which had dominated
the field for several decades (McDougle et al.
2005).
With the discovery of copy number variants as
important structural genomic risk factors for a
variety of human diseases has come the realization that these variants might play an important
role in the etiology of ASD (Marshall et al. 2008).
Copy number variants represent deletions or
duplications covering more than 1000 kbs of
DNA. In general, copy number variants can be
common or rare, and they can be either de novo
or inherited from parents. The first copy number
variant identified in ASD in a genome-wide study
was a rare deletion in the neurexin gene (Szatmari
et al. 2007), and this has been followed up with
many reports of other copy number variants
affecting important brain-expressed genes (for a
review see Malhotra and Sebat 2012). These
copy number variants tend to be extremely rare,
found in less than 1% of the ASD samples, and
tend to rise de novo. This would explain the frequent lack of family history in ASD families and
would provide a mechanism whereby the prevalence of the disorder remains stable in the context
of a condition associated with reduced fertility
(Power et al. 2012). Common genetic variants
(either polymorphisms or copy number variants)
have not been reliably identified in ASD, although
such variants likely do exist and will require very
large sample sizes for their identification (Devlin
et al. 2011). If they do exist, their effect size must
241
be very small or else they operate as modifying
factors as opposed to susceptibility genes.
Currently, copy number variants explain less than
10% of cases in ASD, and the population attributable risk is less than 5% (Pinto et al. 2010). The
etiology of the vast majority of cases of ASD
remains to be explained. The next generation of
genetic studies will focus on much smaller copy
number variants or DNA sequence variants identified either in exomes or in whole genome
sequencing.
2.3
Nongenetic Contributions
The search for environmental risk factors has
been underway for many decades. There seems
reasonably good evidence that certain maternal
anticonvulsants such as carbamazepine, valproic
acid, and perhaps other anticonvulsants increase
risk for ASD (Palac and Meador 2011). Other
putative environmental risk factors that may have
been more common in earlier decades such as
congenital rubella have now virtually disappeared. A complication in all this work is disentangling the sometimes complex issues of severe
mental handicap and “autistic-like” behaviors,
particularly since stereotyped mannerisms
become much more common with severe intellectual deficiency, but characteristic socialcommunicative features may be either difficult to
judge or not clearly present (Volkmar et al. 2005).
Pregnancy and birth complications have been
studied extensively, and other than low birth
weight or being small for gestational age, it is difficult to identify a risk factor (see Gardener et al.
2009 for a recent meta-analysis). It should be
noted, however, that low birth weight may in fact
represent a consequence of genetic vulnerability
rather than an environmental risk factor interacting with genetic susceptibility (Rutter 2005).
Other potential environmental markers signaled
by inflammatory indices are of interest but remain
highly speculative at this point (Muller et al.
2015). Perhaps the most convincing evidence for
an environmental risk factor is that of advanced
parental age (Shelton et al. 2010). Since paternal
and maternal age is so highly correlated, it is
I.E. Drmic et al.
242
difficult to determine if both are risk factors or if
only one. This is important as the mechanism by
which the risk factor might operate will be quite
different based on the maternal/paternal risk factor (Veltman and Brunner 2012). De novo copy
number variants are more common with advanced
paternal age, while chromosomal abnormalities
(like trisomies) are more common with advanced
maternal age.
2.4
Epigenetic Contributions
Epigenetic studies of non-syndromic ASD are in
their infancy but do hold promise once technological and methodical issues associated with this
area of study are resolved. Genes that regulate
DNA methylation have been implicated in the
etiology of ASD, and single gene disorders associated with ASD have implicated epigenetic
mechanisms (Loke et al. 2015). It is anticipated
that this will be an active field in the future and
will yield novel insights in how personenvironment interactions lead to the development
of ASD.
2.5
Physical Biomarkers
Magnetic resonance imaging and diffusion tensor
imaging studies have provided some insight into
brain structure, circuitry, and connectivity in
ASD. Courchesne and colleagues followed toddlers from 12 months to 48 months and found an
abnormal brain growth trajectory in autism
(Schumann et al. 2010). Toddlers, who were ultimately diagnosed with ASD, showed enlargement of cerebral gray and white matter, which
was most pronounced in the frontal, temporal,
and cingulate cortices, and some gray matter
regions showed an abnormal growth rate. In addition, in those infants who later developed ASD,
there was a difference in the development of
white matter tracks between 6 and 12 months of
age (i.e., increases in fractional anisotropy), with
a slowing in development by 24 months (Wolff
et al. 2012). Thus, abnormal neurodevelopmental
connectivity early in life could potentially serve
as an important biomarker for ASD.
2.6
Genotype-Phenotype
Contributions
In addition, genotype-phenotype correlations
have been virtually impossible to establish, and
the fact that the genetic variants identified in
ASD also appear to be playing a role in intellectual disability, epilepsy, schizophrenia, and
ADHD has emphasized the nonspecificity of
these susceptibility risk factors (State 2010).
These genetic findings in ASD have influenced
healthcare recommendation and practice, such
that microarray testing is part of the standard of
care for diagnosis of ASD (McGrew et al. 2012).
It is likely that in the future, whole exome
sequencing will also become standard clinical
practice. However, to date, no studies have
directly assessed the impact of genetic testing on
health outcomes such as earlier age of diagnosis
or reduced use of other diagnostic tools such as
MRI (Sun et al. 2015). In the future, we anticipate that there will be an increased application of
genetics and neuroimaging to the early identification of ASD (Dawson and Bernier 2013). It is
also likely that the discovery of genetic variants
that impact the development of the synapse will
lead to clinical trials of new (or “repurposed”)
drugs targeting those mechanisms. Such trials are
already underway in fragile X and tuberous sclerosis but so far with disappointing results
(Geschwind and State 2015).
3
Outcome Studies
3.1
An Overview
of Developmental Course
Before reviewing approaches to assessing outcome, an overview of what is known, and what is
not known, about the course of autism spectrum
disorders may be helpful. It must be emphasized
that outcome is usually very specifically defined as
adult independence and that little information is
available on health-related outcomes and vanishingly little relates to older individuals with
ASD. Variations in life course are, in part, one of
the rationales used to support previous diagnostic
distinctions between subtypes. In more “classical”
Life Course Health Development in Autism Spectrum Disorders
autism, it is clear that diagnosis in infancy is of
great interest but also somewhat problematic until
about age 3 when reasonable stability in clinical
manifestations occurs. Before that time, the issue
usually is between change from (or to) a more
classic picture to a more “subthreshold” condition
(an issue that may be mitigated by the broad
approach adopted in DSM-5). Often the difficulty,
using the DSM-IV, was that social and communication problems are present but the “restricted
interests” criteria were not fully met (even though
some likely precursor behaviors are present).
Rarely a child moves off the autism spectrum
entirely. There is good evidence (NRC 2001) that
treatments are most effective when started early in
life; although for reasons not well understood,
some children do not improve as dramatically as
others. It is important to emphasize that apart from
a reasonably robust literature on adult outcome
(i.e., early in adult life), the literature on adults
with autism is very limited both in terms of actual
numbers of studies and individuals studied and an
absence of literature on the topic of aging in autism
(Howlin 2013; Perkins and Berkman 2012). For
example, few studies address the issue of rates of
employment among adults with autism/ASD and
rates of suicide or suicide attempts (even though
depression is thought to be a frequent co-occurring
condition), and no studies are available on the life
course changes for individuals with autism/
ASD. Put another way (and viewed in the starkest
possible light), it is as if adults with autism/ASD
vanish from the scene once they’ve become adult
and “had their outcome,” such as capacities in
young adulthood for personal independence and
self-sufficiency.
School-age children with more “classic”
autism often become more sociable as they get
older, and gains in cognitive and communication
skills also typically are made – sometimes dramatic ones and other times not. Social language
use and peer relationships often remain areas of
difficulties, and problems with agitation and selfstimulatory behavior may require behavioral or
pharmacologic intervention. As first observed by
Leo Kanner (Kanner 1971), in adolescence, some
children make major gains, while another group
unfortunately exhibits some degree of deterioration in functioning. A number of outcome studies
243
now suggest a gradual change in overall prognosis presumably reflecting both earlier and better
intervention as well as some changes in diagnostic practice over the time (i.e., including recognizing autism in more cognitively able
individuals) (Howlin 2005). An increasing number of individuals are able to live independently
and self-sufficiently as adults, and many now go
to college or pursue technical/vocational training.
Major predictors of adult outcome include level
of intellectual functioning and communication
competence. Some adults require intermediate
levels of support, and in many cases, affected
individuals never attain independence and must
live in highly structured residential settings. Agerelated vulnerabilities have been noted, e.g., the
risk of seizure disorder is substantially increased
both in early childhood and again in adolescence
and possibly well into adulthood. More able
adults with autism and related conditions also are
apparently more vulnerable to anxiety and
depression which can require specific treatments
(Howlin 2013; Skokauskas and Gallagher 2010;
Szatmari and McConnell 2011; White et al.
2009).
Data on outcome in Asperger’s disorder are,
expectedly, less extensive but suggest (also as
might be expected) overall better outcomes in
terms of independence, educational attainment,
employment, and social independence. However,
comorbid conditions, such as anxiety and depression, can be a complicating factors and negatively
impact outcomes (Skokauskas and Gallagher
2010; Szatmari and McConnell 2011; White et al.
2009; Volkmar et al. 2014). There is some suggestion of increased risk for both psychosis and legal
difficulties in adolescence and adulthood, but the
evidence for this consists largely of case reports
(Skokauskas and Gallagher 2010; Szatmari and
McConnell 2011; White et al. 2009).
Studies of children with “subthreshold” pervasive developmental disorder are very uncommon although, on balance, these individuals
appear to have a better outcome than those with
more classical autism (Towbin 2005). The outcome in childhood disintegrative disorder and
Rett’s does appear, unfortunately, to be poor
(Volkmar et al. 2005; Van Acker et al. 2005). In
childhood disintegrative disorder, children lose
I.E. Drmic et al.
244
skills but then plateau and make relatively minimal gains. In Rett’s, the course is well known
with children becoming somewhat more sociable
over time but also more incapacitated due to neurological and movement problems.
3.2
Adult Independence
Information on long-term prognosis is limited
and difficult to interpret. This review will focus
primarily on long-term follow-up outcome studies that have been published over the past decade
(for reviews of earlier outcome studies see
Gillberg 1991; Henninger and Taylor 2012;
Howlin 2000; Howlin and Moss 2012; Levy and
Perry 2011; Nordin and Gillberg 1998). The
majority of studies that report on “outcomes” for
people with ASD have focused on a few domains
(e.g., work status, residential situation, friendships; see more detailed discussion below).
Predicting outcome has been difficult due to the
wide variability in cognitive, linguistic, social,
and behavioral abilities in this group of individuals. Consistent interpretation of different findings
has also been difficult due to varied sample size
(generally small), sampling procedures (clinical
versus population based), differences in diagnostic criteria over time, wide range of age at diagnosis, differences in amount of time between
initial and follow-up assessments, differences in
measures used, imprecise and/or poor quality
data on early intellectual functioning, and the
lack of information about early intervention services. These factors may account, in part, for the
substantial variability reported in adult outcomes
for people with ASD.
As noted previously, although psychosocial
outcomes have generally been poor, there is a
trend over time for higher rates of improvement
(see Table 1 – description of how the outcome
categories were defined is provided below;
Beadle-Brown et al. 2002; Billstedt et al. 2005,
2007, 2011; Eaves and Ho 2008; Farley et al.
2009; Gillberg 1991; Hofvander et al. 2009;
Howlin 1998; Howlin et al. 2004; Kobayashi
et al. 1992; Lockyer and Rutter 1969, 1970;
Lotter 1978; Lovaas 1987; Nordin and Gillberg
1998; Rutter et al. 1967; Rutter and Lockyer
1967; Szatmari et al. 1989). Studies conducted
from the 1950s to 1970s reported that less than
15% of individuals had good/very good outcomes
and the majority had poor/very poor outcomes
(Eisenberg 1956; Rutter et al. 1967; Lotter 1974).
Over the past three decades, outcomes have
improved but remain variable; the proportion of
good to very good outcomes ranges from 4% to
62% and poor to very poor outcome ranges from
3% to 78% (see Table 1). Outcomes have been
classified into different outcome categories ranging from “very poor to very good” by various
studies over the years. Of note, these ratings were
more subjective in earlier studies (pre-2000),
whereas more objective criteria have been used in
recent studies (for a detailed discussion, see
Henniger and Taylor 2012).
The group of studies published within the last
decade used quantifiable outcome criteria to classify individuals with ASD into these different
outcome categories ranging from very poor to
very good (see Howlin et al. 2004, Appendix 2,
for detailed criteria). In general, the criteria used
to determine outcome were based on a 5-point
composite rating of overall social and independent living functioning that was derived by summing individual scores for work status (ranging
from 0 = employed or self-employed to 3 = no
occupation), residential situation (ranging from
0 = living independently to 5 = being in hospital
care or at home because nowhere else would
accept the individual), and number and quality of
meaningful friendships (ranging from 0 = more
than one friendship to 3 = no friendships or joint
activities) . Based on the 5-point composite rating, a score of 0 indicated a “very good” outcome
(i.e., total for all three areas between 0 and 2);
1 = “good” outcome (total 3–4); 2 = “fair” outcome (total 5–7); 3 = “poor” outcome (total
8–10); and 4 = “very poor” outcome (total 11).
Thus, the concept of “outcome” is related to
practical independence and autonomy achieved
in adult life.
Howlin and her colleagues (2004) surveyed
68 adults (average age of 29 years) with autism
who had a performance IQ of 50 or above in
childhood and found that the majority (58%) had
Mean age at intake
(IN) and follow-up
(FU) (years)
IN: 3.9
FU: 26.6
IN: <10 years
FU: 25.5 (17–40)
Authors
Gillespie-Lynch et al.
(2012)
Billstedt et al. (2011)b
N
AUT = 20
Farley et al. (2009)
AUT = 48
IN: 7.2 (3–25)
FU: 32.5 (22–46)
Eaves and Ho (2008)
AUT = 48
ASD = 40
ASP = 70
AUT = 70
(includes
AUT & ATY)
IN: 6.8 (3–12)
FU: 24.0 (19–31)
ASP
IN: 11.3 (5–24)
FU: 21.5 (16–33)
AUT
IN: <10.0
FU: 24.5 (16–36)
Billstedt et al. (2005)
TOT = 108
(AUT = 73
ATY = 35)
IN: <10.0
FU: 25.5 (17–40)
Howlin et al. 2004
AUT = 68
IN 7.24 (3–15)
FU 29.3 (21–48)
Cederlund et al. (2008)
TOT = 108
(AUT = 73
ATY = 35)
Functioning level at
intake (IN) and
follow-up (FU)
IN: 54.7a
FU: n/a
AUT
IN: 21% IQ > 70
FU: 7% IQ > 70
ATY
IN: 14% IQ > 70
FU: 3% IQ > 70
IN: IQ > 70 (IQ
Estimate = 86.66)
FU: FSIQ = 88.93
IN: 17% IQ ≥ 70
FU: n/a
ASP:
IN: 100% IQ ≥ 70
(FSIQ =101.4)
FU: 98% IQ ≥ 70
(FSIQ = 103.0)
AUT:
IN: 19% IQ ≥ 70
FU: 7% IQ ≥ 70
AUT:
IN: 21% IQ > 70
FU: 7% IQ > 70
ATY
IN: 14% IQ > 70
FU: 3% IQ > 70
IN: PIQ = 80.21
VIQ = 61.49
FU: PIQ = 75.00
VIQ = 79.78
Mean
follow-up Method of initial
time (years) assessment
–
DSM-III
Dx
AUT
Very
poor to
poor
50
Restricted to Good to
very good
fair
20
30
17.8
DSM-III-R
AUT&
ATY
12
26
62
24.4
DSM-III
AUT
17
34
48
–
DSM I–III; Rutter
AUT
46
32
21
–
Gillberg’s criteria,
DSM-III,
DSM-III-R
ASP
AUT
3
76
70
24
27
0
17.8
Rutter, DSM-III
AUT&
ATY
78
21
0
–
Criteria applicable
at time of
diagnosis
(1950–1979)
AUT
58
19
22
Life Course Health Development in Autism Spectrum Disorders
Table 1 Review of adult outcome studies
(continued)
245
246
Table 1 (continued)
Authors
Engstrom et al. (2003)
N
ASP = 32
HFA = 10
Howlin (2000) and
Mawhood et al. (2000)
AUT = 19
Larsen and Mouridsen
(1997)
AUT = 9
ASP = 9
Ruble and Dalrymple
(1996)
Kobayashi et al. (1992)
AUT = 46
AUT = 201
AUT = 23
Lotter 1974
AUT = 32
Rutter et al. (1967) and
Rutter and Lockyer (1967)
IP = 57
Eisenberg (1956)
AUT = 63
IN:
ASP = 9.2;
AUT = 5.9
FU:
ASP 39.1
(33–44); AUT
36.5 (32–39)
IN: 5.2
FU: 17.1
IN: 6.4
FU: 21.5
IN: <6
FU: (16–23)
IN: (8–10)
FU: (16–18)
IN: 5.9 (2–10)
FU: 15.6
IN: 6
FU: 15
Very
poor to
poor
12
Restricted to Good to
fair
very good
75
12
AUT
74
10
16
ICD-10
ASP
AUT
22
67
44
11
33
22
8.6
DSM-III-R
AUT
100
0
0
15.4
DSM-III-R
AUT
46
27
27
6–11
DSM-III; Kanner
& Eisenberg;
Rutter
Creak (1961)
AUT
44
48
4
AUT
63
24
13
Functioning level at
intake (IN) and
follow-up (FU)
IN: n/a
FU: 100% IQ > 70
Mean
follow-up Method of initial
time (years) assessment
–
DSM-IV
IN: NVIQ ≥ 70
NVIQ = 94.28
VIQ = 66.56
FU:
NVIQ = 82.78
VIQ = 82.33
IN:
ASP: 100% IQ > 70
AUT: 56% IQ > 70
FU: N/A
–
Rutter
30
IN: 26.7% IQ >70
FU: 15.2% IQ > 70
IN: 23.4% IQ ≥ 70
FU: n/a
IN: 26% IQ > 70
FU: n/a
IN: 66.7% IQ < 55
FU: n/a
IN: 29% IQ > 70
(mean IQ 62)
FU: n/a
n/a
8
Dx
ASP&
HFA
9.7
Agreement among
psychiatrists
IP
61
25
14
9
Unclear
AUT
73
22
5
ASP Asperger syndrome, ATY atypical autism, AUT autism, HFA high-functioning autism, IP infantile psychosis
a
Developmental quotient (mental age divided by chronological age)
b
QOL scale
I.E. Drmic et al.
Gillberg and Steffenburg
(1987)
Mean age at intake
(IN) and follow-up
(FU) (years)
IN: HFA 13.5
(4–20); ASP 28.0
(15–41)
FU: 30.8 (18–49)
IN: (7–8)
FU: 23.8 (21–26)
Life Course Health Development in Autism Spectrum Disorders
poor or very poor outcomes, 19% had fair outcome, and 22% had good or very good outcome.
Those individuals with a performance IQ of at
least 70 in childhood had significantly better outcome than those with an IQ below 70. More specifically, of those individuals with a performance
IQ between 50 and 69 (n = 23), only one individual had very good outcome, three were rated
as fair, and the remaining had poor or very poor
outcome. Individuals in this IQ category had
much poorer prognosis with few having jobs,
friends, or achieving independence as adults. In
the group of individuals with near average to
average IQ, outcome was generally better,
although still quite variable. Of those individuals
with an IQ of 70 or above (n = 45; parental data
missing on one individual), 14 individuals were
rated as good or very good, 10 as fair, and 20 as
poor or very poor. Furthermore, there was no difference in outcome in those individuals with an
IQ over 100 and those with and initial IQ between
70 and 99. Interestingly, many individuals with
an IQ between 70 and 99 made better progress in
terms of academics, jobs, and general level of
functioning, than those with an IQ above 100.
Thus, having an IQ of about 70 seemed to be a
critical cutoff point for better outcome (e.g., level
of independence); however, outcome in these
individuals was still quite variable.
Other studies found a similar pattern of results.
Billstedt et al. (2005) examined a group of lowto middle-functioning individuals (82% had an
IQ below 70) with autism or atypical autism and
found that 91% had poor or very poor outcome
and none were reported to have good outcome.
Eaves and Ho (2008) reported somewhat better
outcomes in a group of low- to middle-functioning
individuals (majority had IQ < 70) with approximately half having fair to good outcome and half
poor (but none very poor). Similar outcomes
were reported by Gillespie-Lynch et al. (2012),
with 50% reporting poor outcome and 50% fair
to very good outcome. Engstrom, Ekstrom, and
Emilsson (2003) reported on the psychosocial
functioning of a systematically selected sample
of patients with Asperger syndrome (AS) and
high-functioning autism (HFA) all of whom had
an IQ above 70. The majority were rated as hav-
247
ing a fair outcome (75%), with few being rated as
good or poor (12% each) and none being rated as
very poor. Farley et al. (2009) also examined
adult outcomes for 41 individuals with average
or near average cognitive abilities and found better outcomes compared to the other studies with
48% showing good or very good outcome, 34%
with fair outcome, and only 17% with poor outcome (none with very poor outcome). Taken
together, these studies suggest that outcomes for
adults may be improving and that this is likely
related to availability of services for individuals
with ASD (Gillespie-Lynch et al. 2012;
Kobayashi et al. 1992). However, it is important
to also take into consideration the pattern of attrition in longitudinal studies. For instance, selective attrition of lower functioning individuals
may account for the more positive outcomes in
some studies (Eaves and Ho 2008; GillespieLynch et al. 2012).
3.3
Quality of Life
Although the vast majority of studies have based
outcomes on level of independence achieved in
adulthood and IQ, symptom severity and
language development are thought to be important factors in these outcomes; there is increasing
interest in examining outcomes based on quality
of life (QOL) indictors. QOL is a more comprehensive and multidimensional concept that also
encompasses one’s subjective evaluations of
their experiences (Burgess and Gutstein 2007).
According to the World Health Organization
(WHO), the concept of QOL not only includes
level of independence and social relationships
but also is composed of the person’s health, personal beliefs, and their relationship to salient features of their environment (Saxena and Orley
1997).
Generally, studies show that overall QOL and
health-related QOL is relatively low for people
with ASD (Kamp-Becker et al. 2011; van Heijst
and Geurts 2015; Potvin et al. 2015). van Heijst
and Geurts (2015) conducted a meta-analysis on
QOL studies that included children, adolescents,
and adults with ASD and found that QOL is
248
lower in people with ASD compared to people
without ASD and age did not have an effect. The
lack of association with age has previously been
reported (Kamino et al. 2012; Kamp-Becker
et al. 2011), although it had also been reported
that increasing age is associated with decreasing
quality of life (Bennett et al. 2005). Characteristics
that were relevant predictors in the traditional
adult outcome studies, including symptom severity, IQ, and early language, were not examined
because information was too sparse. Given the
dearth of information about the elderly, the
authors also conducted an empirical study examining QOL in intellectually able elderly individuals with ASD and again found large difference in
QOL in people with versus without ASD with
lower QOL in individuals with ASD. Interestingly,
age, IQ, and symptom severity did not predict
QOL in the elderly.
Billstedt et al. (2011) examined aspects of
quality of life (QOL) in adults diagnosed with
autism or atypical autism in childhood and found
that the majority of the ASD group remained
dependent on parents for support in education,
recreation, and occupational situations in late
adolescents and early adulthood, but QOL was
quite positive (independent of intellectual functioning). The QOL scale used in this study measured both objective and subjective (although by
proxy) indicators of well-being. The authors who
evaluated the participants completed a QOL scale
assessing “autism-friendly environment” using a
five-item scale (QOL-1). The item categories
were (1) staff and caregivers have specific
“autism knowledge,” (2) applied structured education implemented, (3) individual specific treatment/training plan for the person with autism
implemented, (4) occupation or everyday life
activity corresponding to his/her level of capacity, and (5) overall quality of life. Each item was
rated on a 1 (very good) to 5 (very poor). A second QOL scale assessing “parent/care rating of
individual’s well-being” was obtained to estimate
how the individual with autism enjoyed or liked
his/her residential conditions (QOL-2). Sixtytwo percent of individuals were rated as having a
very good or good QOL by the authors (QOL-1),
and 91% of families reported high or very high
I.E. Drmic et al.
residential well-being (QOL-2). This is consistent with reports among young adults with intellectual disabilities who reported satisfaction with
their lives even though they remained quite
dependent on caregivers (Keogh et al. 2004) and
a greater sense of empowerment than those individuals who were still in the school system
(Kraemer et al. 2003). Howlin (2013) emphasized that traditional definitions of “good outcome” or “independent living” may not be the
most suitable goal to strive for individuals with
ASD, because it may lead to a life with little support that is isolated and lacking in stimulation.
More research examining QOL is needed in
ASD. Raphael et al. (1996) conceptualize a QOL
model, which is relevant to persons with developmental disabilities, as containing three domains
that include “being, belonging, and becoming”
(p. 28). The “being” domain includes being who
one is or wants to become, “belonging” includes
how one fits within their environment, and
“becoming” involves pursuit of personal goals
and wants. They also noted that within each
domain, it is important to consider the perceived
importance, enjoyment experienced, perceived
control, and opportunities for change and
enhancement. Thus, it is important to also
examine and understand the subjective experience of the individuals with ASD and their families when assessing outcome. As with any chronic
disability, having a condition like ASD may predispose to other problems – particularly difficulties with depression and anxiety disorder; the
relative dearth of work on adult outcome and
complexities of the problems of comorbidity and
diagnosis limit our understanding of the developmental pathways that result in these outcomes.
Similarly, even for the most cognitively able
adults (e.g., those who attend college), challenges
remain in various areas including adaptive (realworld) skills and generalization, social interaction, organization abilities, and so forth (Farley
et al. 2009). As noted previously, the significant
limitations of the available literature must be
emphasized – issues of QOL have not frequently
been assessed, even more cognitively able individuals may lead lives of social isolation with
comorbid mental health conditions, medication
Life Course Health Development in Autism Spectrum Disorders
use is quite high, limitations in daily life can
result from poor adaptive skills, and essentially
no information is available on aging (Howlin
2013). Thus, given the large differences between
quality of life in people with and without ASD, a
great deal of work needs to be done to understand
how to support optimal health development in
individuals with ASD and provide their families
with the kinds of interventions that will enable
them to achieve a higher QOL and promote wellbeing. A shift in research focus is needed to
understand factors that lead to a better QOL
across the life course. There is some evidence
indicating that a higher QOL is related to earlier
diagnosis (Kamino et al. 2012), greater perceived
support (Renty and Roeyers 2006), and supported
employment, residential, and leisure programs
(Garcia-Villamisar et al. 2002, García-Villamisar
and Dattilo 2010; Gerber et al. 2011). Thus, considerable work is needed to improve QOL in
people with ASD, and a better understanding of
the developmental trajectory across the lifespan
may help to better understand the need and interventions required to support these individuals.
3.4
Predictors of Adult
Independence
Intellectual functioning (IQ) and language have
been most consistently identified as the best predictor of adult personal independence as outcome
in ASD (Billstedt et al. 2005, 2007; Gillberg and
Steffenburg 1987; Gillespie-Lynch et al. 2012;
Howlin et al. 2004; Kobayashi et al. 1992; Lotter
1974; Nordin and Gillberg 1998; Rumsey et al.
1985), although reports have been variable
(Farley et al. 2009; Gillespie-Lynch et al. 2012).
Farley et al. (2009) reported that early childhood
variables were weakly associated with adult outcomes, but the participants in their study all had
communicative phrase speech or better language
by age 6 and near average to average IQ. GillespieLynch et al. (2012) also found that early intellectual functioning did not predict adult outcomes
but suggested that this may be because the average age of the first assessment was quite young
(mean age of 3.9 years). Cognitive testing of chil-
249
dren with autism older than 4–5 years of age is
reported to give more stable measures than in
those children who are tested at younger ages
(Lord and Schopler 1989; Howlin et al. 2004). In
general, however, the presence of an intellectual
disability (ID) is associated with poorer outcome
in people with ASD in adult life (Cederlund et al.
2008; Gillberg and Steffenberg 1987; Howlin
et al. 2004; Rumsey et al. 1985; Shattuck et al.
2007), with the poorest outcomes for those individuals with an IQ below 50 (Lord and Bailey
2002). Few people with this level of cognitive
impairment (IQ < 50), regardless of an ASD
diagnosis, achieve a high level of independence
as adults (Keogh et al. 2004; Kraemer and
Blacher 2001). Conversely, higher levels of intellectual functioning (IQ > 70) are associated with
better outcome (Billstedt et al. 2007; Eaves and
Ho 2008; Gillberg and Steffenberg 1987; Howlin
et al. 2004; Rumsey et al. 1985; Szatmari et al.
1989), but outcome can still be quite variable in
these individuals (Billstedt et al. 2005; Howlin
et al. 2004). These data further underscore the
importance of focusing on early language development and learning (IQ) in early intervention
programs for children with ASD.
With regard to verbal and performance (nonverbal) IQ, the results are more inconsistent.
Some have reported that childhood verbal IQ is a
better predictor of later functioning than performance IQ (Billstedt et al. 2005; Lord and Bailey
2002), whereas another study found neither to be
a consistent prognostic indicator (Howlin et al.
2004). In addition, the presence of good early
language skills before age 5–6 is crucial for positive outcome in ASD (Billstedt et al. 2007;
Gillberg and Steffenberg 1987; Gillespie-Lynch
et al. 2012; Lockyer and Rutter 1969; Lotter
1978; Szatmari et al. 2003). Mawhood et al.
(2000) have shown that individuals who tended
to have better linguistic outcome in adulthood
were functioning at a higher cognitive level more
generally.
Other factors have also been suggested to be
related to outcome, including symptom severity,
repetitive/stereotyped behaviors in childhood,
adaptive behavior, education, gender, the presence of a diagnosed medical disorder, onset of
250
epilepsy before age 5 years, and joint attention
skills (Billstedt et al. 2007; De Meyer et al. 1973;
Farley et al. 2009; Gillespie-Lynch et al. 2012;
Howlin and Goode 1998; Totsika et al. 2010;
Venter et al. 1992). For instance, Farley et al.
(2009) reported that adaptive behavior measures
(in particular the Daily Living Skills domain)
evaluated at follow-up were most closely correlated to better outcomes. They described a group
of individuals with a high IQ but poor practical
living skills who felt frustrated by these limitations. Furthermore, they also described individuals who had a low IQ but who were able to
manage with little assistance and obtained good
or very good outcome rating despite poor cognitive skills. These results have important implications for intervention programs that foster these
practical living skills in adulthood.
The development of joint attention skills may
be another important area to target in early intervention programs because of the relationship of
this developmental skill set with later health
development outcomes. Joint attention skills typically develop between 6 and 18 months of age
and refer to ability of the infant to coordinate
their attention with another individual during a
social interaction. Joint attention skills are important because they are considered early markers of
social cognitive development; that is, they reflect
the early development of the infant’s ability to
think about the perspective of another individual
(Tomasello 1995). These skills are critical to
social learning and social information processing
(Mundy and Jarrold 2010), language development (Sigman and Kasari 1995), and the emergence of social and behavioral competence in
childhood (Vaughan Van Hecke et al. 2007).
Children with ASD present with deficits in joint
attention regardless of their developmental or
intellectual level, and these deficits are neither
absolute nor uniform (Mundy et al. 2009).
In a recent longitudinal study into adult outcomes, responsiveness to joint attention, but not
initiation of joint attention (IJA), was found to
predict later outcomes. Responsiveness to joint
attention was found to predict gains in IQ from
early childhood (M = 3.9 years) to later childhood (M = 11.7 years) and receptive language in
I.E. Drmic et al.
late adolescents/early adulthood (M = 18.3 years)
(Sigman and McGovern 2005; Sigman and
Ruskin 1999). In terms of adult outcomes,
responsiveness to joint attention predicted adult
nonverbal communication, social skills, and
symptoms (Gillespie-Lynch et al. 2012). In addition, responsiveness to joint attention and early
language both predicted a composite measure of
adult social functioning and independence. Taken
together, responsiveness to joint attention measured in early childhood may reflect early emerging social cognition, and preliminary evidence
suggests that it may be a good predictor of social
behaviors in adulthood. Gillespie-Lynch and colleagues also suggested examining the relationship with responsiveness to joint attention and
early executive functioning in childhood on later
outcomes given that executive functioning is
related to adaptive functioning in children with
autism (Gilotty et al. 2012).
Gulsrud et al. (2014) examined the developmental trajectories of specific joint attention gestures (i.e., pointing, showing, coordinated joint
looking) and expressive language over a 6-year
period and found that coordinated joint looking
and showing increased over time and pointing to
share interest increased over the preschool years
(3–5 years of age) and then decreased. They also
reported a causal relationship between early
pointing and later language development; that is,
those children with high rates of pointing in the
preschool years had better expressive language
skills later. Hence, an understanding of the developmental course of joint attention skills allows
for better consideration deciding “when” to intervene. Based on the data, the authors suggested
that it may be useful to focus on pointing in early
childhood when the child is preverbal and less
useful when the child begins to use spoken language. Thus, the interval when early joint attention skills develop may represent a critical or
sensitive period to intervene and potentially
impact the developmental trajectory and outcomes for individuals with ASD. In addition, the
children who received the joint attention intervention showed the fastest improvement compared to the control condition or child who
received a different intervention. Participation in
Life Course Health Development in Autism Spectrum Disorders
the joint attention treatment placed these children
on a different developmental trajectory. This
highlights that “what” you target and “when” you
intervene are important (Gulsrud et al. 2014).
3.5
Diagnostic Stability
and Symptom Severity
There is considerable debate in the literature
regarding the stability of a diagnosis of autism
and other forms of ASD over time. Some studies
report that individuals diagnosed in childhood
continue to meet criteria for the disorder at follow-up (Billstedt et al. 2005; Cederlund et al.
2008; Howlin et al. 2004; Lotter 1978); whereas
others report a change in diagnosis or movement
“off” the spectrum in some individuals (Daniels
et al. 2011; Kleinman et al. 2008; Turner and
Stone 2007). Two recent studies reported that a
diagnosis of autism was reasonably stable,
whereas for other forms of ASD such as pervasive developmental disorder, the results were
more variable (Rondeau et al. 2011; Woolfenden
et al. 2012). Rondeau et al. (2011) did a metaanalysis on eight longitudinal studies, published
between 1996 and 2009, examining autism and
pervasive developmental disorder. All diagnoses
were made before 36 months of age, and the
interval between the initial and follow-up assessment was short, ranging from 12 to 84 months.
They reported that an autism diagnosis tended to
be a more stable than pervasive developmental
disorder. Of those with pervasive developmental
disorder, only 35% retained the same diagnosis
(versus the 76% reported for autism), 39% moved
to autism, and 25% moved off the spectrum. The
authors concluded that those children diagnosed
with pervasive developmental disorder at a young
age should be reassessed at an older age.
The diagnosis of autism is relatively reliable
and stable over time. Issues arise for very young
children (the full syndrome picture sufficient for
diagnosis may not be observed until age 3 years)
and for individuals with “autistic-like” or “spectrum” conditions not so clearly satisfying strict
diagnostic criteria. For example, Woolfenden
et al. (2012) conducted a systematic review of 23
251
longitudinal studies (many overlapping with
Rondeau et al. 2011) examining the stability of
the diagnosis of children initially diagnosed with
autism and other forms of ASD. The mean age of
children at baseline ranged from 1.8 to 11.3
years, and mean duration at follow-up ranged
from 2.1 to 32.5 years. Overall, autism was found
to be a stable diagnosis although lower estimates
(as low as 53%) were reported for children diagnosed in the preschool years (below 3 years of
age). A small percentage of individuals diagnosed with autism, between 12% and 15%, move
off the spectrum altogether. For other ASD’s
(including pervasive developmental disorder,
Asperger’s disorder, but not autism), based on the
highest quality studies, the pattern of stability
was more variable with 14–61% retaining their
initial diagnosis at follow-up. A few studies
reported on optimal outcome in autism (Fein
group), and while these adults are indeed quite
independent and functional, it appears that some
autistic traits remain.
Taken together, a diagnosis of autism is more
stable than the other ASD subtypes such as pervasive developmental disorder (Rondeau et al. 2011;
Woolfenden et al. 2012), which is consistent with
the rationale for the DSM-5 (Szatmari 2011).
Given that the ASD category is less stable due to
the inclusion of pervasive developmental disorder
individuals, this presents challenges to clinicians
when sharing a diagnosis of ASD with parents in
terms of making appropriate intervention recommendations (Woolfenden et al. 2012).
Impairments in the core ASD symptoms
(impairment in communication, reciprocal social
interaction, and repetitive or stereotyped behaviors and interests) also tend to persist into adulthood (Billstedt et al. 2007; Howlin 2003; Matson
et al. 2008), although symptom severity is
reported to decrease in some individuals over
time (Seltzer et al. 2003; Shattuck et al. 2007).
Totsika et al. (2010) found that an older (50 years
or older) group of adults with ASD and ID presented with fewer behavioral problems than the
younger (<50 years) group, which is consistent
with reports of decreases in behavior problems in
the general adult intellectual disability population (Holden and Gitlesen 2006). Billstedt et al.
I.E. Drmic et al.
252
(2007) found that social interaction problems
were still present in the vast majority of adults
with ASD, but the occurrence of behavioral
impairments (maladaptive and stereotyped
behaviors) was much more variable with only a
few symptoms in each category affecting half or
more of the study sample. Others have reported
that behavioral problems continue into the adult
years (Ballaban-Gil et al. 1996) and that unusual
responses to sensory (e.g., touch, pain, auditory,
and visual) stimuli also remain common in adulthood (Billstedt et al. 2007; Leekam et al. 2006).
3.6
Cognitive Stability
There is considerable variability in measures of
cognitive ability over time. Some have reported
that IQ is relatively stable in ASD (e.g., BallabanGil et al. 1996; Begovac et al. 2009; Howlin et al.
2004; Lockyer and Rutter 1970; Venter et al.
1992), whereas others have reported gains and/or
losses over time (Billstedt et al. 2005; Farley
et al. 2009; Mawhood et al. 2000). Howlin and
colleagues reported that IQ remained remarkably
stable over the life course; however, stability
tended to be greatest among individuals with an
initial IQ of at least 70. On the other hand,
Billstedt et al. (2005) found that many individuals who initially had near average intelligence
(IQ between 71 and 85) were later diagnosed
with a mild (IQ 50–70) or severe (IQ < 50) intellectual disability, and those individuals with a
severe intellectual disability (IQ < 50) in the original diagnostic study were all still in this category
at follow-up. Overall, they found a downward
shift of IQ level from the diagnostic study to the
follow-up evaluation. A decrease in IQ may be an
important biological marker in a subset of individuals with ASD (Farley et al. 2009). Farley
et al. (2009) reported that over half of the sample
had either large gains or losses in cognitive ability of greater than 1 standard deviation. Cognitive
gain was associated with better outcome, as was
better adaptive functioning. Some of the variability in results may be, in part, accounted for by the
way IQ was measured or assessed, the age of initial assessment (IQ less stable in young children),
and level of functioning of the participant, and
the sometimes highly varied cognitive profiles of
individuals which may evolve over time. This
variability in IQ trajectory may also be due to the
cumulative or time-specific impact of other risk
or protective factors on the plasticity of IQ
development.
4
Transitions Over the Life
Course
4.1
Childhood
Children with ASD, like all other children,
undergo several transitions throughout their
school years. For instance, this may include the
transition from early intervention to preschool,
changing grades each year and moving into adolescence and then into adult life. Individuals with
ASD also struggle with smaller transitions that
are required on a daily basis, such as transitioning from one context to another. For example,
this may include a transition between classes or
transitions to unfamiliar settings (e.g., doctor’s
office). It is important to plan for these transitions
because ineffective transition planning can have a
negative impact on social and academic progress
(Adreon and Stella 2001). Thus, numerous studies have emphasized the importance of transition
planning for all children with special needs
(Polloway et al. 2001), and this is particularly
significant for children with ASD who frequently
struggle with transitions. The ease with which a
child transitions depends on the strategies
adopted by educators when working with the
child with ASD and their family (Harris and
Handelman 2000). Parents of young children
with ASD identified a number of factors that support successful and effective transitions, such as
good communication between the school and
home, transition plans that are child-centered,
and inclusion of parents (Stoner et al. 2007).
Longitudinal studies of ASD have emphasized
documenting the prevalence of certain outcomes
(such as the stability of the diagnosis) rather than
a more detailed look at developmentally related
changes over time. Some studies are now taking
Life Course Health Development in Autism Spectrum Disorders
a life course health development perspective by
focusing on trajectories (Kim et al. 2015). The
Pathways in ASD study, for example, followed
roughly 400 children from the point of diagnosis
at 2–4 years of age to the transition into primary
school at 6 years of age allowing for the possibility of heterogeneous trajectories of autism symptoms and adaptive functioning. The results
suggested (Szatmari et al. 2015) that heterogeneity in the rate of change of both phenotypes
occurred, with some children doing remarkably
well in adaptive function, but autism symptom
severity remained more stable. There was little
overlap of the autistic symptom severity and
adaptive functioning developmental trajectories
so that the course of the two seemed relatively
independent, thereby highlighting the importance of close surveillance and treatment of these
two domains independently over time because
improvement in one area does not ensure
improvement in the other area. These data also
suggested that at least for the transition into primary school, continued improvement is possible
for the majority of children with ASD.
A growing evidence base suggests that behavioral interventions can be associated with positive outcomes for children with ASD, with little
evidence and generally poor quality studies available in adolescents and adults (Lounds Taylor
et al. 2012; Weitlauf et al. 2014; National ASD
Center National Standards Project 2009, 2015).
Early interventions that were based on highintensity applied behavior analysis over extended
periods of time were associated with improvements in cognitive functioning and language
skills, although the magnitude of effects varied
across studies (Weitlauf et al. 2014). This variability may be related to the scarcity of information about modifying factors related to subgroups
(Weitlauf et al. 2014). For instance, the majority
of evidence supports the efficacy of comprehensive early intensive behavioral interventions
(EIBI), which is currently the standard of care in
many clinics and schools (Peters-Scheffer et al.
2011; Reichow 2012). Meaningful gains have
been shown in IQ, language skills, and adaptive
outcomes with medium to large effects (Reichow
2012; Strauss et al. 2013) and a decrease in
253
autism severity over time (Zachor and Itzchak
2010). Gains are not universal, some making
rapid progress and others only modest or no
progress (Peters-Scheffer et al. 2011; Reichow
and Wolery 2009). Inclusion of parents in skill
generalization has led to overall higher effect
sizes (Strauss et al. 2013). Early intensive parent
programs have been shown to modify parent
behavior during interactions, but there is less data
about their ability to improve developmental
skills beyond language gains for some children
(Weitlauf et al. 2014). Predictors of positive outcome and/or changes in trajectory include
younger age at entry into EIBI programs (between
18 months and 5 years), higher IQ or adaptive
functioning or language (mild to moderate range
of functioning), and less severe autism symptoms
(Strauss et al. 2013). With regard to program
characteristics, intensity of EIBI programs was
related to gains in IQ, adaptive functioning, and
language ability, whereas duration of the program was generally not related to gains with the
exception of some evidence that it was related to
language and adaptive behavior gains (Strauss
et al. 2013). Contemporary early intervention
ABA approaches are more naturalistic, childdirected, and guided by developmental theory
and are currently referred to as naturalistic developmental behavioral interventions (NDBI’s) (for
detailed discussion see Schreibman et al. 2015).
Other behavioral interventions that have shown
some positive effects include, but are not limited
to, social skills, treatment targeting joint attention, and cognitive behavior therapy (Weitlauf
et al. 2014). Research is beginning to show that
behaviorally based interventions have an effect
on patterns of brain activity, particularly in
regions involved in social processing (Dawson
et al. 2012), and that neural plasticity in response
to intervention may exist throughout the lifespan
(Faja et al. 2012).
Thus, the evidence supports that early interventions are effective in ASD; however, less is
known about how sustainable these intervention
effects are after transitions from early intervention into preschool and elementary school. There
is emerging evidence that young children with
ASD who received early intervention sustained
I.E. Drmic et al.
254
social, language, and/or nonverbal cognitive
gains through a 6–37-month follow-up period
(e.g., Kasari et al. 2010; Landa et al. 2011; Landa
and Kalb 2012). For instance, Landa and Kalb
(2012) examined outcomes in 2-year-old toddlers
enrolled in a 6-month nursery school-based intervention immediately following intervention (M
age = 35 months), at short-term (M
age = 41 months) and long-term follow-up (M
age = 72 months; 37 months after children completed intervention). At long-term follow-up,
they found gains in IQ and communication; however, increases were not evenly distributed across
the different time points. Gains in IQ and communication were robust initially from pre- to
post-intervention, followed by a period of stabilization, which was then followed by another
developmental burst. In contrast, the trajectory of
autism severity was different with an initial
decrease in severity form pre- to post-intervention,
followed by an increase in severity to preintervention levels by the final time point. Magiati
et al. (2011) reassessed outcomes even later at 5
years after departing preschool and found that
children maintained standardized IQ scores, but
adaptive functioning scores decreased. Longerterm outcome studies are needed to determine the
sustainability of gains, as well as to determine
whether there are sensitive periods in development as well as the developmental scaffolding
that is necessary, especially during key life course
transitions that are likely to promote optimal
health development outcomes (e.g., age, type of
intervention, duration, intensity, content, etc.)
(Landa and Kalb 2012).
4.2
Transition from Adolescence
into Adulthood
For youth with ASD and their families, the transition into adulthood is a time of uncertainty and
loss of entitlement to many services that were
available while in the school system. In comparison to early childhood, there often is a dearth of
appropriate supports and opportunities for adults
with ASD. Leaving high school can have a disruptive effect on youth and young adults with
ASD in a number of areas, including ASD symptom presentation, maladaptive behaviors, and
family functioning (Taylor and Seltzer 2010,
2011a, b). Taylor and Seltzer (2010) examined
the impact of exiting high school and observed
that in general, autism symptoms and maladaptive behaviors continued to improve while adolescents were part of the secondary school
system, but improvement significantly slowed
(by over half) after exiting high school for internalizing behavior and all but one of the autism
symptom domains (i.e., verbal communication
impairments). The authors suggested that this
change was likely related to the lack of disabilityrelated services or that occupational or day program services were less stimulating than those in
the school system. Interestingly, youth without
comorbid intellectual disability improved more
in both autism symptoms and maladaptive
behaviors while in high school compared to
those with intellectual disability (Shattuck et al.
2007), but after exiting high school, improvement slowed more for those individuals without
an ID (Taylor and Seltzer 2010). Indeed, only
6% of individuals with ASD without comorbid
ID were receiving day services immediately
after high school, compared to 74% with ASD
and ID, and over one-quarter had no occupational, educational, or day activities. In addition,
the improvement in the mother-youth relationship seen in high school slowed or stopped after
high school (i.e., decreasing maternal warmth),
with the greatest impact on relationships for
youth with ASD and no ID (Taylor and Seltzer
2011a). Thus, leaving high school had a negative
impact on individuals with ASD, with the most
pronounced effect on those youth with ASD
without an ID, suggesting a particular lack of
adequate educational and occupational activities
for this group of individuals in adulthood. It also
highlights that individuals with ASD and no ID
are often impaired in their adaptive skills and
encounter difficulty generalizing these skills in
daily life. When services are inadequate, parents
will often take on this responsibility, leading
some families to experience this transition period
as a time of stress and anxiety (Howlin 2005).
Other families view this period as an opportunity
Life Course Health Development in Autism Spectrum Disorders
for new and more positive possibilities, such as
finding more appropriate occupational placements and leaving high school, which for some
youth, was a negative experience (Taylor 2009).
Access to autism-related services is also
related to family income. Families with lower
incomes and less parental education may have
less access to autism-related services in childhood compared to families with higher incomes
(Liptak et al. 2008; Thomas et al. 2007).
Improvement in maladaptive behaviors slows
after high school more so for those families with
a lower income (Taylor and Seltzer 2011b). Thus,
it is important to better understand the factors
that promote optimal transition for individuals
with ASD, as well as the impact on families.
Taylor (2009) underscores the importance of
examining contextual and environmental factors
that are amenable to change or can be targeted in
intervention in order to improve success with
transition and suggests that exclusively focusing
on characteristics such as IQ and early language
that although are predictive of later outcome is
more difficult to intervene once the individual
with ASD becomes an adolescent or adult. For
instance, the implementation of transition planning has been associated with more successful
achievement of adult milestones (Frank and
Siltington 2000). Indeed, factors such as the posthigh school goals for the student and parental
expectations were predictive of participation in
postsecondary education (Chiang et al. 2012). In
addition, the development of appropriate educational, occupational, and day programs for individuals with ASD of all ability levels is important
as is a lifelong emphasis on generalization of
skills.
4.2.1 Educational Attainment
With regard to education, many individuals in the
follow-up studies published in the last decade
attended specialized schools or classrooms
(Billstedt et al. 2011; Farley et al. 2009, Howlin
et al. 2004). Only a few individuals with ASD
obtained formal qualifications (Eaves and Ho
2008; Howlin et al. 2004), although results of
recent studies have been more promising
(Hofvander et al. 2009; Farley 2009). For example,
255
Farley and colleagues reported that 44% completed
high school at grade 12 with a diploma, and 39%
went on to postsecondary education. With regard to
those students who are capable of entry into college
or university, many do not seek or gain entry or
drop out prematurely (Glennon 2001; VanBergeijk
et al. 2008) due to a number of factors including
social isolation and loneliness, difficulty managing
changing routines and schedules, difficulty with
independent living, and the absence of monitoring
and guidance (Howlin et al. 2004; Jobe and White
2007; White et al. 2011).
Students with ASD are able to succeed both
academically and socially in postsecondary education when the transition is carefully planned,
appropriate accommodations are obtained, and
supports are available (for detailed discussion,
see VanBergeijk et al. 2008). Preparing students
for the transition to postsecondary education is
important for many adolescents but is particularly important for individuals with ASD, most of
whom have difficulty with transitions.
VanBergeijk and colleagues stress the importance
of a good fit between the student and the prospective academic institution. For instance, this may
include careful consideration of the size of the
school and classes, level of supports, and receptiveness to serving students with disabilities –
specifically ASD, access to mental health
services, and availability of supports to address
both academic concerns (e.g., organizational
help, accommodations related to evaluation and
test taking, support with complex assignments,
etc.), as well as nonacademic concerns including
life skills, issues of daily living (e.g., laundry,
budgeting), organizational difficulties, communication limitations, and social skills (e.g., socializing with peers, getting along with roommates,
romantic relationships). Indeed, adolescents with
ASD reported greater concern about social acceptance and being able to function independently
than about accessing academic accommodations
and resources in postsecondary educational settings, and thereby identified a need for built in
social supports and access to trained staff members as a resource (Camarena and Sarigiani
2009). Many individuals with ASD also have difficulty with issues related to sexuality and may
256
require support or direct instruction (see Volkmar
2004). Furthermore, postsecondary educational
programs must also include planning and preparation for the transition to work and independent
living that includes vocational training and life
coaching (VanBergeijk et al. 2008). Families and
educators are also encouraged to support the
development of self-advocacy skills which are
important to the transition process so that adolescent and young adults are able to navigate the
journey through higher education, work, and
independent living (Camarena and Sarigiani
2009). Thus, for students with aspirations for
postsecondary education, careful planning and
supports are required to help students with ASD
achieve academic success.
4.2.2
Employment, Independent
Living, and Social Life
The research on transition into adulthood in individuals with ASD has primarily focused on the
traditional view of transition which describes the
degree of independence in living arrangements,
ability to gain and maintain employment, and the
presence of intimate relationships outside of the
family unit (Fussell and Furstenberg 2005). The
majority of individuals with ASD have difficulty
achieving the developmental tasks of adulthood
and remain dependent on parents/caregivers for
support in employment and accommodation, and
most do not have intimate relationships (friendship or romantic).
Many persons with ASD are able to work successfully within the community (GarciaVillamisar et al. 2000; Mawhood and Howlin
1999); however, the majority experience difficulty securing meaningful employment (Billstedt
et al. 2011; Eaves and Ho 2008; Howlin et al.
2004; Newman et al. 2011). Findings from the
National Longitudinal Transition Study-2
(NLTS2) examined a nationally representative
sample of youth and young adults with disabilities in the USA and found that up to 8 years posthigh school, only 63% of adults with autism had
worked at some point after high school and only
37% were employed at the time of the survey
(Newman et al. 2011). In a group of individuals
I.E. Drmic et al.
with near average to average intelligence, the
outcomes were somewhat more promising with
54% in full-time or part-time independent paid
jobs, 12% in supported employment or part-time
volunteer positions, 24% in day programs, and
10% unemployed (Farley et al. 2009). Even
among those who are employed, many worked
few hours, and most jobs tended to be low level,
were found by parental contacts (versus open job
market), and were poorly paid such that individuals did not have adequate financial support to live
independently (Cimera and Cowan 2009; Eaves
and Ho 2008; Howlin et al. 2004). Furthermore,
many individuals were in part-time, volunteer,
supported, or sheltered employment; few worked
independently with little or no support, and some
had no daytime occupation at all. Jobs often
ended prematurely because of social or behavioral difficulties or other work-related difficulties
(Mawhood and Howlin 1999). Cimera and
Cowan (2009) examined cost of services and
employment outcomes achieved by adults with
ASD in the vocational rehabilitation system. The
study found that adults with autism were
employed at higher rates than other disability
groups; however, the rate of employment was
only 41% (60% do not obtain employment), and
as noted earlier, they worked fewer hours and
earned lower wages. Adults with autism were
also the most costly group of individual to serve,
and services were difficult to implement. Howlin
(2013) underscores that generic employment
services do not meet the needs of individuals
with ASD, and there is a need for more programs and research specific to individuals with
ASD. Although limited, research is beginning to
emerge showing that supported employment
models for individuals with ASD are effective
(Howlin et al. 2005; Keel et al. 1997; Shattuck
et al. 2012; Taylor et al. 2012).
Critical factors in finding and maintaining a
long-term placement include a thorough assessment to appropriately match the skills and abilities of the person with ASD to the requirements
of the job, as well as having the appropriate level
and type of employment support (Mawhood and
Howlin 1999; Smith and Paulippen 1999).
Life Course Health Development in Autism Spectrum Disorders
Hagner and Cooney (2005) found that a set of
supervisory accommodation strategies were
commonly associated with successful supervision, including maintaining a consistent schedule
and set of job responsibilities, using organizers to
structure the job, minimizing unstructured or idle
time, being direct with communication, and providing reminders and reassurances. Longer job
retention has been correlated with higher levels
of social inclusion and acceptance on the job
(Belcher and Smith 1994). Furthermore, engaging in work is often related to other meaningful
outcomes, such as contributing to one’s selfworth and confidence, providing opportunities to
shares one’s strengths that are valued by others,
opportunities to develop friendships or other supportive relationships, as well as engagement in
the larger community (Lee and Carter 2012). On
the other hand, failure to find suitable work
resulted in frustration and in some serious problems with depression and/or anxiety (Mawhood
and Howlin 1999). With supported employment,
individuals with ASD are able to find and maintain suitable employment (Mawhood and Howlin
1999).
Most individuals with ASD live with their parents or in other supported arrangements (e.g.,
community-based group home), and few live
independently either with or without some level
of support (Billstedt et al. 2011; Farley et al.
2009). Eaves and Ho (2008) reported that 56%
lived with their parents, 35% were in supported
arrangements such as group homes or foster care,
and the remainder lived more or less independently. With regard to friendship, it has been
reported that up to approximately 30% of individuals have one or more friends (Eaves and Ho
2008; Howlin et al. 2004). Social contacts often
result from special interests and skills rather than
close friendships (Howlin 2000). Some individuals have been reported to want friendships but
were unable to form these relationships, whereas
others had no interest or did not understand the
concept of friendship (Billstedt et al. 2011). Few
individuals had close intimate relationships or
were married at some point (Eaves and Ho 2008;
Farley et al. 2009; Hofvander et al. 2009; Howlin
257
et al. 2004). In a group of higher functioning individuals with ASD, Farley and colleagues reported
that 7% were married, 5% divorced, 7% were in
long-term relationships, 44% never dated, 32%
dated in both group and couple situations, and
22% dated only in group settings. When parents
were asked whether they felt their adult child
wished to have a romantic relationship, of those
participants not already in relationships, 44% felt
their son or daughter would like to be in a relationship. On the other hand, 41% did not want a
romantic partner because parents thought their
children were concerned about passing on autism
to a child or themselves being too difficult for a
partner to manage.
4.2.3 Summary
Taken together, the research to date focuses primarily on the attainment of traditional skills that
are indicative of successful transition into adulthood and less on more subjective or quality of life
indicators. Although outcomes are quite variable,
in general, outcomes related to level of independence, employment status, and intimate relationships are poor, suggesting that the transition to
adulthood is fraught with many challenges.
Evidence suggests that better outcomes are related
to better cognitive abilities and earlier language
skills, although even these individuals struggle
with attaining the developmental tasks of adulthood. Furthermore, availability of appropriate
resources, services, and supports for the individuals with ASD and their families are key ingredients
for successful transition into adulthood and better
outcomes throughout adult life. Lee and Carter
(2012) identified seven elements that are important for high-quality transition services, which
include individualized services that reflect the
strengths of the individual, positive career development and early work experiences, collaboration
and interagency involvement, family supports and
expectations, fostering self-determination and
independence, social- and employment-related
skill instruction, and establishment of job-related
supports. Thus, the impetus of future research is to
examine factors that will support successful transition (e.g., environmental and contextual factors)
I.E. Drmic et al.
258
and find other ways to define transition such as
from a psychosocial or attachment perspective (for
review, see Taylor 2009), especially as the epidemiology of ASD changes from autism with ID to a
preponderance of ASD without ID.
4.3
Aging and ASD
Autism is a lifelong condition; however, little is
known about older adults with ASD. Indeed,
much of the research to date has examined the
aging ASD population but not the aged (65 plus)
(Perkins and Berkman 2012). Although information is quite limited, research suggests that life
expectancy is shortened compared to the general
population and causes of death include high rates
of seizures and accidental deaths (e.g., drowning)
(Mouridsen et al. 2008; Piven and Rabins 2011).
It is unclear whether deaths that are related to diseases (e.g., nervous and sensory disease) in ASD
are due to a predisposition to certain health concerns or due to poor health surveillance leading
to poor diagnosis and management (Perkins and
Berkman 2012). A number of medical and psychiatric concerns have been reported in children
and adolescents with ASD, such as sleep disorders, psychiatric symptoms, gastrointestinal disorders, feeding selectivity and aversions,
metabolic conditions, and epilepsy; however,
there is little information about these concerns as
individuals age (Piven and Rabins 2011).
Similarly, it is unclear whether treatments that
are effective in children and adolescents would
also benefit adults. Given the dearth of information of older adults with ASD, a group of experts
were convened to characterize the gaps in knowledge and define a research agenda to bridge this
gap (see Piven and Rabins 2011). Research priorities included developing diagnostic criteria
and tools to diagnose older adults, conducting
cross-sectional descriptive studies in adults of
phenomenology and associated features, longitudinal studies of lifespan trajectories, neurobiological studies examining underlying changes
over time, intervention studies, and mechanism
to support clinical and research training in aging.
This research is needed to improve the lives of
individuals with ASD and to inform clinical practice and policies that can address these healthcare
disparities (Piven and Rabins 2011).
5
Mental Health Issues in ASD
5.1
Prevalence Rates
In terms of quality of adult life, there is increasing
evidence suggesting that comorbid psychiatric
symptoms and disorders are common in individuals with ASD, with estimated rates ranging from
30% to 70% (e.g., Bradley et al. 2004; Gjevik
et al. 2011; Leyfer et al. 2006; Moseley et al.
2011; Simonoff et al. 2008; Skokauskas and
Gallagher 2010). Understanding comorbid mental
health problems is important not only because of
the high prevalence but also because of the impact
on daily functioning, long-term prognosis, and
the added burden and challenges faced by individuals and their families. Individuals with ASD
have higher rates of comorbidity compared to the
general population (Ghaziuddin et al. 1998;
Moseley et al. 2011), a psychiatrically referred
population of youth without ASD (Joshi et al.
2010), as well as individuals with ID alone
(Bradley et al. 2004; Brereton et al. 2006; LoVullo
and Matson 2009), although the latter has not
been consistently found (Tsakanikos et al. 2006).
The most commonly reported psychiatric disorders include anxiety and depression (MacNeil
et al. 2009; Sterling et al. 2008; White et al. 2009).
ASD has also been found to co-occur with obsessive compulsive disorder (OCD), disruptive
behavioral disorders, attention deficit hyperactivity disorder (ADHD), bipolar disorder, Tourette’s
disorder, tic disorder, and eating disorders (Drmic
amd Szatmari 2014; Skokauskas and Gallagher
2010; Szatmari and McConnell 2011; Volkmar
et al. 2014; White et al. 2009). Studies examining
schizophrenia have been mixed; some studies
report high rates (Chang et al. 2003), whereas others have not found an increased rate (Seltzer et al.
2004; Volkmar and Cohen 1991). Although the
two disorders are quite different, misclassification
of ASD as schizophrenia may also contribute to
the variability in reported rates (Perlman 2000;
Life Course Health Development in Autism Spectrum Disorders
Palucka, Bradley, Lunsky 2008). Furthermore,
ASD often co-occurs with multiple psychiatric
disorders (Leyfer et al. 2006; Simonoff et al.
2008). For instance, Simonoff et al. (2008), using
a population-based cohort, conducted a structured
psychiatric interview (not modified for ASD) with
112 10–14-year olds and found that 70% of ASD
participants had at least one comorbid disorder
and 41% had two or more. It is important to better
understand this comorbidity given that many
individuals with ASD are likely to struggle with
mental health concerns, and more importantly,
psychiatric problems may cause considerable distress and interference with daily functioning and
significantly impact long-term prognosis and outcome (Kim et al. 2000; Mattila et al. 2010; Muris
et al. 1998; Russell and Sofronoff 2005; White
et al. 2009).
To date, the majority of research on psychiatric comorbidity has been conducted in children
and adolescents (Amr et al. 2012; Gjevik et al.
2011; Leyfer et al. 2006; Mattila et al. 2010;
Skokauskas and Gallagher 2010). In a review
conducted by Skokauskas and Gallagher (2010)
examining comorbid disorders in children with
autism and Asperger syndrome, wide ranges in
rates of co-occurring symptoms or diagnoses
were found: 0–6% for schizophrenia, 5–35% for
generalized anxiety, 10–64% for simple phobias,
1–37% for OCD, and 0–50% for affective disorders or symptoms. Fewer studies have examined
this issue in adults, although research in this area
is beginning to emerge (Cederlund et al. 2010;
Ghaziuddin and Zafar 2008; Hofvander et al.
2009; La Malfa et al. 2007; Lugnegard et al.
2011; Moseley et al. 2011), and a number of follow-up studies have reported on psychiatric outcomes (Billstedt et al. 2005; Eaves and Ho 2008;
Farley et al. 2009). A study that examined young
adults with Asperger syndrome found high rates
of depression and anxiety, consistent with the literature in children and adolescents with ASD,
whereas psychotic and substance-induced disorders were uncommon (Lugnegard et al. 2011).
Hofvander et al. (2009) assessed 122 consecutively referred adults with ASD and normal intelligence ranging in age from 16 to 60 years.
Lifetime axis I comorbidity was common, and
259
they also found that mood (53%) and anxiety disorders (50%) were most common, followed by
ADHD (43%), OCD (34%), and chronic tic disorders (20%). With regard to the rate of lifetime
axis II disorders, 62% met criteria for at least one
personality disorder. Other studies have reported
somewhat lower rates of anxiety (Farley et al.
2009; Moseley et al. 2011) and depression
(Billstedt et al. 2005; Eaves and Ho 2008; Farley
et al. 2009; Moseley et al. 2011). A study examining adolescents and young adults with autism
(over three-quarters had an intellectual disability)
found that 42% had an additional mental health
disorder, a rate that is two to four times seen in
typically developing young people (Moseley
et al. 2011). There is some evidence that the rates
of mood and anxiety disorders in parents of children with autism is higher than that in the general
population and when compared to Down syndrome (Bolton et al. 1998; Mazefsky et al. 2008).
This may reflect an effect of stress on family
members. A more thorough examination of
genetic variable expressivity would involve a
study of mood disorders in second- and thirddegree relatives.
5.2
Mental Health Services
Given the high rate of psychiatric comorbidity,
there is a need for appropriate mental health services for individuals with ASD of all ages. This
includes preventive interventions designed to
optimize mental functioning and life course trajectories. In a study investigating the healthcare
experiences of children with ASD and their families, it was found that 15% of families reported
unmet mental health services, and provider lack
of skills was a barrier to obtaining these services
(Chiri and Warfield 2012). With regard to the utilization of mental health services by individuals
with ASD, the reports are mixed, with some
reporting underutilization of services (Bryson
et al. 2008) and others reporting relatively high
rates of service use (Narendorf et al. 2011).
Narendorf et al. (2011) reported that of those
youth who received mental health services (i.e.,
46%), 49% received it through the school.
I.E. Drmic et al.
260
African-American youth and youth from lowerincome families were more likely to use schoolbased services than accessing community-based
services, underscoring the importance of schools
in providing these services for underserved group
and the importance of ensuring the continuation
of services after high school. For students with
ASD, there is emerging research examining the
delivery of mental health services in schools for
anxiety (Drmic, Aljunied, Reaven 2017; Luxford,
Hadwin, Kovshoff 2016). Bryson et al. (2008)
found that short-term crisis-oriented mental health
services were effective at stabilizing children with
ASD but that many remained highly symptomatic
at discharge, suggesting that changes are needed
to the current system in order to serve those individuals with chronic and long-term needs.
Many individuals with ASD may be at risk for
psychiatric hospitalization. Indeed, approximately 11% of children and youth aged
5–21 years experienced at least one psychiatric
hospital inpatient admission (Mandell 2008), and
48% used psychiatric hospital services by age 40
(Mouridsen et al. 2008). Children with a diagnosis of Asperger’s disorder and pervasive developmental disorder, compared to children with
autism, were more likely to experience inpatient
hospitalization (Bryson et al. 2008). Lunsky et al.
(2009) compared patients with ASD and ID to
other individuals accessing tertiary level mental
healthcare in Ontario and found that individuals
with ASD and ID were younger, spent more days
in hospitals, and were less likely to have a psychotic disorder diagnosis than both patients with
and without ID. Interestingly, fewer than half of
the individuals with ASD and ID had an additional psychiatric diagnosis, although almost all
(91%) were prescribed psychotropic medication,
which is consistent with previous studies
(Tsakanikos et al. 2006, 2007a). Tsakanikos et al.
(2007b) reported that behavioral management
problems predicted psychotropic medication and
use of psychiatric services in adults with pervasive developmental disorder and ID. A study
examining referral trends to specialist mental
health services in south London from 1983 to
2000 for individuals with ASD and ID reported
an increase in diagnosable psychiatric disorders
over time and a significant reduction of medication at time of referral, but no change in the use of
other therapeutic interventions, which is in contrast to services received by younger children
with ASD. Thus, there continues to be a need for
specialized inpatient and outpatient mental health
services for individuals with ASD, as well as
more training for service providers.
6
Summary
ASD is a neurodevelopmental disorder that represents a chronic, lifelong condition based on
atypicalities in brain development. While there
are changes in syndrome expression over time,
new challenges often emerge. Much of the current research has focused on static or crosssectional outcomes as opposed to developmental
trajectories and changes in those trajectories.
This can include looking at the developmental
course of the disorder, the developmental course
of specific skills that are important to later outcomes such as joint attention skills, as well as
trajectories, related to the effects of early intervention. Longer-term studies are critical to treatment research in order to determine the
sustainability of early intervention treatment
gains, to explore whether there are sensitive periods of child development when treatments are
most effective, and to determine factors that promote treatment outcome. Research needs to
include enough information about factors that
might be important to outcome so that predictors
can be better elucidated, as well as a shift to focus
on mediators and moderators of outcome. There
is a need for more longitudinal studies and some
consensus about the type of information that
should be gathered by all research groups so that
findings are comparable among different studies
and can be combined into meta-analyses. For
instance, standardized IQ measures could be used
to document intellectual abilities. It may be beneficial to convene a group of experts as well as
parents and individuals with ASD to determine
what basic set of factors are important.
The current review highlights that outcomes
for people with ASD have been improving over
Life Course Health Development in Autism Spectrum Disorders
time; although relative to typically developing
children and adolescents, outcomes are still generally poor. Much of the work to date has focused
on achieving the traditional developmental tasks
of adulthood, including independence in living
arrangements, gaining and maintaining employment, and developing intimate relationships outside of the family unit. Research examining
quality of life is beginning to emerge, which is an
important next step to further our understanding
of how to help improve the lived experiences of
individuals with ASD. QOL studies need to shift
focus and include information about factors and
developmental processes that are thought to be
important to achieving better life course health
development outcomes, such as IQ, symptom
severity, and language development. In addition,
examining other factors that promote thriving in
individuals with ASD is needed. Understanding
what those factors are is critical so that better
programs and interventions can be developed,
which include the developmental scaffolding
that children and families need to optimize longterm health development outcomes. It is also
important to understand what impact access to
adequate resources and intervention has on the
identification, service provision, and likely outcomes on people with ASD. Another key theme
is the development and emergence of mental
health comorbidities which often represent an
enormous challenge for young adults with ASD
and their families. Research needs to include
information about the developmental pathways
that lead to these outcomes so that risk and protective factors can be targeted early, with the
intent of optimizing the trajectory of mental
health outcomes. There is also a need for more
basic research such as epigenetic studies of nonsyndromic ASD, genetic studies that focus on
much smaller copy number variants or DNA
sequence variants identified either in exomes or
in whole genome sequencing, and continued
research examining environmental risk factors.
A number of recommendations to facilitate
the transition periods and support individual with
ASD and their families can be gleaned from the
existing literature. Implementation of a transition
plan is crucial since it is associated with more
261
successful transition from early intervention into
preschool (Stoner et al. 2007) and attainment of
adult milestones (Frank and Siltington 2000) and
may help temper the disruptive effect of leaving
high school (Taylor and Seltzer 2010, 2011a, b).
Parents, clinicians, and educators are encouraged
to begin the transition planning process for the
transition to adulthood early and include planning for treatment, independence (i.e., daily living skills, skills for independent living),
education, employment, living arrangements,
mental health, and medical care. For instance,
good practical living skills, even in those individuals who have a cognitive disability, led to
better outcomes and quality of life (Farley et al.
2009). Therefore, focusing on developing independence in daily living skills starting from a
young age is an important target for intervention
programs and transition planning. This highlights
the importance of planning early so that skills can
be integrated into treatment and educational programs at younger ages, allowing for ample practice and supervision of these skills well before
the individual is expected to be able to manage on
their own.
For many individuals and families, the transition to adulthood is a time of worry and uncertainty because many services that were available
through the educational system are no longer
available and there are few appropriate supports
or services for adults with ASD. Taylor and
Seltzer (2010) showed that the improvement
slowed significantly after exiting high school, particularly for those youth with ASD and no ID. This
change was likely related to a lack of services or
that available services were less stimulating than
those provided in the educational system. Thus,
the development of new models of care that incorporate appropriate educational, occupational and
day programs for adults with ASD of all ability
levels is essential. However, simply having a program is not enough; it is also important to ensure
that the appropriate training and support is provided on an ongoing basis to the individual with
ASD and that service providers are adequately
trained to work with this group of individuals. For
instance, given the prevalence and impact of mental health issues in individuals with ASD and their
I.E. Drmic et al.
262
families, there is a need for ongoing mental health
services and staff who are trained in this area and
able to support families. A worrying trend is the
increased use of psychopharmacology in adults
with ASD often as a result of lack of psychosocial
services that might better meet those needs in a
way that improves life course outcomes and promotes thriving. Another example relates to successfully obtaining and maintaining long-term
employment, which may involve supportive
employment programs that include training
related to general job skills (e.g., resume writing,
interviewing skills, proper social behavior), specific skills needed for a particular job (e.g., learning job requirements and skills, learning about the
culture and expectations for that job), and ongoing support (e.g., job coach). Training and support
should also be provided to the employers in order
to educate them about ASD in general, help them
understand the needs of a particular individual, as
well as highlight their unique skills and abilities
that can positively contribute to workplace.
Indeed, parents, educators, employers, and other
professionals are encouraged to appreciate the
many strengths, skills, and unique talents of individuals with ASD and approach current programs
and future planning from a strength-based
approach, leveraging each individual’s unique set
of assets, versus a deficits-based approach that
focuses on what needs to be fixed.
There is a stark lack of information about
ASD in the aging and elderly population. Given
this gap in knowledge, a group of experts were
convened to identify and describe these gaps and
define a research agenda (see Piven and Rabins
2011). The research proprieties included developing diagnostic criteria and tools to diagnose
older adults, conducting cross-sectional descriptive studies in adults of phenomenology and
associated features, longitudinal studies of lifespan trajectories, neurobiological studies examining underlying changes over time, and
intervention studies and mechanism to support
clinical and research training in aging.
This change in perspective might be part of a
shift from treatment of deficits to accommodation of unique characteristics of the adult with
ASD. There comes a time when the relentless
pursuit of treatment to “make things better” will
have a negative impact on the person with ASD
and their family. People with ASD should not
always be considered “disabled,” and taking a
lifespan perspective to developmental trajectories
should be part of this switch for service providers
as well as parents.
7
Research Priorities
There has been a marked increase in ASD-related
research over the past decades. Considerable
advances have been made in key areas, including: the genetic basis of ASD and its diagnosis;
studies of the social brain; the connections
between timing of diagnosis and long-term outcomes; new treatment models, some pharmacological and behavioral treatments can now be
viewed as evidence based; psychiatric co-morbidities with particular emphasis on anxiety and
depression; and, research on basic issues like
employment, living arrangements, medical
survelillance, etc. Thus, a number of gaps remain
that need to be addressed. The following summarizes a number of recommendations for
research, some of which were noted earlier in the
paper, to better understand life course health
development in ASD.
7.1
Research Priorities
(i) Basic mechanism research:
(a) Better understanding of epigenetic
mechanisms in non-syndromic ASD.
(b) Continued research in the area of genetics and neuroimaging. For example, this
could include:
• Genetic studies that focus on much
smaller copy number variants or DNA
sequence variants identified either in
exomes or in whole genome sequencing
• Studies that assess the impact of genetic
testing (e.g., microarrays) on health
outcomes
• Studies combining genetics and neuroimaging work
Life Course Health Development in Autism Spectrum Disorders
• Neuroimaging studies examining the trajectories of brain development
• Neuroimaging studies examining the
impact of behavioral treatments on the
brain
(c) Continued research examining environmental risk factors, as well as geneenvironment interactions.
(d) Identification of biomarkers (e.g.,
genetics, neural circuitry) is a high priority. For instance, the National
Institutes of Health Interagency
Coordinating Committee Strategic Plan
called for the identification of biological
markers, which separately or in combination with behavioral markers accurately identified ASD before age 2
(Dawson and Bernier 2013).
(e) Development of novel therapeutics,
e.g., creating medicine that can target
core ASD symptoms.
(f) Focus on early developmental skills or
early markers of development that may
impact the developmental trajectory.
For instance, there is evidence that early
joint attention skills are important for
social cognitive development and later
language and adaptive skills. Further,
interventions targeting joint attention
have been shown to be effective. Thus,
research further examining the role of
joint attention skills in ASD would be
beneficial. It is also important to identify other potential early developing
skills that may provide early targets for
intervention.
(ii) Clinical Research:
(a) Early diagnosis remains a treatment priority. The recent advances in technology
(e.g., neuroimaging) and genetics suggest the next wave of advances in our
ability to detect autism through the use
of biomarkers and new screening tools.
Thus, research examining the utility of
these biomarkers in clinical contexts
will be needed. The availability of predictive biomarkers could index risk for
ASD before or at the onset of symptoms,
263
thereby preventing the symptoms from
developing or alleviating symptoms and
improving the trajectory of outcomes.
(b) Development of more evidence-based
treatment research throughout the lifespan is needed.
• A growing body of literature has shown
that interventions can be associated with
positive outcomes in children; however,
there is still a need for studies across setting and improvements in methodological
rigor.
• Little evidence is available about treatment in adolescents and adults. There is a
need to develop treatment across the lifespan and determine “which” treatments are
most needed and effective at the different
life stages and “when” these treatments
are most effective.
• Substantial scientific advances are needed
to improve our understanding of which
interventions are most effective for specific children with ASD and to elucidate
the factors or components of interventions
most associated with positive outcomes.
• Exciting potential to bring together basic
science and treatment in development of
new metrics/measures (e.g., predicting
response to treatments – recent work on
EEG/fMRI and treatment change).
(c) Very little evidence is available for specific treatment approaches in adolescents and adults; this is especially the
case for evidence-based approaches to
support the transition of youth with
autism to adulthood. (Lounds et al.
2012). Thus, more research is needed to
focus transition period.
(d) Little information exists about the aging
population in ASD with virtually no
information about the aged (65+). More
research is needed in various areas from
basic and clinical research to data/
method development and translational
research. A research agenda spanning
all these areas for the aging population
was described in Piven and Rabins
(2011). As noted earlier, the research
I.E. Drmic et al.
264
priorities included developing diagnostic criteria and tools to diagnose older
adults, conducting cross-sectional
descriptive studies in adults of phenomenology and associated features, longitudinal studies of lifespan trajectories,
neurobiological studies examining
underlying changes over time, and
intervention studies and mechanism to
support clinical and research training in
aging.
(e) Studies examining quality of life and
the person, environmental, and personenvironmental interactions that lead to a
thriving life.
(iii) Translational research:
(a) Need innovative solutions for training
physicians, clinicians, and other professionals working with individuals with
ASD throughout the lifespan. In particular, there is a serious lack of welltrained healthcare professional to serve
the large number of adults with
ASD. This includes multiple areas,
including diagnosis and assessment of
adult, mental health and medical supports, and supports for daily life.
(b) Research related to transition planning
as it relates to treatment, independence,
education, employment, living arrangements, mental health, and medical care.
7.2
Data/Method Development
Priorities
(a) A shift toward longitudinal studies to better
understand developmental trajectories (with
attention to child and family characteristics –
e.g., SES, parental education)
(b) Inclusion of characteristics that might be
important to outcome (e.g., IQ, language
ability, etc.) so that predictors of outcome can
be better elucidated, as well as a shift to focus
on mediators and moderators of outcome.
(c) A shift in focus examining not only risk factors but also on factors that are protective and
promote thriving.
(d) Better metrics throughout the lifespan.
There is an exciting potential to bring
together basic science and treatment in
development of new metrics/measures (e.g.,
predicting response to treatments as was
seen in recent work on fMRI and treatment
change).
(e) Development of better measures to look at
outcome determination and more focus on
quality of life (QOL) measures for the individual and family.
(f) Greater focus in critical or sensitive periods
of development when environemntal exposures can have long-lasting effects on
outcomes.
7.3
Translational Priorities
(a) Early diagnosis. A wide gap remains
between our knowledge about best practices
in ASD (e.g., screening for ASD at 18 and
24 months) and Centers for Disease Control
and Prevention (CDC) data showing that the
average age of diagnosis in the USA is
5 years and higher for individuals with
milder forms of ASD. This is a problem
given that much of the literature on early
intervention shows best outcomes when
intervention is initiated early (between
18 months and 5 years). This is a major challenge and will require innovative solutions
and a focus on dissemination and implementation science.
(b) Better coordination of different systems
related to children’s health development
(e.g., school and health systems).
(c) Development of physician guidelines for
healthcare.
(d) Challenge in providing access to behavioral,
mental health, and medical healthcare and,
thus, the need for improved and expanded
healthcare services with care providers who
are well trained in working with individuals
with ASD and their families.
(e) Innovative solutions to better serve those in
remote regions or underserved population
(e.g., individuals living in poverty).
Life Course Health Development in Autism Spectrum Disorders
(f) Incorporating independent living skills into
treatment/educational programs starting at a
young age.
(g) Although our understanding of autism has
increased, there is a major gap in translating
this to public school settings.
(h) Development of new models/systems of care
that incorporate appropriate educational,
occupational and day programs for adults
with ASD of all ability levels is essential.
(i) Vocational system for adults.
(j) Generic employment services do not meet
the needs of individuals with ASD, therefore
the need for more programs and research specific to individuals with ASD (e.g., supported
employment models). Supportive employment programs – training related to general
job skills, specific skills needed for a particular job, and ongoing support (e.g., job coach).
(k) Training and support for employers.
8
Conclusions
Taken together, there has been a tremendous
growth in research in the ASD field. However,
there remain a number of gaps that need to be
addressed, with a particular focus on optimizing
life course trajectories for individuals and populations with ASD. The principles of the life
course health development framework describe
health as an emergent set of developmental
capacities that develop continuously over the
lifespan in a complex, nonlinear process occurring in multiple dimensions, levels, and phases
and are sensitive to timing. This is a valuable
framework to direct the research agenda to continue to move the field forward and to improve
the lives of individuals with ASD, their families,
and their communities.
References
Adreon, D., & Stella, J. (2001). Transition to middle
and high school: Increasing the success of students
with Asperger syndrome. Intervention in School and
Clinic, 36, 266–271.
American Psychiatric Association. (1980). Diagnostic
and statistical manual. Washington, DC: APA Press.
265
Amr, M., Raddad, D., El-Mehesh, F., Bakr, A., Sallam,
K., & Amin, T. (2012). Comorbid psychiatric
disorders in Arab children with autism spectrum disorders. Research in Autism Spectrum Disorders, 6,
240–248.
Asperger, H. (1944). Die “autistichen Psychopathen”
im Kindersalter. Archive fur psychiatrie und
Nervenkrankheiten, 117, 76–136.
Baker, T. B., Breslau, N., Covey, L., & Shiffman, S.
(2012). DSM criteria for tobacco use disorder and
tobacco withdrawal: A critique and proposed revisions for DSM-5. Addiction, 107(2), 263–275.
doi:10.1111/j.1360-0443.2011.03657.x.
Ballaban-Gil, K., Rapin, I., Tuchman, R., & Shinner, S.
(1996). Longitudinal examination of the behavioral,
language and social changes in a population of adolescents and young adults with autistic disorder. Pediatric
Neurology, 15, 217–223.
Baron-Cohen, S. (2011). The science of evil: On empathy
and the origins of cruelty. New York: Basic Books.
Beadle-Brown, J., Murphy, G., Wing, L., Gould, J., Shah,
A., & Holmes, N. (2002). Changes in social impairment for people with intellectual disabilities: A follow-up of the Camberwell cohort. Journal of Autism
and Developmental Disorders, 32, 195–206.
Begovac, I., Begovac, B., Majic, G., & Vidovic, V. (2009).
Longitudinal studies of IQ stability with childhood
autism-literature survey. Psychiatria Danubina, 21(3),
310–319.
Belcher, R., & Smith, M. D. (1994). Coworker attitudes
towards employees with autism. Journal of Vocational
Rehabilitation, 4, 29–36.
Bennet, H. E., Wood, C. L., & Hare, D. J. (2005).
Providing care for adults with autistic spectrum disorders in learning disability services: Needs-based
or diagnosis-driven? Journal of Applied Research in
Intellectual Disabilities, 18, 57–64.
Billstedt, E., Gillberg, C., & Gillberg, C. (2005). Autism
after adolescents: Population-based 13- to 22-year follow-up study of 120 individuals with autism diagnosed
in childhood. Journal of Autism and Developmental
Disorders, 35(3), 351–360.
Billstedt, E., Gillberg, C., & Gillberg, C. (2007). Autism
in adults: Symptom patterns and early childhood predictors. Use of the DISCO in a community sample followed from childhood. Journal of Child Psychology
and Psychiatry, 48(11), 1102–1110.
Billstedt, E., Gillberg, I. C., & Gillberg, C. (2011). Aspects
of quality of life in adults diagnosed with autism in
childhood: A population-based study. Autism, 15(1),
7–20.
Bolton, P. F., Pickles, A., Murphy, M., & Rutter, M.
(1998). Autism, affective, and other psychiatric disorders: Patterns of familial aggregation. Psychological
Medicine, 28, 385–395.
Bradley, E. A., Summers, J. A., Wood, H. L., & Bryson,
S. E. (2004). Comparing rates of psychiatric and
behavior disorders in adolescents and young adults
with severe intellectual disability with and without autism. Journal of Autism and Developmental
Disorders, 34, 151–161.
266
Brand, B. L., Lanius, R., Vermetten, E., Loewenstein,
R. J., & Spiegel, D. (2012). Where are we going? An
update on assessment, treatment, and neurobiological
research in dissociative disorders as we move toward
the DSM-5. Journal of Trauma & Dissociation, 13(1),
9–31.
Brereton, A. V., Tonge, B. J., & Einfeld, S. L. (2006).
Psychopathology in children and adolescents with
autism compared to young people with intellectual
disability. Journal of Autism and Developmental
Disorders, 36, 862–870.
Brown, J. R., & Rogers, S. J. (2003). Cultural issues in
autism. In R. L. Hendren, S. Ozonoff, & S. Rogers
(Eds.), Autism spectrum disorders (pp. 209–226).
Washington, DC: American Psychiatric Press.
Brugha, T. S., McManus, S., Bankart, J., Scott, F.,
Purdon, S., Smith, J., et al. (2011). Epidemiology of
autism spectrum disorders in adults in the community
in England. Archives of General Psychiatry, 68(5),
459–466.
Bryson, S. A., Corrigan, S. K., McDonald, T. P., &
Holmes, C. (2008). Characteristics of children with
autism spectrum disorders who received services
through community mental health centers. Autism, 12,
65–82.
Burgess, A. F., & Gutstein, S. E. (2007). Quality of life for
people with autism: Raising the standard for evaluating successful outcomes. Child and Adolescent Mental
Health, 12, 80–86.
Camarena, P. M., & Sarigiani, P. A. (2009). Postsecondary
educational aspirations of high-functioning adolescents with autism spectrum disorders and their parents. Focus on Autism and Other Developmental
Disabilities, 24(2), 115–128.
Cederlund, M., Hagberg, B., Billstedt, E., Gillberg, I. C.,
& Gillberg, C. (2008). Asperger syndrome and autism:
A comparative longitudinal follow-up study more than
5 years after original diagnosis. Journal of Autism and
Developmental Disorders, 38, 72–85.
Cederlund, M., Hagberg, B., & Gillberg, C. (2010).
Asperger syndrome in adolescent and young adult
males. Interview, self – And parent assessment of
social, emotional, and cognitive problems. Research
in Developmental Disabilities, 31, 287–298.
Centers for Disease Control and Prevention (CDC).
(2014). Prevalence of Autism Spectrum disorder among children aged 8 years — Autism and
Developmental disabilities monitoring network, 11
sites, United States, 2010. Morbidity and Mortality
Weekly Report, 63(SS02), 1–21.
Chang, H. L., Juang, Y. Y., Wang, W. T., Huang, C. I.,
Chen, C. Y., & Hwang, Y. S. (2003). Screening for
autism spectrum disorder in adult psychiatric outpatients in a clinic in Taiwan. General Hospital
Psychiatry, 25, 284–288.
Chawarska, K., Klin, A., & Volkmar, F. R. (2008). Autism
spectrum disorders in infants and toddlers: diagnosis,
assessment, and treatment. Autism spectrum disorders
in infants and toddlers: Diagnosis, assessment, and
I.E. Drmic et al.
treatment (pp. 327–336, 348). New York: Guilford
Press.
Chiang, H.-M., Cheung, Y. K., Hickson, L., Xiang,
R., & Tsai, L. Y. (2012). Predictive factors of participation in postsecondary education for high
school leavers with autism. Journal of Autism and
Developmental Disorders, 42, 685–696. doi:10.1007/
s10803-011-1297-7.
Chiri, G., & Warfield, M. E. (2012). Unmet needs and
problems accessing core health care services for
children with autism spectrum disorders. Journal of
Child and Maternal Health, 16, 1081–1091.
Cimera, R. E., & Cowan, R. J. (2009). The costs of services and employment outcomes achieved by adults
with autism in the US. Autism, 13(3), 285–302.
Constantino, J. N., Zhang, Y., Frazier, T., Abbacchi, A. M.,
& Law, P. (2010). Sibling recurrence and the genetic
epidemiology of autism. The American Journal of
Psychiatry, 167(11), 1349–1356.
Creak, M. (1961). Schizophrenic syndrome in childhood.
Developmental Medicine and Child Neurology, 3,
501–504.
Daniels, A. M., Rosenberg, R. E., Law, J. K., Lord, C.,
Kaufmann, W. E., & Law, P. A. (2011). Stability of
initial disorder diagnoses in community settings.
Journal of Autism and Developmental Disorders,
41(1), 110–121.
Dawson, G., & Bernier, R. (2013). A quarter century of progress on early detection and treatment
of autism spectrum disorder. Development and
Psychopathology, 25, 1455–1472.
Dawson, G., Jones, E. J., Merkle, K., Venema, K., Lowy,
R., Faja, S., et al. (2012). Early behavioral intervention is associated with normalized brain activity in
young children with autism. Journal of the American
Academy of Child & Adolescent Psychiatry, 51,
1150–1159.
DeMyer, M. K., Barton, S., DeMyer, W. E., Norton, J. S.,
Allen, J., & Steele, R. (1973). Prognosis in autism:
A follow-up study. Journal of Autism and Childhood
Schizophrenia, 3, 199–246.
Devlin, B., & Scherer, S. W. (2012). Genetic architecture in autism spectrum disorder. Current Opinion in
Genetics and Development, 22, 229–237.
Devlin, B., Melhem, N., & Roeder, K. (2011). Do common variants play a role in risk for autism? Evidence
and theoretical musings. Brain Research, 1380, 78–84.
Drmic, I., Aljunied, M., & Reaven, J. (2017). Facing
your fears in schools: Implementing CBT for teens
with ASD and anxiety in Singapore. Journal of
Autism and Developmental Disabilities. doi:10.1007/
s10803-016-3007-y.
Drmic, I., & Szatmari, P. (2014). Emotional dysregulation and comorbidity in Autism Spectrum Disorder.
Invited review for Cutting Edge Psychiatry in Practice
– Autism Issue, 1, 119–131.
Eaves, L. C., & Ho, H. H. (2008). Young adult outcomes
of autism spectrum disorders. Journal of Autism and
Developmental Disorders, 38, 739–747.
Life Course Health Development in Autism Spectrum Disorders
Eisenberg, L. (1956). The autistic child in adolescence.
The American Journal of Psychiatry, 112, 607–612.
Elsabbagh, M., Divan, G., Koh, Y.-J., Kim, Y. S., Kauchali,
S., Marcin, C., et al. (2012). Global prevalence of
autism and other pervasive developmental disorders.
Autism Research, 5(3), 160–179. doi: http://dx.doi.
org/10.1002/aur.239.
Engstrom, I., Ekstrom, L., & Emilsson, B. (2003).
Psychosocial functioning in a group of Swedish adults
with Asperger syndrome or high-functioning autism.
Autism, 7(1), 99–110.
Faja, S., Webb, S. J., Jones, E., Merkle, K., Kamara, D.,
Bavaro, J., et al. (2012). The effects of face expertise training on the behavioral performance and brain
activity of adults with high functioning autism spectrum disorders. Journal of Autism and Developmental
Disorders, 42, 278–293.
Farley, M. A., McMahon, W. M., Fombonne, E., Jenson,
W. R., Miller, J., Gardner, M., et al. (2009). Twentyyear outcome for individuals with autism and average
or near-average cognitive abilities. Autism Research,
2, 109–118.
Fombonne, E. (2005a). Epidemiology of autistic disorder
and other pervasive developmental disorders. Journal
of Clinical Psychiatry, 66(Suppl 10), 3–8.
Fombonne, E. (2005b). The changing epidemiology of
autism. Journal of Applied Research in Intellectual
Disabilities, 18(4), 281–294. doi: http://dx.doi.
org/10.1111/j.1468-3148.2005.00266.x.
Frank, A. R., & Siltington, P. L. (2000). Young adults with
mental disbailities – Does transition planning make a difference? Education and Training in Mental Retardation
and Developmental Disabilities, 35, 119–134.
Fussell, E., & Furstenberg, F. F. (2005). The transition
to adulthood during the twentieth century. In R. A.
Settersten, F. F. Furstenberg, & R. G. Rumbaut (Eds.),
On the frontier of adulthood: Theory, research and
public policy (pp. 29–75). Chicago: University of
Chicago Press.
García-Villamisar, D. A., & Dattilo, J. (2010). Effects
of a leisure pro-gramme on quality of life and stress
of individuals with ASD. Journal of Intellectual
Disability Research, 54, 611–619.
Garcia-Villamisar, D., Ross, D., & Wehman, P. (2000).
Clinical differential analysis of persons with autism
in a work setting: A follow-up study. Journal of
Vocational Rehabilitation, 14, 183–185.
García-Villamisar, D., Wehman, P., & Navarro, M. D.
(2002). Changes in the quality of autistic people’s
life that work in supported and sheltered employment: A 5-year follow-up study. Journal of Vocational
Rehabilitation, 17, 309–312.
Gardener, H., Spiegelman, D., & Buka, S. L. (2009).
Prenatal risk factors for autism: Comprehensive metaanalysis. British Journal of Psychiatry, 195(1), 7–14.
Gerber, F., Bessero, S., Robbiani, B., et al. (2011).
Comparing residential programmes for adults with
autism spectrum disorders and intellectual disability:
Outcomes of challenging behaviour and quality of
life. Journal of Intellectual Disability Research, 55,
918–932.
267
Geschwind, D.H., & State, M.W. (2015). Gene hunting
in autism spectrum disorder: on the path to precision
medicine. Lancet Neurology, Published online http://
dx.doi.org/10.1016/S1474-4422(15)00044-7.
Ghaziuddin, M., & Zafar, S. (2008). Psychiatric comorbidity in adults with autism spectrum disorders.
Clincial Neuropsychiatry, 5, 9–12.
Ghaziuddin, M., Weidmer-Mikhail, E., & Ghaziuddin,
N. (1998). Comorbidity of Asperger syndrome: A
preliminary report. Journal of Intellectual Disability
Research, 42, 278–283.
Gillberg, C. (1991). Outcome in autism and autistic-like
conditions. Journal of the American Academy of Child
and Adolescent Psychiatry, 30, 375–382.
Gillberg, C., & Steffenburg, S. (1987). Outcomes and
prognostic factors in infantile autism and similar
conditions: A population-based study of 46 cases
followed through puberty. Journal of Autism and
Developmental Disorders, 17, 272–288.
Gillespie-Lynch, K., Sepeta, L., Wang, Y., Marshall, S.,
Gomez, L., Sigman, M., & Hutman, T. (2012). Early
childhood predictors of the social competence of adults
with autism. Journal of Autism and Developmental
disorder, 42, 161–174.
Gilotty, L., Kenworthy, L., Sirian, L., Black, D. O., &
Wagner, A. E. (2012). Adaptive skills and executive function in autism spectrum disorders. Child
Neuropsychology, 8(4), 241–248.
Gjevik, E., Eldevik, S., Fjaeran-Granum, T., & Sponheim,
E. (2011). Kiddie-SADS reveals high rates of DSM-IV
disorders in children and adolescents with autism spectrum disorders. Journal of Autism and Developmental
Disorders, 41, 761–769.
Glennon, T. J. (2001). The stress of the university experience for students with Asperger syndrome. Work:
Journal of Prevention, Assessment & Rehabilitation,
17(3), 183–190.
Gulsrud, A. C., Hellemann, G. S., Freeman, F. N., &
Kasari, C. (2014). Two to ten years: Developmental
trajectories of joint attention with ASD who received
targeted social communication interventions. Autism
Research, 7, 207–215.
Hagner, D., & Cooney, B. F. (2005). “I do that for everybody”: Supervising employees with autism. Focus
on Autism and Developmental Disabilities, 20(2),
91–97.
Halfon, N., & Forrest, C. B. (2017). The emerging theoretical framework of life course health development. In
N. Halfon, C. B. Forrest, R. M. Lerner, & E. Faustman
(Eds.), Handbook of life course health-development
science. Cham: Springer.
Halfon, N., Larson, K., Lu, M., Tullis, E., & Russ, S.
(2014). Lifecourse health development: Past, present and future. Journal of Maternal Child Health, 18,
344–365.
Hallmayer, J., Cleveland, S., Torres, A., Phillips, J.,
Cohen, B., Torigoe, T., et al. (2011). Genetic heritability and shared environmental factors among twin
pairs with autism. Archives of General Psychiatry,
68(11), 1095–1102. doi:10.1001/archgenpsychiatry.
2011.76.
268
Harris, L. J., & Handelman, J. S. (Eds.). (2000). Preschool
education programs for children with autism (2nd
ed.). Austin: PRO-ED.
Henninger, N. A., & Taylor, J. L. (2012). Outcomes
in adults with autism spectrum disorders:
A historical perspective. Autism, 0(0), 1–14.
doi:10.1177/1362361312441266.
Hermelin, B. (2001). Bright Splinters of the mind: A personal story of research with Autistic savants. London:
Jessica Kingsley.
Hofvander, B., Delorme, R., Chaste, P., Nyden, A., Wentz,
E., Stahlberg, O., et al. (2009). Psychiatric and psychosocial problems in adults with normal-intelligence
autism spectrum disorders. BMC Psychiatry, 9, 35.
doi:10.1186/1471-244X-9-35.
Holden, B., & Gitlesen, J. P. (2006). A total population study
of challenging behavior in the country of Hedmark,
Norway: Prevalence, and risk markers. Research in
Developmental Disabilities, 27(4), 456–465.
Howlin, P. (1998). Children with autism and Asperger
syndrome. A guide for practitioners and carers.
Chichester: John Wiley & Sons.
Howlin, P. (2000). Outcome in adult life for more able
individuals with Autism or Asperger Syndrome.
Autism, 4(1), 63–83.
Howlin, P. (2003). Outcome in high-functioning adults
with autism with and without early language delays:
Implications for the differentiation between autism
and Asperger syndrome. Journal of Autism and
Developmental Disorders, 33, 3–13.
Howlin, P. (2005). Outcomes in autism spectrum disorders. In P. Howlin (Ed.), Handbook od autism and pervasive developmental disorder, volume 1: Diagnosis,
development, neurobiology, and behavior (3rd ed.).
Hoboken: John Wiley & Sons.
Howlin, P. (2013). Outcomes in adults with autism spectrum disorders. In F. Volkmar, S. Rogers, R. Paul, &
K. Pelphrey (Eds.), Handbook of autism (4th ed.).
Hoboken: Wiley.
Howlin, P., & Goode, S. (1998). Outcomes in adult life for
individuals with autism. In F. Volkmar (Ed.), Autism
and developmental disorders. New York: Cambridge
University Press.
Howlin, P., & Moss, P. (2012). Adults with autism spectrum disorders. Canadian Journal of Psychiatry,
57(5), 275–283.
Howlin, P., Alcock, J., & Burkin, C. (2005). An 8 year
follow-up of a specialist supported employment service for high-ability adults with autism or Asperger
syndrome. Autism, 9(5), 533–549.
Howlin, P., Goode, J., Hutton, J., & Rutter, M. (2004).
Adult outcomes for children with autism. Journal of
Child Psychology and Psychiatry, 45, 212–229.
Huerta, M., Bishop, S. L., Duncan, A., Hus, V., & Lord,
C. (2012). Application of the DSM-5 criteria for
autism spectrum disorder to three samples of children
with DSM-IV diagnoses of pervasive developmental
disorders. American Journal of Psychiatry, 169(10),
1056–1064.
I.E. Drmic et al.
Jobe, L., & White, S. W. (2007). Loneliness, social relationships, and a broader autism phenotype in college
students. Personality and Individual Differences, 42,
1479–1489.
Joshi, G., Petty, C., Wozniak, J., Henin, A., Fried,
R., Galdo, M., et al. (2010). The heavy burden of
psychiatric comorbidity in youth with autism spectrum disorders: A large comparative study of psychiatrically referred population. Journal of Autism and
Developmental Disorders, 40, 1361–1370.
Kamio, Y., Inada, N., & Koyama, T. (2012). A nationwide
survey on quality of life and associated factors of
adults with high-functioning autism spectrum disorders. Autism, 17, 15–26.
Kamp-Becker, I., Schröder, J., Muehlan Remschmidt,
H., et al. (2011). Health-related quality of life in children and adolescents with autism spectrum disorder.
Zeitschrift für Kinder - und Jugendpsychiatrie und
Psychotherapie, 39(2), 123–131.
Kanner, L. (1943). Autistic disturbances of affective contact. Nervous Child, 2, 217–250.
Kanner, L. (1971). Follow-up study of eleven autistic
children originally reported in. Journal of Autism &
Childhood Schizophrenia, 1(2), 119–145.
Kasari, C., Gulsrud, A. C., Wong, C., Kwon, S., & Locke,
J. (2010). Randomized controlled caregiver mediated joint engagement intervention for toddlers with
autism. Journal of Autism Developmental Disorders,
40(9), 1045–1056.
Keel, J. H., Mesibov, G. B., & Woods, A. V. (1997).
TEACCH-supported employment program. Journal
of Autism and Developmental Disabilities, 27(1), 3–9.
Keogh, B. K., Bernheimer, L. P., & Guthrie, D. (2004).
Children with developmental delays twenty years
later: Where are they? How are they? American
Journal on Mental Retardation, 109(3), 219–230.
Kim, S.H., Macari, S., Koller, J., & Chawarska, K.
(2015). Examining the phenotypic heterogeneity of
early autism spectrum disorder: Subtypes and shortterm outcomes. Journal of Child Psychology and
Psychiatry. doi:10.1111/jcpp.12448.
King, M., & Bearman, P. (2009). Diagnostic change and
the increased prevalence of autism. International
Journal of Epidemiology, 38(5), 1224–1234. Epub
2009 Sep 7. PubMed PMID: 19737791.
Kleinman, J., Ventola, P., Pandey, J., Verbalis, A., Barton,
M., Hodgson, S., et al. (2008). Diagnostic stability in
very young children with autism spectrum disorder.
Journal of Autism and Developmental Disorders,
38(4), 606–615. doi:10.1007/s10803-007-0427-8.
Klin, A., Volkmar, F. R., Sparrow, S. S., Cicchetti, D. V., &
et al. (1996). Validity and neuropsychological characterization of Asperger syndrome: Convergence with nonverbal learning disabilities syndrome. Annual Progress
in Child Psychiatry & Child Development, 241–259.
Kobayashi, R., Murata, T., & Yoshinaga, K. (1992). A follow-up study of 201 children with autism in Kyushu
and Yamaguchi areas, Japan. Journal of Autism and
Developmental Disorders, 22, 395–411.
Life Course Health Development in Autism Spectrum Disorders
Kraemer, B. R., & Blacher, J. (2001). Transition for young
adults with severe mental retardation: School preparation, parent expectations, and family involvement.
Mental Retardation, 39, 432–455.
Kraemer, B. R., McIntyre, L. L., & Blacher, J. (2003).
Quality of life for young adults with mental retardation
during transition. Mental Retardation, 41, 250–262.
Kim, J., Szatmari, P., Bryson, S. E., & Wilson, F. J. (2000).
Prevalence of anxiety and mood problems among children with autism and Asperger syndrome. Autism, 4,
117–132.
LaFramboise, T., Winckler, W., & Thomas, R. K. (2009).
A flexible rank-based framework for detecting copy
number aberrations from array data. Bioinformatics,
25(6), 722–728.
La Malfa, G., Lassi, S., Salvini, R., Giganti, C., Bertelli,
M., & Albertini, G. (2007). The relationship between
autism and psychiatric disorders in intellectually disabled adults. Research in Autism Spectrum Disorders,
1, 218–228.
Landa, R. J., & Kalb, L. G. (2012). Long-term outcomes
of toddlers with autism spectrum disorders exposed
o short-term intervention. Pediatrics, 130(2), S186–
S190. doi:10.1542/peds.2012-0900Q.
Landa, R. J., Holman, K. C., & Garrett-Mayer, E. (2007).
Social and communication development in toddlers
with early and later diagnosis of autism spectrum
disorders. Archives of General Psychiatry, 64(7),
853–864.
Landa, R. J., Holman, K. C., O’Neill, A. H., & Stuart,
E. A. (2011). Intervention targeting development of
socially synchronous engagement in toddlers with
autism spectrum disorder: A randomized controlled
trial. Journal of Child Psychology and Psychiatry,
52(1), 13–21.
Larsen, F. W., & Mouridsen, S. E. (1997). The outcome
in children with childhood autism and Asperger syndrome originally diagnosed as psychotic. A 30-year
follow-up study of subjects hospitalized as children.
European Child and Adolescent Psychiatry, 6(4),
181–190.
Lee, G. K., & Carter, E. W. (2012). Preparing transitionage students with high-functioning autism spectrum
disorders for meaningful work. Psychology in the
Schools, 49(10), 988–1000. doi:10.1002/pits.21651.
Leekam, S. R., Nieto, C., Libby, S. J., Wing, L., & Gould,
J. (2006). Describing the sensory abnormalities of
children and adults with autism. Journal of Autism and
Developmental Disorders, 37, 894–910. doi:10.1007/
s10803-006-0218-7.
Levy, A., & Perry, A. (2011). Outcome in adolescents and
adults with autism: A review of the literature. Research
in Autism Spectrum Disorders, 5, 1271–1282.
Levy, S. E., Mandell, D. S., & Schultz, R. T. (2009).
Autism. Lancet, 374, 1627–1638.
Leyfer, O. T., Folstein, S. E., Bacalman, S., Davis, N. O.,
Dinh, E., Morgan, J., et al. (2006). Comorbid psychiatric disorders in children with autism: Interview development and rates of disorders. Journal of Autism and
Developmental Disorders, 36, 849–861.
269
Li, X., Zou, H., & Brown, W. T. (2012). Genes associated with autism spectrum disorder. Brain Research
Bulletin, 88(6), 543–552.
Liptak, G. S., Benzoni, L. B., Mruzek, D. W., Nolan, K. W.,
Thingvoll, M. A., Wade, C. M., & Fryer, G. E. (2008).
Disparities in diagnosis and access to health services
for children with autism: Data from the national survey of children’s health. Journal of Developmental &
Behavioral Pediatrics, 29(3), 152–160.
Lockyer, L., & Rutter, M. (1969). A five- to fifteenyear follow-up study of infantile psychosis: III.
Psychological aspects. The British Journal of
Psychiatry, 115, 865–882.
Lockyer, L., & Rutter, M. (1970). A five- to fifteen-year
follow-up study of infantile psychosis: IV. Patterns
of cognitive ability. British Journal of Social and
Clinical Psychology, 9(2), 152–163.
Loke, Y. J., Hannan, A. J., & Craig, J. M. (2015). The
role of epigenetic change in autism spectrum disorders. Frontiers in Neurology, 6, 107. doi:10.3389/
fneur.2015.00107.
Lord, C., & Bailey, A. (2002). Autism spectrum disorders.
In M. Rutter & E. Taylor (Eds.), Child and adolescent psychiatry (4th ed., pp. 664–681). Oxford, MA:
Blackwell Scientific.
Lord, C., & Schopler, E. (1989). The role of age at assessment, developmental level, and test in the stability of
intelligence scores in young autistic children. Journal
of Autism and Developmental Disorders, 19(4),
483–499.
Lotter, V. (1974). Social adjustment and placement of
autistic children in Middlesex: A follow-up study.
Journal of Autism and Childhood Schizophrenia, 4,
11–32.
Lotter, V. (1978). Follow-up studies. In M. Rutter &
E. Schopler (Eds.), Autism: A reappraisal of concepts
and treatment (pp. 475–495). New York: Plenum Press.
Lounds Taylor, J., Dove, D., Veenstra-VanderWeele,
J., Sathe, N.A., McPheeters, M.L., Jerome, R.N.,
Warren, Z.. (2012). Interventions for adolescents
and young Adults with Autism Spectrum disorders.
Comparative effectiveness review no. 65. (Prepared
by the Vanderbilt evidence-based practice Center
under contract no. 290–2007-10065-I.) AHRQ publication no. 12-EHC063-EF. Rockville, MD: Agency
for Healthcare Research and Quality. www.effectivehealthcare.ahrq.gov/reports/final.cfm
Lovaas, I. (1987). Behavioral treatment and normal
educational and intellectual functioning in young
autistic children. Journal of Consulting and Clinical
Psychology, 55(1), 3–9.
LoVullo, S. V., & Matson, J. L. (2009). Comorbid
psychopathology in adults with autism spectrum
disorders and intellectual disabilities. Research
in Developmental Disabilities, 30, 1288–1296.
doi:10.1016/j.ridd.2009.05.004.
Lugnegard, T., Hallerback, M. U., & Gillberg, C. (2011).
Psychiatric comorbidity in young adults with a clinical diagnosis of Asperger syndrome. Research in
Developmental Disabilities, 32, 1910–1917.
270
Lunsky, Y., Gracey, C., & Bradley, E. (2009). Adults with
autism spectrum disorders using psychiatric hospitals
in Ontario: Clinical profile and service needs. Research
in Autism Spectrum Disorders, 3, 1006–1013.
Luxford, S., Hadwin, J. A., & Kovshoff, H. (2016).
Evaluating the effectiveness of a school-based cognitive behavioural therapy intervention for anxiety in
adolescents diagnosed with autism spectrum disorder. Journal of Autism and Developmental Disorders,
doi:http://dx.doi.org/10.1007/s10803-016-2857-7
Magiati, I., Moss, J., Charman, T., & Howlin, P. (2011).
Patterns of change in children with autism spectrum
disorders who received community based comprehensive interventions in their pre-school years: A seven
year follow-up study. Research in Autism Spectrum
Disorders, 5(3), 1016–1027.
Malhotra, D., & Sebat, J. (2012). Copy number variants:
Harbingers of a rare variant revolution in psychiatric
genetics. Cell, 148(6), 1223–1241.
Mattila, M.-L., Hurtig, T., Haapsamo, H., Jussila, K.,
Kuusikko-Gauffin, S., Kielinen, M., et al. (2010).
Comorbid psychiatric disorders associated with
Asperger syndrome/high functioning autism: A community and clinic-based study. Journal of Autism and
Developmental Disorders, 40, 1080–1093.
Mattilla, M.-L., Kielinen, M., Linna, S.-L., Jussilla, K.,
Ebeling, H., Bloigu, R., et al. (2011). Autism Spectrum
disorders According to DSM-IV-TR and comparison with DSM-5 draft criteria an epidemiological
study. Journal of the American Academy of Child &
Adolescent Psychiatry, 50(6), 583–592.
MacNeil, B. M., Lopes, V. A., & Minnes, P. M. (2009).
Anxiety in children and adolescents with autism
spectrum disorders. Research in Autism Spectrum
Disorders, 3, 1–21. doi:10.1016/j.rasd.2008.06.001.
Mandell, D. (2008). Psychiatric hospitalization among
children with autism spectrum disorders. Journal of
Autism and Developmental Disorders, 38, 1059–1065.
Marshall, C. R., Noor, A., Vincent, J. B., Lionel, A. C.,
Feuk, L., Skaug, J., et al. (2008). Structural variation of
chromosomes in autism spectrum disorder. American
Journal of Human Genetics, 82(2), 477–488.
Matson, J. L., Wilkins, J., & Ancona, M. (2008). Autism
in adults with severe intellectual disability: An
empirical study of symptom presentation. Journal of
Intellectual & Developmental Disability, 33, 36–42.
doi:10.1080/13668250701829837.
Mawhood, L., & Howlin, P. (1999). The outcome of a
supported employment scheme for high functioning
adults with autism or Asperger syndrome. Autism, 3,
229–254.
Mawhood, L., Howlin, P., & Rutter, M. (2000). Autism
and developmental receptive language disorder-a
comparative follow-up in early adult life I: Cognitive
and language outcomes. Journal of Child Psychology
and Psychiatry, 41, 547–559.
Mazefsky, C. A., Folstein, S. E., & Lainhart, J. E.
(2008). Overrepresentation of mood and anxiety
disorders in adults with autism and their first degree
relatives: What does it mean? Autism Research, 1,
193–197.
I.E. Drmic et al.
McGrew, S. G., Peters, B. R., Crittendon, J. A., &
Veenstra-VaderWeele, J. (2012). Diagnostic yield
of chromosomal microarray analysis in autism primary care practice: Which guidelines to implement?
Journal of Autism and Developmental Disorders,
42(8), 1582–1591.
McDougle, C. J., Erickson, C. A., Stigler, K. A., & Posey,
D. J. (2005). Neurochemistry in the pathophysiology
of autism. Journal of Clinical Psychiatry, 66(Suppl
10), 9–18.
Meyer, J. A., & Minshew, N. J. (2002). An update on
neurocognitive profiles in Asperger syndrome and
high-functioning autism. Focus on Autism and Other
Developmental Disabilities, 17(3), 152–160. http://
dx.doi.org/10.1177/10883576020170030501.
Moseley, D. S., Tonge, B. J., Brereton, A. V., & Einfeld,
S. L. (2011). Psychiatric comorbidity in adolescents
and young adults with autism. Journal of Mental
Health Research in Intellectual Disabilities, 4,
229–243.
Mouridsen, S. E., Rich, B., Isager, T., & Nedergaard, N. J.
(2008). Psychiatric disorders in individuals diagnosed
with infantile autism as children: A case control study.
Journal of Psychiatric Practice, 14, 5–12.
Muller, N., Myint, A., & Schwarz, M. J. (2015).
Immunology and psychiatry: From basic research
to therapeutic interventions. Cham: Springer
International Publishing.
Mundy, P., & Jarrold, W. (2010). Infant joint attention, neural networks and social cognition. Neural Networks,
23, 985–997. doi:10.1016/j.neunet.2010.08.009.
Mundy, P., Sullivan, L., & Mastergeorge, A. M. (2009).
A parallel and distributed processing model of joint
attention, social cognition, and autism. Autism
Research, 2, 2–21.
Muris, P., Steerneman, P., Merckelbach, H., Holdrinet, I.,
& Meesters, C. (1998). Comorbid anxiety symptoms
in children with pervasive developmental disorders.
Journal of Anxiety Disorders, 12, 387–393.
Narendorf, S. C., Shattuck, P. T., & Sterzing, P. R. (2011).
Mental health services among adolescents with an
autism spectrum disorder. Psychiatric Services, 62,
975–978.
National Autism Center. (2015). Findings and conclusions: National standards project, phase 2. Randolph,
MA: Author.
National Autism Center. (2009). National Standards
Report – Addressing the need for evidence-based
practice guidelines for Autism Spectrum Disorders.
Massachusetts: National Autism Center.
National Research Council. (2001). Educating young
children with autism. Washington, DC: National
Academy Press.
Newman, L., Wagner, M., Knokey, A. M., Marder, C.,
Nagle, K., Shaver, D., & Wei, X. (2011). The posyhigh school outcomes of young adults with disabilities up to 8 years after high school. Menlo Park: SRI
International.
Nordin, V., & Gillberg, C. (1998). The long-term course
of autistic disorders: Update on follow-up studies.
Acta Psychiatrica Scandinavica, 97, 99–108.
Life Course Health Development in Autism Spectrum Disorders
Noterdaeme, M., Wriedt, E., & Höhne, C. (2010).
Asperger's syndrome and high-functioning autism:
Language, motor and cognitive profiles. European
Child & Adolescent Psychiatry, 19(6), 475–481.
Palac, S., & Meador, K. J. (2011). Antiepileptic drugs and
neurodevelopment: An update. Current Neurology
and Neuroscience Reports, 11(4), 423–427.
Palucka, A. M., Bradley, E., & Lunsky, Y. (2008). A case
of unrecognized intellectual disability and autism
misdiagnosed as schizophrenia: Are there lessons to
be learned? Mental Health Aspects of Developmental
Disabilities, 11(2), 55–60.
Parr, J. R., Le Couteur, A., Baird, G., Rutter, M., Pickles,
A., Fombonne, E., Bailey, A. J., & International
Molecular Genetic Study of Autism Consortium.
(2011). Early developmental regression in autism
spectrum disorder: Evidence from an international
multiplex sample. Journal of Autism & Developmental
Disorders, 41(3), 332–340.
Paul, R. (2008). Communication development and assessment. In K. Chawarska, A. Klin, & F. Volkmar (Eds.),
Autism Spectrum disorders in infants and toddlers:
Diagnosis, Assessment, and treatment (pp. 76–103).
New York: Guilford Press.
Pinto, D., Pagnamenta, A. T., Klei, L., Anney, R., Merico,
D., Regan, R., et al. (2010). Functional impact of
global rare copy number variation in autism spectrum
disorders. Nature, 466(7304), 368–372.
Piven, J., & Rabins, P. (2011). Autism spectrum disorders
in older adults: Toward defining a research agenda.
Journal of the American Geriatrics Society, 59(11),
2151–2155.
Perkins, E. A., & Berkman, K. A. (2012). Into the
unknown: Aging with autism spectrum disorders.
American Journal on Intellectual and Developmental
Disabilities, 117(6), 478–496.
Perlman, L. (2000). Adults with Asperger disorder misdiagnosed as schizophrenic. Professional Psychology:
research & Practice, 31(2), 221–225. False.
Peters-Scheffer, N., Didden, R., Hubert, K., & Sturmey,
P. (2011). A meta-analytic study on the effectiveness
of comprehensive ABA-based early intervention programs for children with Autism Spectrum disorders.
Research in Autism Spectrum Disorders, 5(1), 60–69.
Polloway, E. A., Patton, J. R., & Serna, L. (2001).
Strategies for teaching learners with special needs.
Upper Saddle River: Merrill Prentice Hall.
Potvin, M. C., Snider, L., Prelock, P. A., Wood-Dauphinee,
S., & Kehayia, E. (2015). Health-related quality of
life in children with high-functioning autism. Autism,
19(1), 14–19.
Power, R. A., Kyaga, S., Uher, R., Maccabe, J. H.,
Långström, N., Landen, M., et al. (2012). Fecundity
of patients with schizophrenia, Autism, bipolar disorder, depression, anorexia nervosa, or substance abuse
vs their unaffected siblings. Archives of General
Psychiatry, 12, 1–8.
Raphael, D., Brown, I., Renwick, R., & Rootman, I.
(1996). Assessing the quality of life of persons with
developmental disabilities: Description of a new
271
model, measuring instruments, and initial findings.
International Journal of Diability, 43(1), 25–42.
Reichow, B. (2012). Overview of meta-analyses on early
intensive behavioral intervention for young children
with autism spectrum disorders. Journal of Autism and
Developmental Disorders, 42, 512–520.
Reichow, B., & Wolery, M. (2009). Comprehensive synthesis of early intensive behavioral interventions
for young children with autism based on the UCLA
young Autism Project model. Journal of Autism and
Developmental Disorders, 39(1), 23–41.
Renty, J., & Roeyers, H. (2006). Quality of life in highfunctioning adults with autism spectrum disorder: The
predictive value of disability and support characteristics. Autism, 10, 511–524.
Ritvo, E. R., Jorde, L. B., Mason-Brothers, A., Freeman, B.
J., Pingree, C., Jones, M. B., et al. (1989). The UCLAUniversity of Utah epidemiologic survey of autism:
Recurrence risk estimates and genetic counseling.
American Journal of Psychiatry, 146(8), 1032–1036.
Robinson, S., Goddard, L., Dritschel, B., Wisley, M., &
Howlin, P. (2009). Executive functions in children
with autism spectrum disorders. Brain and Cognition,
http://dx.doi.org/10.1016/j.
71(3),
362–368.
bandc.2009.06.007
Rogers, S. J. (2009). What are infant siblings teaching us
about autism in infancy? Autism research : Official
Journal of the International Society for Autism
Research, 2(3), 125–137.
Rondeau, E., Klein, L. S., Masse, A., Bodeau, N., Cohen,
D., & Guile, J.-M. (2011). Is pervasive developmental
disorder not otherwise specified less stable than autistic disorder? A meta-analysis. Journal of Autism and
Developmental Disorders, 41, 1267–1276.
Ruble, L. A., & Dalrymple, N. J. (1996). An alternative
view of outcome in autism. Focus on Autism and
Other Developmental Disabilities, 11(1), 3–14.
Rumsey, J. M., Rapoport, J. L., & Sceert, W. R. (1985).
Autistic children as adults: Psychiatric, social, and
behavioral outcomes. Journal of the American
Academy of Child Psychiatry, 24(4), 465–473.
Russ, S. A., Larson, K., Tullis, E., & Halfon, N. (2014).
A lifecourse approach to health development:
Implications for the maternal and child health research
agenda. Maternal and Child Health Journal, 18(2),
497–510.
Russell, E., & Sofronoff, K. (2005). Anxiety and
social worries in children with Asperger syndrome.
Australian and New Zealand Journal of Psychiatry,
39, 633–638.
Rutter, M. (1967). The autistic child. Royal Institute of
Public Health & Hygiene Journal, 30(4), 130–132.
Rutter, M. (1973). Why are London children so disturbed?
Proceedings of the Royal Society of Medicine, 66(12),
1221–1225.
Rutter, M. (1978). Diagnosis and definition of childhood
autism. Journal of Autism & Childhood Schizophrenia,
8(2), 139–161.
Rutter, M. (1982). Prevention of children's psychosocial disorders: Myth and substance. Pediatrics, 70(6), 883–894.
272
Rutter, M. (2005). Environmentally mediated risks for psychopathology: Research strategies and findings. Journal
of the American Academy of Child & Adolescent
Psychiatry, 44(1), 3–18.
Rutter, M., & Lockyer, L. (1967). A five to fifteen year
follow-up study of infantile psychosis I. Description
of sample. British Journal of Psychiatry, 113(504),
1169–1182.
Rutter, M., Greenfield, D., & Lockyer, L. (1967). A five
to fifteen year follow-up of infantile psychosis II
social and behavioral outcome. The British Journal of
Psychiatry, 113, 1183–1199.
Schreibman, L., Dawson, G., Stahmer, A. C., Landa, R.,
Rogers, S., et al. (2015). Naturalistic developmental
behavioral interventions: Empirically validated treatments for autism spectrum disorder. Journal of Autism
and Developmental Disorders, 45(8), 2411–2428.
Saxena, S., & Orley, J. (1997). Quality of life assessment:
The world health organization perspective. European
Psychiatry, 12, 263s–266s.
Schumann, C. M., Bloss, C. S., Barnes, C. C., Wideman,
G. M., Carper, R. A., Akshoomoff, N., et al. (2010).
Longitudinal magnetic resonance imaging study
of cortical development through early childhood in
autism. Journal of Neuroscience, 30, 4419–4427.
Seltzer, M. M., Krauss, M. W., Shattuck, P. T., Orsmond,
G., Swe, A., & Lord, C. (2003). The stmptoms
of autism spectrum disorders in adolescents and
adulthood. Journal of Autism and Developmental
Disorders, 33, 565–581.
Seltzer, M. M., Shattuck, P., Abbeduto, L., & Greenberg,
J. S. (2004). Trajectory of development in adolescents and adults with autism. Mental Retardation and
Developmental Disabilities, 10, 234–247.
Shattuck, P. T., Durkin, M., Maenner, M., Newschaffer,
C., Mandell, D. S., Wiggins, L., et al. (2009). Timing
of identification among children with an autism spectrum disorder: Findings from a population-based surveillance study. Journal of the American Academy of
Child & Adolescent Psychiatry, 48(5), 474–483.
Shattuck, P. T., Roux, A. M., Hudson, L. E., Taylor,
J. L., Maenner, M. J., & Trani, J.-F. (2012). Services
for adults with autism spectrum disorder. Canadian
Journal of Psychiatry, 57(5), 284–291.
Shattuck, P. T., Seltzer, M. M., Greenberg, J. S., Orsmond,
G. I., Bolt, D., Kring, S., et al. (2007). Change in
autism symptoms and maladaptive behaviors in adolescents with an autism spectrum disorder. Journal of
Autism and Developmental Disorders, 37, 1735–1747.
Shelton, J. F., Tancredi, D. J., & Hertz-Picciotto, I. (2010).
Independent and dependent contributions of advanced
maternal and paternal ages to autism risk. Autism
Research, 3(1), 30–39.
Sigman, M., & Kasari, C. (1995). Joint attention across
contexts in normal and autistic children. (pp. 189–
203). Hillsdale: Lawrence Erlbaum Associates, Inc.
Retrieved from http://search.proquest.com/docview/6
18769168?accountid=14771
Sigman, M., & McGovern, C. W. (2005). Improvement in
cognitive and language skills from preschool to adoles-
I.E. Drmic et al.
cence in autism. Journal of Autism and Developmental
Disorders, 35(1), 15–23.
Sigman, M., & Ruskin, E. (1999). Continuity and change
in the social competence of children with autism, down
syndrome, and developmental delays. Monographs of
the Society for Research in Child Development, 64(1),
1–139.
Simonoff, E., Pickles, A., Charman, T., Chandler, S.,
Loucas, T., & Baird, G. (2008). Psychiatric disorders in
children with autism spectrum disorders: Prevalence,
comorbidity, and associated factors in a populationderived sample. Journal of the American Academy of
Child and Adolescent Psychiatry, 47, 921–929.
Skokauskas, N., & Gallagher, L. (2010). Psychosis,
affective disorders, and anxiety in autistic spectrum
disorder: Prevalence and nosological considerations.
Psychopathology, 43, 8–16.
Smith, M., & Paulippen, L. (1999). Community integration and supported employment. In D. Zager (Ed.),
Autism: Identification, education, treatment (pp. 301–
319). Mahwah: Erlbaum.
State, M. W. (2010). The genetics of child psychiatric
disorders: Focus on autism and Tourette syndrome.
Neuron, 68(2), 254–269.
Sterling, L., Dawson, G., Estes, A., & Greenson, J. (2008).
Characteristics associated with presence of depressive
symptoms in adults with autism spectrum disorder.
Journal of Autism and Developmental Disorders, 38,
1011–1018.
Stoner, J. B., Angell, M. E., House, J. J., & Bock, S. J.
(2007). Transitions: Perspectives from parents of
young children with autism spectrum disorders
(ASD). Journal of Developmental Disabilities, 19,
23–39.
Strauss, K., Mancini, F., & Fava, L. (2013). Parent inclusion in early intensive behavior interventions for young
children with ASD: A synthesis of meta-analyses from
220 to 2011. Reseach in Developmental Disabilities,
34(9), 2967–2985.
Sullivan, P. F., Daly, M. J., & O'Donovan, M. (2012).
Genetic architectures of psychiatric disorders: The
emerging picture and its implications. Nature Reviews
Genetics, 13(8), 537–551.
Sun F, Oristaglio J, Levy SE, Hakonarson H, Sullivan N,
Fontanarosa J, Schoelles KM. 2015. Genetic testing
for developmental disabilities, intellectual disability, and Autism Spectrum disorder. Technical brief
no. 23. (prepared by the ECRI institute–Penn medicine evidence-based practice Center under contract
no. 290–2012-00011-I.) AHRQ publication no.15EHC024-EF. Rockville, MD: Agency for Healthcare
Research and Quality. www.effectivehealthcare.ahrq.
gov/reports/final.cfm.
Szatmari, P. (2011). New recommendations on autism
spectrum disorder shifting the focus from subtypes to
dimensions carriers potential costs and benefits. BMJ,
342. doi:10.1136/bmj.d2456.
Szatmari, P., & McConnell, B. (2011). Anxiety and mood
disorders in individuals with autism spectrum disorder. In D. G. Amaral, G. Dawson, & D. H. Geschwind
Life Course Health Development in Autism Spectrum Disorders
(Eds.), Autism spectrum disorders (pp. 330–338).
New York: Oxford University Press, Inc.
Szatmari, P., Bartolucci, G., Brenner, R., Bond, S., &
Rich, S. (1989). A follow-up study of high-functioning
autistic children. Journal of Autism and Developmental
Disorders, 19, 213–225.
Szatmari, P., Bryson, S. E., Boyle, M. H., Streiner, D. L.,
& Duku, E. (2003). Predictors of outcome among high
functioning children with autism and Asperger syndrome. Journal of Child Psychology and Psychiatry,
44(4), 529–528.
Szatmari, P., Georgiades, S., Duku, E., Bennett, T. A.,
Bryson, S., et al. (2015). Developmental trajectories of symptom severity and adaptive functioning
in an inception cohort of preschool children with
autism spectrum disorder. JAMA Psychiatry, 72(3),
276–283.
Szatmari, P., Jones, M. B., Zwaigenbaum, L., & MacLean,
J. E. (1998). Genetics of autism: Overview and new
directions. Journal of Autism and Developmental
Disorders, 28(5), 351–368.
Szatmari, P., Paterson, A. D., Zwaigenbaum, L., Roberts,
W., Brian, J., Autism Genome Project Consortium,
et al. (2007). Mapping autism risk loci using genetic
linkage and chromosomal rearrangements. Nature
Genetics, 39(3), 319–328.
Taylor, J. L. (2009). The transition out of high school
and into adulthood for individuals with autism and
for their families. International Review of Research
in Mental Retardation, 38, 1–32. doi:10.1016/
S0074-7750(08)38001-X.
Taylor, J. L., & Seltzer, M. M. (2010). Changes in the
autism behavioral phenotype during the transition
to adulthood. Journal of Autism and Developmental
Disorders, 40, 1431–1446.
Taylor, J. L., & Seltzer, M. M. (2011a). Changes in the
mother-child relationship during the transition to
adulthood for youth with autism spectrum disorders.
Journal of Autism and Developmental Disorders, 41,
1397–1410. doi:10.1007/s10803-010-1166-9.
Taylor, J. L., & Seltzer, M. M. (2011b). Employment and
post-secondary educational activities for young adults
with autism spectrum disorders during the transition
to adulthood. Journal of Autism and Developmental
Disorders, 41, 566–574.
Taylor, J. L., McPheeters, M. L., Sathe, N. A., Dove, D.,
Veenstra-VanderWeele, J., & Warren, Z. (2012). A systematic review of vocational interventions for young
adults with autism spectrum disorders. Pediatrics,
130(3), 531–538. doi:10.1542/peds.2012-0682.
Thomas, K. C., Ellis, A. R., McLaurin, C., Daniels, J.,
& Morrissey, J. P. (2007). Access to care for autismrelated services. Journal of Autism and Developmental
Disorders, 37, 1902–1912.
Totsika, V., Flece, D., Kerr, M., & Hastings, R. P. (2010).
Behavior problems, psychiatric symptoms, and quality of life for older adults with intellectual disability with and without autism. Journal of Autism and
Developmental Disorders, 40, 1171–1178.
273
Tomasello, M. (1995). Joint attention as social cognition.
In C. Moore, & P. J. Dunham (Eds.), Joint attention:
Its origins and role in development; joint attention:
Its origins and role in development (pp. 103-130,
Chapter vii, 286 Pages). Hillsdale: Lawrence Erlbaum
Associates, Inc.
Towbin, K. E. (2005). Pervasive developmental disorder
not otherwise specified. In F. R. Volkmar, A. Klin,
R. Paul, & D. J. Cohen (Eds.), Handbook of autism
and pervasive developmental disorders (Vol. 1, 3rd
ed., pp. 165–200). Hoboken: Wiley.
Tsakanikos, E., Costello, H., Holt, G., Bouras, N.,
Sturmey, P., & Newton, T. (2006). Psychopathology
in adults with autism and intellectual disability.
Journal of Autism and Developmental Disorders, 36,
1123–1129.
Tsakanikos, E., Costello, H., Holt, G., Sturmey, P., &
Bouras, N. (2007a). Behavior management problems
as predictors of psychotropic medication and use of
psychiatric services in adults with autism. Journal of
Autism and Developmental Disorders, 37, 1080–1085.
Tsakanikos, E., Sturmey, P., Costello, H., Holt, G., &
Bouras, N. (2007b). Referral trends in mental health
services for adults with intellectual disability and
autism spectrum disorders. Autism, 11, 9–17.
Turner, L. M., & Stone, W. L. (2007). Variability in outcome for children with an ASD diagnosis at age 2. The
Journal of Child Psychology and Psychiatry, 48(8),
793–802.
Van Acker, R., Loncola, J. A., & Van Acker, E. Y. (2005).
Rett syndrome: A pervasive developmental disorder.
In F. R. Volkmar, R. Paul, A. Klin & D. Cohen (Eds.),
Handbook of autism and pervasive developmental
disorders: Diagnosis, development, neurobiology, and
behavior (vol. 1) (3rd ed) (pp. 126–164, Chapter xxv,
703 Pages). Hoboken: John Wiley & Sons Inc.
VanBergeik, E., Klin, A., & Volkmar, F. (2008).
Supporting more able students on the autism spectrum: College and beyond. Journal of Autism
and Developmental Disorders, 38, 1359–1370.
doi:10.1007/s10803-007-0524-8.
van Heijst, B. F. C., & Geurts, H. M. (2015). Quality of life
in autism across the lifespan: A meta-analysis. Autism,
19(2), 158–167. doi:10.1177/1362361313517053.
Vaughan Van Hecke, A., Mundy, P., Acra, C. F., Block,
J. J., Delgado, C. E. F., Parlade, M. V., et al. (2007).
Infant joint attention, temperament, and social competence in preschool children. Child Development,
78(1), 53–69.
Veltman, J. A., & Brunner, H. G. (2012). De novo mutations in human genetic disease. Nature Reviews
Genetics, 13(8), 565–575.
Venter, A., Lord, C., & Schopler, E. (1992). A follow-up
study of high-functioning autistic children. Journal of
Child Psychology and Psychiatry, 33(3), 489–507.
Volkmar, F. R. (2004). Adolescence and sexuality. In
F. R. Volkmar & L. A. Wiesner (Eds.), Healthcare
for children on the autism spectrum (pp. 245–260).
New York: Harbour House.
274
Volkmar, F. R., & McPartland, J. (2014). From Kanner
to DSM-5: Autism as an evolving diagnostic concept.
Annual Review of Clinical Psychology, 10, 193–212.
Volkmar, F. R., & Cohen, D. J. (1991). Comorbid association of autism and schizophrenia. The American
Journal of Psychiatry, 148(12), 1705–1707.
Volkmar, F. R., Koenig, K., & State, M. (2005). Childhood
disintegrative disorder. In F. R. Volkmar, A. Klin,
R. Paul, & D. J. Cohen (Eds.), Handbook of autism
and pervasive developmental disorders (Vol. 1, 3rd
ed., pp. 70–78). Hoboken: Wiley.
Volkmar, F. R., Klin, A., & McPartland, J. (2014).
Asperger's syndrome: An overview. In J. McPartland,
A. Klin, & F. Volkmar (Eds.), Asperger syndrome:
Assessing and treating high-functioning autism spectrum disorders (pp. 1–42). New York: Guilford Press.
Watts, G. (2012). More psychiatrists attack plans for
DSM-5. BMJ, 344, e3357. http://dx.doi.org/10.1136/
bmj.e3357
Weitlauf, A.S., McPheeters, M.L., Peters, B., Sathe,
N., Travis, R., Aiello, R., Williamson, E., VeenstraVanderWeele, J., Krishnaswami, S., Jerome, R.,
Warren, Z. (2014). Therapies for Children With
Autism Spectrum Disorder: Behavioral Interventions
Update. Comparative Effectiveness Review No. 137.
(Prepared by the Vanderbilt Evidence-based Practice
Center under Contract No. 290–2012-00009-I.)
AHRQ Publication No. 14-EHC036-EF. Rockville,
MD: Agency for Healthcare Research and Quality.
www.effectivehealthcare.ahrq.gov/reports/final.cfm
I.E. Drmic et al.
Wing, L. (1980). Childhood autism and social class: a
question of selection? British Journal of Psychiatry,
137, 410–417.
White, S., Oswald, D., Ollendick, T., & Scahill, L. (2009).
Anxiety in children and adolescents with ASD.
Clinical Psychology Review, 29, 216–229.
Wolff, J. J., Gu, H., Elison, J. T., Styner, M., Gouttard, S.,
et al. (2012). Differences in white matter fiber tract
development present from 6 to 24 months in infants
with autism. The American Journal of Psychiatry,
169, 589–600.
Woodbury-Smith, M. R., & Volkmar, F. R. (2009).
Asperger syndrome. European Child & Adolescent
Psychiatry, 18(1), 2–11.
Woolfenden, S., Sarkozy, V., Ridley, G., & Williams, K.
(2012). A systematic review of the diagnostic stability of autism spectrum disorder. Research in Autism
Spectrum Disorders, 6, 345–354. doi:10.1016/j.
rasd.2011.06.008.
Zachor, D. A., & Itzchak, E. B. (2010). Treatment
approach, autism severity and intervention outcomes
in young children. Research in Autism Spectrum
Dosorders, 4(3), 425–432.
Zwaigenbaum, L., Bryson, S. E., Szatmari, P., Brian,
J., Smith, I. M., Roberts, W., et al. (2012). Sex differences in children with autism spectrum disorder
identified within a high-risk infant cohort. Journal
of Autism and Developmental Disorders, 42(12),
2585–2596.
Open Access This chapter is licensed under the terms of the Creative Commons Attribution 4.0 International License
(http://creativecommons.org/licenses/by/4.0/), which permits use, sharing, adaptation, distribution and reproduction in
any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license and indicate if changes were made.
The images or other third party material in this chapter are included in the chapter’s Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the chapter’s Creative Commons license
and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain
permission directly from the copyright holder.
Self-Regulation
Megan McClelland, John Geldhof, Fred Morrison,
Steinunn Gestsdóttir, Claire Cameron, Ed Bowers,
Angela Duckworth, Todd Little,
and Jennie Grammer
1
Self-Regulation
Self-regulation has received enormous attention
in recent years as a key predictor of a variety of
outcomes, including obesity (Evans et al. 2012),
school readiness (Blair and Razza 2007;
McClelland et al. 2007; Morrison et al. 2010),
academic
achievement
in
adolescence
(Duckworth et al. 2010b), and long-term health
and educational outcomes (McClelland et al.
2013; Moffitt et al. 2011). Although researchers
have focused on self-regulation from a diverse
set of perspectives (Geldhof et al. 2010;
M. McClelland (*)
Human Development and Family Sciences,
245 Hallie E. Ford Center for Healthy Children
and Families, Oregon State University,
Corvallis, OR 97331, USA
e-mail: megan.mcclelland@oregonstate.edu
J. Geldhof
Oregon State University, Human Development and
Family Sciences, Corvallis, OR, USA
F. Morrison
University of Michigan, Department of Psychology,
Ann Arbor, MI, USA
S. Gestsdóttir
University of Iceland, Department of Psychology,
Reykjavik, Iceland
C. Cameron
University at Buffalo, SUNY, Learning and
Instruction, Buffalo, NY, USA
McClelland et al. 2010), there is consensus that
self-regulation has important implications for
individual trajectories of health and well-being
across the life course. Indeed, over a decade ago,
it was suggested that “understanding self-regulation is the single most crucial goal for advancing
the understanding of development” (Posner and
Rothbart, 2000, p. 427).
Self-regulation is fundamental to successful
accomplishment of adaptive developmental
tasks at all stages of life. In the field of maternal
and child health, a recent emphasis utilizing a
life course health development (LCHD) perspective has shed new light on how these trajectories are shaped by dynamic mechanisms such
as self-regulation. This perspective is captured
by the seven LCHD principles—as described
by Halfon and Forrest (2017)—which are also
consistent with the relational developmental
E. Bowers
Clemson University, Youth Development Leadership,
Clemson, SC, USA
A. Duckworth
University of Pennsylvania, Department of
Psychology, Philadelphia, PA, USA
T. Little
Texas Tech University, Department of Educational
Psychology and Leadership, Lubbock, TX, USA
J. Grammer
University of California, Los Angeles, Graduate
School of Education and Information Studies,
Los Angeles, CA, USA
© The Author(s) 2018
N. Halfon et al. (eds.), Handbook of Life Course Health Development,
DOI 10.1007/978-3-319-47143-3_12
275
M. McClelland et al.
276
systems (RDS) perspective in the field of human
development.
The development of self-regulation is a prime
example of many of the LCHD principles in
action. For example, the notion that health develops continuously over the life span would imply
that individual pathways in self-regulation skills
are formed partly through life course transitions
and turning points or the points in a person’s life
which can influence developmental pathways in
either positive (protective) or negative (maladaptive) ways, and in fact this is the case. Similarly,
the notion that the timing and structure of environmental exposures are important for health
development applies very well to self-regulation,
the development of which is significantly and
adversely affected by persistent and chronic
stress, especially prenatally and in the first few
years of life. (Conversely, protective factors such
as sensitive and engaged caregiving can be a buffer for a child’s development of these skills during this time.) Additionally, the LCHD notion
that the rhythm of human development is a result
of synchronized timing of molecular, physiological, behavioral, and evolutionary processes and
that the synchronization of these processes contributes to the enormous individual variability in
health development over time is also relevant to
self-regulation.
Another illustration of the degree to which the
development of self-regulation serves as a powerful example of the LCHD framework and its
underlying principles in action is the fact that, at
a time in history when the importance of children’s self-regulation is perhaps greater than in
previous decades due to an increasing academic
focus in school settings, children and youth are
using media to a much greater extent than ever
before, a trend which could be detrimental to the
development of these essential skills. This mismatch between the demands of the environment
and the capacities of the developing individual is
well described by the LCHD principles, which
emphasize how evolution enables and constrains
health development pathways and plasticity, how
different aspects of development are intertwined
over time (e.g., biobehavioral development is
connected to sociocultural development), and
how efforts to promote more optimal health
development can promote survival and enhance
thriving by countering the negative impact of
these kinds of mismatches.
Finally, the LCHD principles capture the
dynamic and complex nature of health development and emphasize that development emerges
as a result of person interactions at multiple levels. This speaks to the importance of integrating
interventions both vertically—meaning along
primary, secondary, and tertiary care continua—
and horizontally, that is, across domains of function (i.e., biological, behavioral, social), as well
as longitudinally (e.g., across life stages and/or
generations). This is especially relevant here
because the capacity for self-regulation has been
shown to be highly malleable and because interventions to promote such skills have been shown
to be more effective when they are integrated
across different levels and contexts (Diamond
and Lee 2011; Raver et al. 2011).
Together, the LCHD principles will guide our
discussion of self-regulation, which are also consistent with an RDS perspective. After providing
a theoretical framework based on RDST, we will
view the seven principles of LCHD to better
understand the determinants and pathways of
self-regulation, methods for studying selfregulation, and translational issues. We conclude
by providing recommendations for better integrating the principles of LCHD with the study of
self-regulation.
1.1
Relational Developmental
Systems Theory
as a Framework
for Self-Regulation
While many processes currently subsumed under
the “self-regulation” moniker have been studied
from the earliest days of psychology (e.g., James
1890), the modern study of self-regulation truly
emerged as psychologists moved away from the
mechanistic neopositivism that dominated their
field during the middle part of the twentieth century. Work by Bandura (1969) and Mischel
(1968), for instance, rejected the notion of the
Self-Regulation
“black box” and instead emphasized the self (and
vicariously behavioral regulation by the self) as
the object of valid scientific inquiry. This renewed
focus on the self has made way for many of the
core concepts that frame modern developmental
science (e.g., that individuals are proactive agents
capable of influencing their own development;
Lerner 1982). Much of the recent work on selfregulation can be subsumed under the metatheoretical stance that Overton (e.g., 2010, 2013)
has termed relational developmental systems
((RDS) theory.
Similar to the principles of LCHD, RDS represents an approach to human development that
rejects the dualistic separation of individual and
context (Overton 2013). Instead, like the principles of LCHD, RDST specifies that the individual is completely embedded as a locally
self-organized component of his or her larger
context. Development of the individual therefore
necessarily influences and is influenced by his or
her surrounding environment. These mutual
influences can be thought of co-regulation (i.e.,
action and development of the individual partially “regulate” and are partially “regulated” by
the surrounding context), resulting in what
Brandstädter (e.g., 2006) has called developmental regulations. Similarly, Lerner (e.g., 1985;
Lerner et al. 2011) has heuristically decomposed
this person-context system and has described
developmental regulations as mutually influential, bidirectional person-context interactions—
similar to LCHD Principle 3. Accordingly, across
the life span, individuals are active agents in the
mutually influential interactions among the variables from the integrated biological, social, cultural, and historical (or temporal) levels of the
dynamic developmental system (as in LCHD
Principles 1, 2, 7).
The co-regulative nature of the person-context
system described in RDST directly informs the
contemporary study of self-regulation. While
person and context are truly inseparable from the
RDST perspective, Gestsdottir and Lerner (e.g.,
2008) note that we can heuristically separate
developmental regulations into those that primarily arise from the individual (i.e., the self) and
those that primarily arise from the context. Using
277
this logic, they proceed to define self-regulation
as comprised of “the attributes involved in and
the means through which the individual contributes to developmental regulations…” (p. 203).
As a broadly defined construct, self-regulation
therefore entails cognitions, emotions, and
actions that arise within the individual and do not
differentiate between conscious and subconscious (or even automatic) action.
Differentiating between consciousness and
sub- or (non)conscious behavior has been a
recurring issue in the study of self-regulation,
and it is now widely acknowledged that all selfregulated action falls along a continuum ranging
from fully intentional to fully automatic. For
instance, work done by Bargh and colleagues
(e.g., Bargh et al. 2001) clearly shows that subconscious goals can influence (i.e., regulate)
behavior outside of the actor’s explicit awareness. Similarly, Gestsdottir and Lerner (2008)
differentiate between organismic and intentional
self-regulation. Here, organismic self-regulation
occurs below the threshold of consciousness and
includes diverse actions ranging from the cardiovascular regulation of blood oxygen levels to the
regulation of outwardly directed behavior
through automatized goal structures. In contrast,
intentional self-regulation includes behavior that
the individual is consciously aware of, representing an agent’s intentional influence over the
person-context system. The remainder of this
chapter focuses specifically on intentional selfregulation. In total, self-regulation may be
defined as “the ability to flexibly activate, monitor, inhibit, persevere and/or adapt one's behavior, attention, emotions and cognitive strategies
in response to directions from internal cues,
environmental stimuli and feedback from others,
in an attempt to attain personally-relevant goals”
(Moilanen 2007, p. 835).
2
Definitions
of Self-Regulation
The study of self-regulation lacks integration
across the life span. Theories that approach
self-regulation within a given period of the life
M. McClelland et al.
278
span are often not integrated with each other
nor are they usually integrated with theories
that focus on subsequent or preceding life periods. In this section, we briefly review several of
the major conceptualizations of self-regulation
in an attempt to highlight the complexity of
self-regulated processes in children and youth.
Inherent in these conceptualizations and definitions are the seven principles of LCHD, which
have important implications for the concepts of
turning points and transitions, how mismatches
can occur in development, and the need to integrate interventions across multiple levels of
influence.
increase throughout adolescence and into early
adulthood (e.g., Hooper et al. 2004). Finally,
working memory is an aspect of executive functioning that includes the ability to actively work
on and process information. In young children, it
is demonstrated by children’s ability to remember and follow instructions (Gathercole et al.
2004; Kail 2003).
The early years are a sensitive period of brain
development, which closely parallel the development of EF. Understanding how EF develops during this developmental window has important
implications for biological, cognitive, and social
development.
2.1
2.2
Executive Functioning
As an instantiation of self-regulation, the study of
executive function (EF) emphasizes the fluid,
cognitive processes that underlie self-regulated
action. While the precise definition of which
skills and processes constitute EF may vary
across studies, researchers studying selfregulation have emphasized a few key skills. In
particular, researchers have studied the importance and development of agentic control over
one’s attention, inhibitory control, and working
memory (McClelland et al. 2010). Research
addressing the development of attentional control
describes the transition from simple arousal to
fully endogenous attention across the first few
years of life (e.g., Colombo 2001) and the subsequent development of attentional capacities from
childhood to late life (e.g., Posner and Rothbart
1998). Attentional processes play a major role in
self-regulated action (e.g., Norman and Shallice’s
(1986) Supervisory Attentional System) and may
especially relate to emotion regulation in infants
and children (Sheese et al. 2008). Children begin
to display inhibitory control by approximately
3 years of age (Posner and Rothbart 1998), a time
that corresponds to the onset of endogenous
attention and also corresponds to the transition
out of Piaget’s preoperational stage (see Geldhof
et al. 2010 for a brief discussion). Inhibitory control continues to develop throughout childhood
(e.g., Backen Jones et al. 2003) and continues to
Self-Regulation
Versus Self-Control
The literature does not consistently distinguish
between the concepts of self-regulation and
self-control, with many authors using the terms
interchangeably. Other authors consider selfregulation and self-control as distinct processes, which follow a sensitive period of
development in infancy. For instance, Kopp
(1982) describes self-control as developing at
around 24 months of age and as including the
ability to behave according to a caregiver’s
requests and to adhere to social expectations in
the absence of external monitors. She distinguishes this from self-regulation, which instead
develops when a child is approximately
36 months old and represents an internalization
of self-control that allows for a degree of flexibility, allowing children to meet the changing
demands of a dynamic context. According to
Kopp, the distinction between self-control and
self-regulation is therefore “a difference in
degree, not in kind” (Kopp 1982, p. 207). In
other words, self-regulation is an outgrowth of
self-control that allows for flexible adaptation
to real-world demands but which develops rapidly over the infant and toddler years. As such,
this progression reflects the principles of LCHD
especially for our understanding of how transitions and sensitive periods influence self-regulation development.
Self-Regulation
2.3
Effortful Control
In addition to the terms executive functions, selfregulation, and self-control, effortful control is a
related construct that stems from the temperament literature. Rothbart and colleagues have
defined the effortful control dimension of childhood temperament as “the ability to inhibit a
dominant response to perform a subdominant
response” (Rothbart and Bates 1998, p.137).
Measures of effortful control for preschool children encompass several facets, including attention focusing and inhibitory control over
inappropriate impulses (Rothbart et al. 2001).
Rothbart distinguishes effortful control from two
temperament factors that encompass more reactive (i.e., less voluntary) tendencies: surgency/
extraversion and negative affect. Moreover,
effortful control seems highly related, both conceptually and empirically, to self-control and
conscientiousness in adolescents and adults
(Eisenberg et al. (2012), under review). While
this definition closely reflects cognitive inhibition, effortful control is instead considered an
aspect of children’s temperament that develops in
tandem with the development of endogenous
attention. Research on infant temperament has
not found a complete analogue to effortful control, for instance, with factor analyses instead
uncovering a factor called orienting/regulation
(e.g., Garstein and Rothbart 2003). Orienting/
regulation contains many “regulatory” components similar to effortful control (e.g., orienting,
soothability) but lacks a truly effortful
component.
Effortful control incorporates the influence
of temperament that infants are born with, along
with the influence of the environment, including
quality of caregiving. This dynamic coaction
can be seen in the temperamental concept of
“goodness of fit.” Goodness of fit refers to the
match (or mismatch) between children’s temperamental states and the quality of caregiving
and temperament of their parents/caregivers.
When there is a positive fit or match between
children and caregivers, children’s development
of self-regulation is optimized. In contrast,
when a mismatch occurs, there is greater potential for difficulty with self-regulation and related
279
outcomes. Thus, effortful control is especially
relevant to understanding self-regulation through
an LCHD framework.
2.4
Delay of Gratification
Delay of gratification is another approach to selfregulation with close ties to both inhibition and
attention. Mischel and colleagues (e.g., Mischel
and Ebbesen 1970) originally studied delay of
gratification using the now-famous delay of gratification task with children. In this task, a
researcher shows a child two rewards (e.g., a
single marshmallow versus several marshmallows) and asks the child which reward he or she
would prefer. Subsequent research has adapted
this task for adults by varying the value of the
rewards—sometimes making them hypothetical—and by extending the delay time to a month
or longer (e.g., Fortsmeier et al. 2011; Duckworth
and Seligman 2005).
Regardless of the delivery, inherent in the construct is the integration of emotion with cognition
in their understanding of self-regulation. Mischel’s
research especially links the ability to delay gratification to endogenous attention through what he
and his colleagues have called the cognitive-affective processing system (e.g., Mischel and Ayduk
2004). This work has shown that when the rewards
are visible to children during the delay period,
children who distract their attention away from the
reward delay longer than children that do not
(Mischel et al. 1972). Similarly, children who
attend only to the cool, non-motivating, features of
the reward (e.g., by treating the actual reward as if
it is instead a picture of the reward) delay longer
than children who do not (Moore et al. 1976).
Delay of gratification thus complements the principles of LCHD by assuming that self-regulated
behavior includes the transactional processes of
emotion and cognition.
2.5
Emotion Regulation
Although the study of emotion regulation is a
complete area of the literature unto itself, there
is some important overlap with the study of
M. McClelland et al.
280
self-regulation more generally defined. Infants’
early regulatory tasks involve regulating their
reactions to stimuli, including affective,
temperament-based reactions that fall under the
emotion regulation umbrella (Eisenberg et al.
2004). Emotion regulation means that children
can modulate their strong emotional reactions
with an appropriate strategy or combination of
strategies (Bridges et al. 2004). Stansbury and
Zimmerman (1999) describe four types of emotion regulatory strategies: instrumental or trying
to change the situation (e.g., bidding for caregiver attention), comforting or soothing oneself
without changing the situation (e.g., thumb-sucking), distraction or redirecting attention elsewhere (e.g., looking away), or cognitive, which is
thought to be the most sophisticated and includes
reframing the situation in a positive light, bargaining, or compromising. Importantly, children
use different strategies depending on their individual characteristics as well as the situational
context (Zimmermann and Stansbury 2003). This
line of work demonstrates that the regulation of
attention and emotion is closely interrelated and
also reflects the principles of LCHD.
Together, the different definitions of selfregulation share many common conceptual
underpinnings and are relevant to how these
skills develop in individuals across the life span.
They also apply to the key principles of LCHD. In
the next section, we apply these principles to the
developmental processes of self-regulation.
3
Developmental Processes
of Self-Regulation
As noted above, the principles of LCHD can
help to inform our understanding of the development of self-regulation. We orient our discussion around these principles by employing
three lenses through which to view the development of self-regulation: (1) the lens of transitions and turning points, (2) the lens of
mismatches, and (3) the lens of intervention
integration. We include important individual,
contextual, and sociocultural factors that influence the development of these skills over time
since such information is critical for developing effective ways to help promote strong selfregulation in individuals.
3.1
Transitions and Turning Points
in the Development
of Self-Regulation
Because of the malleability in self-regulation evident throughout the life course, there are many
transitions and turning points for the development of these skills. The early childhood years
represent one important time in the life course
because they constitute a sensitive period for the
development of self-regulation and underlying
executive function skills. This makes it especially
important for children’s early biological, cognitive,
and
social-emotional
development
(Diamond 2002; Carlson et al. 2013). As noted
above, children’s self-regulation undergoes rapid
change during early childhood, which parallels
brain development, especially of the prefrontal
cortex (e.g., Diamond 2002). The translation of
this development can be seen in turning points in
development, one of which is the transition to
formal schooling for young children.
3.1.1
The Transition to Schooling
as a Turning Point
for Self-Regulation
Several lines of research point to relations
between schooling and self-regulation as a developmental turning point for children. Evidence
points to bidirectional relations between the biological and cognitive factors predicting development of self-regulation as well as the influence of
context such as the schooling environment (e.g.,
Diamond 2002; Carlson et al. 2013; Morrison
et al. 2010). Although much research focuses on
how individual factors influence self-regulation
(e.g., temperament, neurodevelopment of the
prefrontal cortex), research has also examined
how contextual factors such as schooling may
influence self-regulation. For example, researchers have suggested that differences in selfregulation across cultures may be due to early
instructional environments (Morrison et al. 2010)
Self-Regulation
as well as other factors such as temperamental
variables (Hsu et al. 1981) or the prevalence of
particular genes (Chang et al. 1996) that might
contribute to observed advantages in selfregulation (Sabbagh et al. 2006).
Research looking at the transition to formal
schooling has also used a natural experiment
(designated “school cutoff”) design, which examines children whose birth dates cluster closely on
either side of the cutoff date for entering formal
schooling (e.g., kindergarten in the United
States). This method effectively equates the two
groups of children on age (Morrison et al. 2010).
Using this methodology, results from recent
quasi-experimental and experimental investigations have provided further evidence for the
importance of schooling in the development of
self-regulation. For example, Burrage et al.
(2008) examined the influence of experience in
preschool on growth of word decoding, working
memory, and inhibitory control. This quasiexperimental work suggests that schooling, and
more specifically the years of prekindergarten
and kindergarten, improves working memory for
children who attend school compared with sameage peers who, because of arbitrary school cutoff
dates, do not attend at the same time (Burrage
et al. 2008). Together this research suggests that
the early childhood years provide a sensitive
period for the development of self-regulation,
which is influenced by both individual and contextual factors.
3.1.2
Adolescence as a Turning Point
for Self-Regulation
In adolescence, children experience another sensitive period of development, especially for selfregulation. Adolescence, the second decade of
life, is a period of ontogeny characterized by
extraordinary biological, social, and ecological
changes (Lerner and Steinberg 2009). Cognitive
and social development means that the capacities
necessary for advanced, adult-like self-regulation
may for the most part emerge in adolescence.
This is in large part due to the gradual maturation
of the prefrontal context. In particular, as the
frontal lobe develops, so does higher-order,
regulation-relevant cognition, such as metacogni-
281
tion and internalized control. In turn, these skills
enable adolescents to make better interpretations,
choices, and decisions about how to interact with
their environment, especially in accordance with
long-term goals (Brandstädter 2006; Larson
2011; Steinberg 2010). In addition, the formulation of an adaptive identity, which is a major
developmental task of adolescence, allows for the
construction of a personal future that informs
long-term decision-making and goal pursuit
(Brandtstädter 2006; McClelland et al. 2010).
After all, it is impossible to formulate a plan to
reach a long-term goal that has not yet been
determined. Finally, during adolescence, young
people may, for the first time, face decreased
probabilities of achieving major life goals (e.g.,
graduating from high school) that have long-term
consequences. This fact makes self-regulation
particularly pertinent during the adolescent
period (McClelland et al. 2010).
A growing body of research has confirmed the
relation between adolescents’ self-regulation
skills and positive and problematic behaviors.
In the last decade, a body of research has
advanced our understanding of how adolescents
regulate their own learning (Zimmerman 2002;
Zimmerman and Schunk 2001). Self-regulated
learning involves many goal-related skills, such
as the ability to set proximal learning goals, use
appropriate strategies for attaining the goal, selfevaluate the method one has chosen to achieve a
goal, and monitor one’s performance toward that
goal. The use of self-regulated learning skills has
repeatedly been related to school achievement
(Miller and Byrnes 2001; Zimmerman and
Schunk 2001). Similarly, the use of selfregulatory behaviors of youth is positively related
to other positive outcomes, such as measures of
social competence and mental well-being, and
negatively related to indicators of problematic
development, such as sexual risk behaviors, substance abuse, depression, and anxiety (e.g.,
Gestsdottir et al. 2009; Massey et al. 2008; Quinn
and Fromme 2010). In addition, self-regulatory
skills may have particular significance for youth
living in high-risk environments. For instance,
Buckner et al. (2009) found that youth from very
low-income families fared better on a wide range
M. McClelland et al.
282
of developmental outcomes, ranging from academic achievement to anxiety, if they had adaptive self-regulation skills. The authors emphasize
that such skills help youth to cope with stressful
life events, making them less likely to be overwhelmed by the difficulties that they are faced
with, and as such, high levels of self-regulation
are considered a key factor in supporting youth’s
resiliency (Buckner et al. 2009; Quinn and
Fromme 2010). In spite of the growing evidence
that self-regulation has important implications
for healthy functioning in adolescence, as it does
in childhood, there has been limited developmental research on how such important, adult-like
processes develop in adolescence.
In sum, although the understanding about the
nature and development of self-regulatory processes is not complete, recent research confirms
the contribution of adaptive self-regulation to the
healthy development of children and youth.
Furthermore, some recent findings point to an
emerging theme and match both the principles of
LCHD and the RDS framework: complex, adultlike, self-regulatory processes appear to develop
in middle adolescence and continue to grow
through adolescence and early adulthood. In
addition, the function of self-regulation in adolescence may differ in function from that of
childhood and adulthood. As such, the structure
and function of self-regulation may be specific to
this age period and constitute a sensitive period in
development.
3.2
Mismatches (and Matches)
in the Development
of Self-Regulation
In addition to research pointing to the importance
of examining the transaction of how selfregulation develops across multiple levels of
analysis, the match (or mismatch) between different aspects of development is also important.
This can be seen in the notion of goodness of fit,
taken from the child temperament literature,
where an individual’s characteristics and skills
may not fit with those of the environment, such as
the characteristics of caregivers. In the develop-
ment of self-regulation, a child’s individual characteristics and skills may be adversely influenced
by the aspects of their environment, such as
adverse childhood experiences, stress, poor parenting, maternal depression, and the influence of
the media and technology use.
3.2.1
Adverse Childhood Experiences
and Cumulative Risk
Recent research on adverse childhood experiences (ACEs) and toxic stress suggests that multiple and chronic environmental stressors can
have significant and adverse effects on the development of a host of outcomes throughout the life
span (Blair and Raver 2012; Shonkoff et al.
2012). For example, the early and chronic stress
experienced by children living in poverty can
have a profound influence on areas of the brain
most involved in the development of selfregulation (the prefrontal cortex [PFC]; e.g.,
Blair 2010; Blair and Raver 2012). One study
found that low-income children exhibited lower
prefrontal functioning compared to higherincome children. Specifically, the PFC functioning of low-income children in the study was
similar to the level of functioning of individuals
with damage to the PFC (Kishiyama et al. 2009).
In addition to effects on the developing
brain, ACEs are related to poorer executive
function and self-regulation, increased substance use, obesity, and risk-taking behaviors in
adolescents and adults (see Table 1). For example, one study found that children with cumulative risk exposure (e.g., poverty, family turmoil,
substandard housing) gain more weight during
the transition to adolescence than their more
advantaged peers, an effect mediated by lower
levels of self-regulation (Evans et al. 2012).
Such pernicious effects were predicted by
Walter Mischel and colleagues, whose hot/cool
model of self-control specified that stressful
life events would potentiate impulsive (“hot”)
system activity and attenuate slower, more
reflective and voluntary (“cool”) system activity (Metcalfe and Mischel 1999).
Research has also indicated that children from
low-income families are more likely to experience
family and housing instability, a lack of resources,
Self-Regulation
283
Table 1 Examples of direct and/or indirect relations
between self-regulation and health-related outcomes
Predictor
Self-regulation
Health-related outcomes
Obesity
Weight gain and loss
Addiction and substance use
Risk-taking behaviors
Cardiovascular disease
Asthma
Autoimmune diseases
Depression
Liver cancer
Academic achievement
School readiness
Educational attainment
Economic well-being (savings
behavior, financial security,
occupational prestige)
Lack of criminal convictions
Health behaviors
Physical activity
Nutritious eating
ADHD
and lower-quality learning environments in the
home (e.g., Gershoff et al. 2007; Mistry et al.
2010; Obradovic 2010; Sektnan et al. 2010), all
of which have been linked to lower levels of selfregulation. For example, children facing cumulative risk factors may experience significant
difficulty with self-regulation in early childhood
(Wanless et al. 2011).
Partly because of this, children with chronic
environmental stressors are more likely to experience school failure, unemployment, poverty,
violent crime, and incarceration as adults.
Moreover, and perhaps most important for the
long-term implications of ACEs, these children
are less likely as adults to provide supportive
environments for their own children, who in turn
are at significant risk of demonstrating some of
these same issues. In addition to behavioral and
economic effects, chronic and toxic stresses
have been linked to biological changes including
premature aging and death, alterations in
immune functioning, and significant increases in
inflammatory markers. Related to this, ACEs
have been associated with a host of physical
health outcomes, including cardiovascular dis-
ease, liver cancer, asthma, autoimmune diseases,
and depression (Committee on Psychosocial
Aspects of Child and Family Health et al. 2012;
Shonkoff et al. 2012).
Together, this research suggests that ACEs,
toxic stress, and cumulative risk can significantly
impair the development of self-regulation in children. This is also an example of a potential mismatch between children’s own development and
the context in which they live. For example, it is
possible that children facing cumulative risk have
parents who provide fewer opportunities to practice self-regulation (Fuller et al. 2010; Wachs
et al. 2004). These children may also have higher
levels of stress, which interfere with the development of prefrontal cortex, experience more family and housing instability, and have fewer
learning and economic resources (Blair 2010;
Blair and Raver 2012). Thus, there may be few
opportunities for children to experience a positive match between their own developing skills
and those of the environment in which they live.
3.2.2 Parenting and Caregiving
As the research above indicates, poor parenting
can have significant and detrimental effects on
their children’s own self-regulation. For example,
extensive research documents the negative effects
that maternal depression can have on a range of
child outcomes, including self-regulation (Center
on the Developing Child 2011).
In contrast to the conflicted and non-supportive
parent-child relationships that undermine children’s ability to self-regulate, organized and predictable home environments and emotionally
positive parent-child relationships provide a context that allows for the development of selfregulatory competencies (e.g., Bowers et al.
2011; Brody and Ge 2001; Grolnick et al. 2000;
Lewin-Bizan et al. 2010; Moilanen et al. 2010).
For example, parenting that includes a focus on
supporting autonomy and setting limits has significantly predicted stronger self-regulation in
children compared to parenting that is more controlling and focused on compliance (Bernier
et al. 2010; Lengua et al. 2007). A similar line of
work in early childhood classrooms has established
the importance of orienting and organizing
M. McClelland et al.
284
teacher behaviors for children’s self-regulation,
engagement, and academic outcomes (Cameron
and Morrison, 2011; Cameron Ponitz et al. 2009).
Taken together, this work indicates the importance of structured and predictable environments
for helping children’s emerging self-regulatory
capacities. It also demonstrates the importance of
matches between children’s characteristics and
parenting characteristics and behaviors, which
complement the principles of the LCHD
perspective.
3.2.3 Media and Technology Use
Another example of a possible mismatch is the
increasing structure in school settings paired with
the high prevalence of media and television use
by children and adults. Children’s media and
technology use is rapidly increasing, but there
remains little evidence on the positive effects of
such media on children’s development, especially for very young children (Radesky et al.
2014). Many studies have found persistent negative effects of extended television and media
viewing on children’s short- and long-term development (Robertson et al. 2013), including inattention and attention deficit hyperactivity disorder
(ADHD)-related behaviors (Christakis et al.
2004; Nikkelen et al. 2014). These findings indicate that media use is related to poorer selfregulation and that households with heavy media
use may be a poor context for supporting children’s self-regulatory development. Thus, children’s increased media use may run counter to
the increased demands for self-regulated behavior in schools and society.
In addition to the issue of child media use is
the high prevalence of media use by adults and
parents. For example, parents who are distracted
by texting and being on mobile devices may not
be able to adequately respond to and parent their
children. Although limited research exists, one
study found that caregivers who used mobile
devices at a restaurant while with their children
were most often highly absorbed in the content
and were less attentive to the children they were
with. Those caregivers who were highly
absorbed in their mobile devices were also more
likely to respond harshly to child misbehavior
(Radesky et al. 2014). Thus, an increased inattention and distraction on the part of parents and
caregivers may provide children with fewer
opportunities to learn how to self-regulate themselves. Moreover, it is possible that although
children’s self-regulation is needed to successfully navigate increasing structured school settings, children and parents are using media to a
much greater extent than ever before, which
could be detrimental to the development of these
skills. This potential mismatch may have significant long-term implications and is an area ripe
for additional research.
3.3
Integrating Levels of Influence
in Self-Regulation
Interventions
Another LCHD lens through which to view selfregulation processes is the importance of integration across multiple levels of influence, especially
in the context of interventions. This integration
includes lateral integration or integration across
subject domains, vertical integration or integration across levels of analysis, and developmental
integration or integration across time. Because of
the evidence pointing to the malleability of selfregulation, there has been an explosion in recent
years in interventions aiming to foster the development of these skills.
Accumulating evidence suggests that interventions targeting children’s self-regulation at
various levels can be effective at improving
self-regulation and other outcomes. For example, at the sociocultural level, preschool curricula, such as Tools of the Mind, focus on social,
emotional, and executive function skills in addition to literacy and math. Research suggests that
program participation is related to significant
improvement in children’s self-regulation (Blair
and Raver 2014; Diamond et al. 2007), social
behavior (Barnett et al. 2008), academic outcomes (Blair and Raver 2014), and neuroendocrine function (e.g., levels of salivary cortisol
and alpha amylase; Blair and Raver 2014).
Self-Regulation
Some work, however, has not found significant
intervention effects (Farran et al. 2013), suggesting that more work is needed to fully understand the key components of intervention
effectiveness.
Other interventions that include multiple
levels of integration (e.g., at the parent, teacher,
and child level) are the Promoting Alternative
Thinking Strategies (PATHS) and the Head
Start REDI (Research-based, Developmentally
Informed) programs (Bierman et al. 2008a),
which focus on social-emotional skills and
self-regulation. Children receiving these interventions have demonstrated more socially
competent behavior (Domitrovich et al. 2007)
and significant improvements in self-regulation (Bierman et al. 2008b) compared to children in a control group. Another recent study
examining a broad intervention targeting
social-emotional learning and literacy development found that children in intervention
schools demonstrated improvements in a variety of social behaviors and self-regulation
skills (e.g., attention). Improvements were also
found in children’s early math and reading
achievement for those initially most at risk for
behavior problems (Jones et al. 2011).
Further evidence from a school-based intervention that included multiple levels of integration with teachers, mental health consultants,
and children (Raver et al. 2011) reveals that preschool children participating in the Chicago
School Readiness Project exhibited significantly
higher performance on self-regulation tasks than
did their peers in a control group. Moreover,
there was a mediating role of children’s EF on
pre-academic literacy and math skills. These
findings complement those of Connor and colleagues (2010) who also found that an instructional intervention—which emphasized teacher
planning, organization, classroom management,
and opportunities for students to work independently—was most beneficial for children who
started first grade with weaker self-regulation.
Similarly, a recent intervention focusing on
aspects of self-regulation (attentional flexibility,
working memory, and inhibitory control) integrated into classroom games found that partici-
285
pation in the intervention was significantly
related to gains in self-regulation skills and academic achievement compared to children in the
control group (Tominey and McClelland 2011;
Schmitt et al. 2015).
For children with ADHD, research has also
documented that interventions that focus on
strengthening aspects of self-regulation and
underlying executive function skills can be
beneficial (Reid et al. 2005). Such interventions have been found to help children improve
on task behavior, decrease inappropriate
behavior, and increase academic achievement,
although results have been somewhat weaker
for lasting improvement in academic skills
(DuPaul et al. 2011).
Overall, results from a growing number of
randomized control trials suggest that interventions designed to strengthen self-regulation can
improve children’s self-regulation, social
behavior, and academic achievement. It is not
known, however, if these effects persist over
time. More research is needed on the long-term
effects of such interventions and how interventions may work for different subgroups of children (e.g., those most at risk). Moreover,
following the principles of LCHD, interventions
tend to be most effective when they include
multiple levels of influence and are integrated
across domains of functioning and over time
(Jones and Bouffard 2012).
4
Self-Regulation and HealthRelated Outcomes
Although self-regulation has been conceptualized differently in a variety of fields and at different developmental periods, accumulating
evidence demonstrates the importance of selfregulation for a variety of outcomes. Moreover,
our view of self-regulation reflects both the
principles of LCHD and the RDS perspective.
Below we review research on predictive relations between self-regulation and important outcomes such as academic achievement and
educational attainment and health and wellbeing (see also Table 1).
M. McClelland et al.
286
4.1
Academic Achievement,
Educational Attainment,
and Economic Well-Being
Over a century ago, in a series of lectures for
schoolteachers near his home institution of
Harvard University, William James (1899)
declared that much of schoolwork was necessarily “dull and unexciting” in comparison with
other things children might be doing (pp. 104–
105). Consequently, James reasoned that students
who could voluntary control their attention
enjoyed a distinct advantage over students who
regularly succumbed to the “temptation to serve
aside to other subjects” (p.112). Alfred Binet,
Charles Spearman, and David Wechsler all made
similar observations. That three of the most
important figures in the history of intelligence
testing would individually highlight the importance of “will” as a necessary complement to talent is somewhat ironic, given that intellectual
aptitude, rather than self-regulation, was until
very recently given disproportionate emphasis in
the educational psychology literature.
Prospective longitudinal studies have confirmed James’s earlier intuitions. For young children, a large body of evidence now demonstrates
that self-regulation sets the stage for learning in
children even prior to formal schooling. For
example, self-regulation in preschool and during
the transition to kindergarten has uniquely predicted gains in academic achievement after controlling for child IQ and initial achievement
levels (von Suchodoletz et al. 2013; Blair and
Razza 2007; McClelland et al. 2007). In elementary school, strong kindergarten learning-related
skills (including self-regulation and social competence) significantly predicted higher reading
and mathematics achievement between kindergarten and sixth grade and growth in literacy and
mathematics from kindergarten to second grade
after controlling for prior achievement levels,
child IQ, and a host of background variables
(McClelland et al. 2006; see also Duncan et al.
2007; McClelland et al. 2006; McClelland et al.
2007; McClelland et al. 2000). Studies have also
documented the long-term contributions of selfregulation to practically significant outcomes
such as high school graduation and college completion (McClelland et al. 2013; Moffitt et al.
2011). In one recent study, a 4-year-old child
with one standard deviation higher ratings of
attention (one aspect of self-regulation) than
average had 49% greater odds of completing college by age 25 (McClelland et al. 2013).
In terms of economic well-being, the best evidence for the importance of self-regulation comes
from a longitudinal study by Moffitt et al. (2011).
Self-regulation was assessed using parent,
teacher, observer, and self-report ratings at multiple time points in the first decade of life in a
nationally representative sample of New
Zealanders who were followed into adulthood.
Childhood self-regulation predicted income, savings behavior, financial security, occupational
prestige, lack of substance use, and lack of criminal convictions. These benefits were partially
mediated by better decisions in adolescence,
including staying in high school, not becoming a
teenage parent, and not smoking. For a review of
the relevance of self-regulation to academic
achievement, including school readiness and lifetime educational attainment, see Duckworth and
Allred (2012).
4.2
Health and Well-Being
Self-regulation has been shown to be related to a
variety of health behaviors, including recovery
from physical illness or disabilities (e.g., exercise
during and after cardiac rehabilitation (Blanchard
et al. 2002), functional activity of patients undergoing surgical replacement of the hip or knee
(Orbell and Sheeran 2000), physical activity for
individuals in orthopedic rehabilitation (e.g.,
Ziegelmann et al. 2006, 2007), disease prevention (e.g., attendance for cervical cancer screenings, Sheeran and Orbell 2000; performance of
breast self-examinations, Orbell et al. 1997), and
general health (e.g., regulation of body weight
via dieting and exercising/sport activities,
Bagozzi and Edwards 1998; and increased consumption of nutritious foods and other dietary
behaviors [Anderson et al. 2001; Calfas et al.
2002; Jackson et al. 2005]). Many of these stud-
Self-Regulation
ies are framed by Gollwitzer’s model of action
phases (Gollwitzer 1990, 1996).
As an action theory, Gollwitzer’s model of
action phases focuses on the factors that determine how effective one is during the process of
setting a goal to actual goal attainment. A key
construct distinction within this model—and ultimately in predicting one’s success in behavior
change or goal attainment—is between goal
intentions and implementation intentions. A goal
intention indicates a desired behavior or outcome
and is a declaration of one’s commitment to a
goal. Implementation intentions, on the other
hand, specify the “when, where, and how of
responses leading to goal attainment…and thus
link anticipated opportunities with goal-directed
responses” (Gollwitzer 1999, p. 494). As a goal
intention states an individual’s commitment to a
specific goal, the implementation intention states
the individual’s commitment to certain actions in
an effort to attain that particular goal. Gollwitzer’s
model also highlights the contention that selfregulated actions fall along an intentionalautomatic continuum; forming implementation
intentions allows people to “strategically switch
from conscious and effortful control of their
goal-directed behaviors to being automatically
controlled by selected situational cues”
(Gollwitzer 1999, p. 495). In turn, implementation intentions promote goal attainment by helping to initiate action, above and beyond the
effects of goal intentions alone.
Studies applying Gollwitzer’s model to health
behavior have indicated that it is not only important for participants to have goal intentions, but it
is also imperative for them to form implementation intentions and make subsequent planning
strategies to work toward their goals. These strategies allow individuals to pinpoint when, where,
and how they will enact specific goal-related
behaviors. For example, Luszczynska (2006)
examined how well patients who suffered a myocardial infarction utilized physical activity planning strategy and performed moderate physical
activity after engaging in an implementation
intention intervention program. The results indicated that as compared to controls, patients who
participated in the implementation intention
287
intervention more frequently used their planning
strategies and maintained the same levels of
physical activity at 8 months after their infarction
as they did at 2 weeks after rehabilitation.
Furthermore, implementation intentions (as compared to goal intentions) may be more predictive
of health behaviors at later time points (Orbell
and Sheeran 2000; Ziegelmann et al. 2007).
When participants were asked to perform breast
self-examinations, those who made such planning strategies were more likely to perform the
behavior in the manner in which they originally
specified (i.e., time and place) and were less
likely to report forgetting to perform the behavior
(e.g., Orbell et al. 1997). Likewise, the formation
of such plans for breast self-examinations or to
attend cervical cancer screenings can lead to earlier enactment of goal intentions even among a
sample of highly motivated individuals (Orbell
and Sheeran 2000; Sheeran and Orbell 2000) and
influence motivation and adherence (Levack
et al. 2006).
Another work examining the role of intentional self-regulation in health-related behaviors
also focuses on specific self-regulatory cognitions and behaviors. Many studies have highlighted the importance of developing action and
coping plans for successful adoption and maintenance of healthy behaviors such as physical
activity and nutritious eating (e.g., Calfas et al.
2002; Sniehotta et al. 2005; Zeigelmann and
Lippke 2007). Behavioral interventions aimed at
initiating or increasing certain health behaviors—or aiding participants in reaching certain
health goals—were often more effective when
they included the creation of “action plans” (e.g.,
Calfas et al. 2002). The development of these
plans often included having the participant
explicitly identify the goals to pursue and sources
for social support or resources to be utilized for
achieving those goals. In some cases, the action
plans also included identifying possible obstacles
or barriers that might interfere with the implementation of their plans and solutions to overcome them (e.g., Calfas et al. 2002), but separate
“coping plans” were also used for that purpose.
For example, in a sample of 352 cardiac patients
undergoing rehabilitation, Sniehotta et al. (2005)
M. McClelland et al.
288
provided evidence that action planning and coping planning can be identified as distinct strategies; in addition, the combination of forming
both action plans and coping plans was more
effective in increasing health behaviors over time
than forming action plans alone. The additive
benefit of action and coping plans was replicated
in experimental designs (Sniehotta et al. 2006;
Sniehotta et al. 2005; Scholz et al. 2007).
A large body of research also points to the
importance of self-regulation for weight gain
and loss (e.g., Evans et al. 2012; Francis and
Susman 2009; Hofmann et al. 2014), addiction
(Baumeister and Vonasch 2014), and other
health-related outcomes (Moffitt et al. 2011).
Several recent studies have demonstrated that
poor self-regulation predicts unhealthy weight
gain, particularly in adolescence, a period
marked by pubertal changes that influence adiposity and greater latitude to make diet and exercise choices independent of parental control
(Duckworth et al. 2010a; Tsukayama et al. 2010).
In one study, children exposed to a number of
risk factors were significantly more likely to
gain weight during adolescence, which was
mediated by having significantly lower levels of
self-regulation (Evans et al. 2012). Adiposity, in
turn, is a robust predictor of physical vitality
later in life, suggesting one causal pathway linking childhood self-regulation to adult physical
health and, ultimately, mortality.
Issues with self-regulation have also been
implicated in ADHD, with ADHD often characterized as a disorder of self-regulation and underlying executive function components (Barkley
1997, 2011). For example, many individuals with
ADHD exhibit significant difficulties with the
core executive function components of selfregulation, including attentional or cognitive
flexibility, working memory, and inhibitory control. This can be seen in individuals who are inattentive, who lack behavioral inhibition, and who
have difficulty with planning, organizing, and
being goal-oriented. These issues can also lead to
difficulty with emotion regulation. Thus, individuals with ADHD are more likely to have problems with impulse control, be more reactive, and
have diminished social perspective taking abilities (Barkley 2011; Berwid et al. 2005). This
means that children with ADHD may have a
harder time stopping and thinking about a situation before reacting and illustrates why these
children are more at risk for peer rejection and
other behavior problems (Molina et al. 2009).
Children with ADHD also demonstrate significant problems with academic achievement,
which can also be linked back to difficulties with
behavioral and emotional aspects of selfregulation (DuPaul and Kern 2011).
5
Methods for Studying
Self-Regulation
As demonstrated by how self-regulation relates
to the principles of LCHD and RDS, selfregulation shows important transitions and sensitive periods, multiple levels of influence, and
person-context fit in the form of matches or mismatches that can affect health development. Our
understanding of these issues, however, hinges
on how self-regulation is measured and analyzed
in health-related research. In this section, we
examine recent research on ways to measure and
analyze self-regulation.
5.1
Measuring Self-Regulation
Self-regulation is generally treated as a slowly
developing phenomenon, meaning studies that
target the development of self-regulation can easily take advantage of the large sample, small time
point analyses that dominate research in healthrelated fields. Self-regulation research can
accordingly draw on the strengths of modern statistical methods such as latent variable structural
equation modeling, multilevel modeling, and
mixture modeling. In this vein, researchers readily acknowledge that one size rarely fits all
people. Advances in mixture modeling have
allowed us to appropriately model theories that
stem from the person-centered movement and
systems theories. Large sample research can be
Self-Regulation
facilitated by utilizing advances in modern missing data procedures to incorporate planned missing data collection designs. Such designs allow
researchers to collect all the data needed to utilize
modern analytic methods without burdening parents, teachers, or individuals with excessively
long surveys.
It is also important to note, however, that challenges exist with some of these methods because
self-regulation measures change over the developmental years and are often not strongly related
with each other. Thus, developing self-regulation
measures that are reliable and valid over a broad
age range and at important points of transition is
of particular importance. Some progress, however, has been made on this front. For example,
the National Institutes of Health (NIH) Toolbox
has developed brief assessments for a variety of
skills, including aspects of self-regulation, which
are appropriate to use with individuals throughout the life span (Zelazo et al. 2013).
In addition to measures that span a large age
range, other measures capture a broad set of children’s developmental skills, especially at school
entry. Some research has focused on populationbased measures that are based on teacher or caregiver ratings. One example is the Early
Developmental Instrument (EDI; Janus and
Offord 2007), which measures five developmental domains: social, emotional, physical, cognitive, and communicative. Although not
specifically focused on measuring self-regulation,
the measure includes items tapping aspects of
self-regulation mostly in the social and emotional
domains. The measure has been shown to be reliable and valid and significantly related to broad
measures of school readiness, although less
strongly related to direct assessments of children’s skills (Hymel et al. 2011). A strength of
this type of measure is the potential to capture a
range of children’s skills. A weakness, however,
is that there may be considerable construct overlap and variability in how teachers rate children.
An example of a more targeted measure is the
Head-Toes-Knees-Shoulders
(HTKS)
task
(McClelland et al. 2014), which specifically measures behavioral aspects of self-regulation. The
289
HTKS taps children’s ability to pay attention, use
working memory, and demonstrate inhibitory
control by doing the opposite of what was asked.
The task is most appropriate for young children
during the transition to formal schooling, which
is important because this time is a crucial period
for the development of self-regulation. A number
of studies have shown that the HTKS is reliable
and valid and significantly predicts academic
achievement in diverse groups of children in the
US, Asian, and European countries (McClelland
et al. 2007, 2014; von Suchodoletz et al. 2013;
Wanless et al. 2011).
In youth and adults, self-regulation is often
measured either using self-report, parent-report,
or teacher-report questionnaires, delay of gratification tasks, or, ideally, a multi-method battery of
measures. Such measures predict report card
grades and changes in report card grades over
time (Duckworth and Seligman 2005), but the
predictive validity of self-regulation for standardized achievement test scores, in contrast, is less
dramatic (Duckworth et al. 2012). One reason
that report card grades are differentially sensitive
to self-regulation may be their relatively greater
emphasis on effort on the part of the student, to
complete homework assignments on time and
with care, to come to class prepared and pay
attention when present, and to study for quizzes
and tests from provided materials. Notably, report
card grades predict persistence through college
better than standardized test scores, a testament
to the continued importance of self-regulation as
students move through the formal education system (Bowen et al. 2009).
5.1.1 Construct Diversity
The major limitation to measure self-regulation
stems from the fact that self-regulation is not a
single globally measurable construct. Instead,
self-regulation represents an individual’s agentic
attempts to reach distal outcomes by influencing
what Lerner (e.g., Gestsdottir and Lerner 2008)
has called person-context relations. The extant
diversity of theories and measures of selfregulation suggest that the apparently unitary
domain of self-regulation actually consists of
M. McClelland et al.
290
many oblique fragments that differentially influence behavior as a function of context. We therefore need refinements in the measures of and
theories about context-specific self-regulation.
Here, better measurement of the parts will better
inform the whole.
Complementing Nomothetic
Analyses with Idiographic
Analyses
In addition, if we truly see self-regulation as part
of an ongoing process that is unique to each individual, we must begin to complement our existing analyses with more idiographic examinations
of self-regulation over a variety of time spans
(e.g., moments, days). Idiographic analyses such
as dynamic factor analysis and p-technique have
a place in research, and it is important that selfregulation researchers begin to acknowledge this
role. We currently have a poor understanding of
self-regulation as an idiographic phenomenon. A
better understanding of intraindividual differences will allow greater insight into interindividual phenomena related to self-regulation as well
as its intraindividual development.
tion, we suggest key issues and next steps for
self-regulation research.
6.1
Integration in Conceptualizing
and Measuring
Self-Regulation
5.1.2
6
Issues for Future Research
The previous sections demonstrate that, across a
broad spectrum of disciplines, interest has
steadily mounted in self-regulation and related
constructs—executive function (EF), selfcontrol, and effortful control. A growing body of
research has shown the importance of selfregulation for children’s success in school, as
well as for subsequent health, wealth, and criminality (e.g., Moffitt et al. 2011). In addition, the
study of self-regulation can be informed by a
closer appreciation of the principles of LCHD
and RDS, including how turning points and transitions, mismatches, and intervention integration
influence self-regulation trajectories. Despite
advances in many areas, our understanding of
aspects of self-regulation, including the neurological underpinnings of these skills, and efforts
to intervene in the development of self-regulation
for children at risk remains limited. In this sec-
When studied from multiple perspectives and
fields, differences in how self-regulation is
defined and conceptualized arise in part because
its study stems from diverse research traditions
that use distinct methods to examine phenomena
across the life course. For example, research has
burgeoned in basic investigations of selfregulation, including understanding the underlying neurological and behavioral mechanisms
driving these skills in children, adolescents, and
adults (Blair and Raver 2012). It is also the case
that the particular domain of inquiry informs
where and how phenomena and individuals are
studied. Scholars sometimes refer to different
levels of analysis (e.g., neurological activation,
physiological responses, observed behavior, or
self-report) to clarify some of these differences.
More could be done, however, to provide better
integration across different disciplines and contexts to study the development and measurement
of these skills. For example, although the knowledge base of research on different aspects of selfregulation is deep, it lacks breadth, and most of
the work in this area has been conducted in convenience samples of middle-SES North
Americans. More research is needed on how selfregulation develops within different groups and
populations especially as it relates to the principles of LCHD.
Another critical issue is the need to move away
from deficit models of self-regulation (e.g., attribution of undesirable outcomes to having “poor”
self-regulation) and instead take a strength-based
perspective. Each individual carries a unique set
of self-regulatory strengths. By understanding
how to maximize these strengths and the fit
between these strengths and an individual’s contextual resources, the continued study of self-regulation will help researchers promote thriving and
positive outcomes across the life course.
Self-Regulation
6.2
Examining Developmental
Changes in Self-Regulation
Over Time
In addition to issues with conceptualization, it is
also not clear if constructs, as operationalized
across disciplines, are all measuring the same
underlying skills. In addition, longitudinal measurement of the developmental course (both
behavioral and neurological) of the underlying
components of self-regulation over different transitions and turning points is lacking at present.
Although a number of recent investigations provide insight into the structure of self-regulation in
young children (i.e., unitary vs. componential),
very little of this work has involved repeated
assessments over time. As a result, we know a
great deal about the performance of children
before and early in preschool (e.g., Carlson 2005)
but much less about self-regulation as children
move through formal schooling. It is also important to examine whether and how these changing
abilities relate to behavior in real-world contexts.
Indeed, it could be the case that children who
come into school with stronger self-regulation
skills—as assessed from using tasks derived from
cognitive neuroscience—also exhibit stronger
self-regulation on classroom-based measures
(Rimm-Kaufman et al. 2009). It is also possible
that the relations between these sets of skills are
more limited than anticipated and that these different types of tasks tap into different abilities
altogether. Finally, the malleability of selfregulation—and its components, such as working
memory, inhibitory control, and attention control, and particularly the impact of different intervention efforts on these abilities—has not been
extensively charted. We turn to this next.
6.3
Improving Intervention
Efforts
As the research reviewed suggests, there has been
a sharp increase in the number of applied investigations targeting self-regulation, including a
plethora of new programs for young children
(Bierman et al. 2008a; Diamond and Lee 2011;
291
Jones et al. 2011; Raver et al. 2011; Schmitt et al.
2015; Tominey and McClelland 2011). Along
with these changes, there has been an increase in
interdisciplinary collaborations. These collaborations have led to new developments in measurements, analyses, and interventions related to
understanding and promoting self-regulation
skills early in the life course as a way to optimize
development and prevent future difficulties.
Moreover, researchers have started to examine
the complex and dynamic relations among selfregulation and important variables that together
influence individual health and well-being across
the life course (McClelland et al. 2010).
Although research has documented the stability of self-regulation trajectories over time, the
malleability of these skills is also evident. Thus,
although more research is needed to examine the
key components of effective interventions to promote self-regulation and the long-term effects of
such interventions, a few recommendations can
be made. First, in accordance with the principles
of LCHD, self-regulation interventions are likely
most effective when administered to individuals
at turning points or sensitive periods of development, such as the early childhood years (Blair
and Raver 2012). In addition, interventions are
most effective when they integrate multiple levels of influence across different contexts (e.g.,
Jones and Bouffard 2012) and involve repeated
practice of skills that are relevant to behavior in
everyday settings and which increase in complexity over time (e.g., Diamond and Lee 2011).
There is also support for interventions to be most
effective for groups of children who are at the
most risk, such as those living in poverty and/or
experiencing toxic stress and ACEs (Blair and
Raver 2014; Schmitt et al. 2015). Finally, recent
work has examined the impact of additional
intervention components, such as mindfulness
practices and yoga, on children’s self-regulation,
with some encouraging results (Diamond and
Lee 2011; Zelazo and Lyons 2012).
It is also clear that more needs to be done to
translate research and interventions into practice. From a public health perspective, clinicians
and pediatricians need better tools for assessing
children’s self-regulation especially in the early
292
childhood years. Based on the importance of
developing strong self-regulation, it seems plausible that well-child visits include screening of
self-regulation starting when children are 3 years
of age. There are some measures available that
assess aspects of self-regulation such as the EDI
(Janus and Offord 2007), but more work is
needed in this area. In the research realm, some
progress has been made in developing ecologically valid and sensitive measures of selfregulation and in recognizing the roles of context
in the development of these skills (e.g.,
McClelland and Cameron 2012). As noted
above, however, it is unclear if self-regulation
measured in one context relates to self-regulation in another context and how these relations
change over time.
Finally, it is critical that the results of basic and
applied research get translated into policy. Some
efforts are ongoing to bridge the science of selfregulation and child development with policy and
between a diverse number of fields (see, e.g.,
Halfon 2012; Halfon and Inkelas 2003; Shonkoff
2011; Shonkoff and Bales 2011; Shonkoff et al.
2012). Thus, there is great momentum in this
arena. Although more work remains, there is an
increasing energy around translating the importance of self-regulation for important health and
developmental outcomes into policy and practice.
Framing our understanding of self-regulation
within the principles of LCHD and the RDS perspective is a promising way to improve research
and translational efforts and promote healthy
development across the life span.
References
Anderson, E. S., Winett, R. A., Wojcik, J. R., Winett,
S. G., & Bowden, T. (2001). A computerized social
cognitive intervention for nutrition behavior: Direct
and mediated effects on fat, fiber, fruits, and vegetables, self-efficacy, and outcome expectations among
food shoppers. Annals of Behavioral Medicine, 23(2),
88–100.
Backen Jones, L., Rothbart, M. K., & Posner, M. I. (2003).
Development of executive attention in preschool children. Developmental Science, 6(5), 498–504.
Bagozzi, R. P., & Edwards, E. A. (1998). Goal setting
and goal pursuit in the regulation of body weight.
Psychology and Health, 13(4), 593–621.
M. McClelland et al.
Baker, C. E., Cameron, C. E., Rimm-Kaufman, S. E.,
& Grissmer, D. W. (2012). Family and sociodemographic predictors of school readiness among African
American boys in kindergarten. Early Education and
Development, 23, 833–854.
Bandura, A. (1969). Principles of behavior modification.
New York, NY: Holt, Rinehart & Winston.
Bargh, J. A., Gollwitzer, P. M., Lee-Chai, A., Barndollar,
K., & Trötschel, R. (2001). The automated will:
Nonconscious activation and pursuit of behavioral
goals. Journal of Personality and Social Psychology,
81(6), 1014–1027.
Barkley, R. A. (1997). Behavioral inhibition, sustained
attention, and executive functions: Constructing a unifying theory of ADHD. Psychological Bulletin, 121,
65–94.
Barkley, R. A. (2011). Attention-deficit/hyperactivity
disorder, self-regulation, and executive functioning.
In K. D. V. R. F. Baumeister (Ed.), Handbook of selfregulation: Research, theory, and applications (2nd
ed., pp. 551–563). New York, NY: Guilford Press.
Barnett, W. S., Jung, K., Yarosz, D. J., Thomas, J.,
Hornbeck, A., Stechuk, R., & Burns, S. (2008).
Educational effects of the tools of the mind curriculum: A randomized trial. Early Childhood
Research Quarterly, 23(3), 299–313. doi:10.1016/j.
ecresq.2008.03.001.
Baumeister, R. F., & Vonasch, A. J. (2014). Uses of selfregulation to facilitate and restrain addictive behavior. Addictive Behaviors, 44, 3–8. doi:10.1016/j.
addbeh.2014.09.011.
Bernier, A., Carlson, S. M., & Whipple, N. (2010).
From external regulation to self-regulation: Early
parenting precursors of young children’s executive
functioning. Child Development, 81(1), 326–339.
doi:10.1111/j.1467-8624.2009.01397.x.
Berwid, O. G., Curko Kera, E. A., Marks, D. J., Santra,
A., Bender, H. A., & Halperin, J. M. (2005). Sustained
attention and response inhibition in young children
at risk for attention deficit/hyperactivity disorder.
Journal of Child Psychology & Psychiatry, 46(11),
1219–1229.
Bierman, K. L., Domitrovich, C. E., Nix, R. L., Gest,
S. D., Welsh, J. A., Greenberg, M. T., Blair, C.,
Nelson, K. E., & Gill, S. (2008a). Promoting academic and social-emotional school readiness: The
Head Start REDI program. Child Development, 79(6),
1802–1817.
Bierman, K. L., Nix, R. L., Greenberg, M. T., Blair, C.,
& Domitrovich, C. E. (2008b). Executive functions
and school readiness intervention: Impact, moderation, and mediation in the Head Start REDI program.
Development and Psychopathology, 20(03), 821–843.
doi:10.1017/S0954579408000394.
Blair, C. (2010). Stress and the development of selfregulation in context. Child Development Perspectives,
4(3), 181–188. doi:10.1111/j.1750-8606.2010.00145.x.
Blair, C., & Raver, C. C. (2012). Child development in the
context of adversity: Experiential canalization of brain
and behavior. American Psychologist, 67(4), 309–318.
doi:10.1037/a0027493.
Self-Regulation
Blair, C., & Raver, C. C. (2014). Closing the achievement gap through modification of neurocognitive and
neuroendocrine function: Results from a cluster randomized controlled trial of an innovative approach to
the education of children in kindergarten. PLoS One,
9(11), e112393. doi:10.1371/journal.pone.0112393.
Blair, C., & Razza, R. P. (2007). Relating effortful
control, executive function, and false belief understanding to emerging math and literacy ability in
kindergarten. Child Development, 78(2), 647–663.
doi:10.1111/j.1467-8624.2007.01019.x.
Blanchard, C. M., Courneya, K. S., Rodgers, W. M.,
Daub, B., & Knapik, G. (2002). Determinants of exercise intention and behavior during and after phase 2
cardiac rehabilitation: An application of the theory of
planned behavior. Rehabilitation Psychology, 47(3),
308–323.
Bowen, W. G., Chingos, M. M., & McPherson, M. S.
(2009). Test scores and high school grades as predictors. Crossing the finish line: Completing college at America’s public universities (pp. 112–133).
Princeton, NJ: Princeton University Press.
Bowers, E. P., Gestsdottir, S., Geldhof, G., Nikitin, J., von
Eye, A., & Lerner, R. M. (2011). Developmental trajectories of intentional self-regulation in adolescence:
The role of parenting and implications for positive and
problematic outcomes among diverse youth. Journal
of Adolescence, 34(6), 1193–1206.
Brandstädter, J. (2006). Action perspectives on human
development. In W. Damon (Series Ed.), & R. Lerner
(Vol. Ed.), Handbook of child psychology (Theoretical
models of human development). (Vol. 1, pp. 516–568).
Hoboken, NJ: Wiley.
Bridges, L. J., Denham, S. A., & Ganiban, J. M. (2004).
Definitional issues in emotion regulation research.
Child Development, 75(2), 340–345.
Brody, G. H., & Ge, X. (2001). Linking parenting processes and self-regulation to psychological functioning and alcohol use during early adolescence.
Journal of Family Psychology, 15(1), 82–94.
doi:10.1037/0893-3200.15.1.82.
Buckner, J. C., Mezzacappa, E., & Beardslee, W. R.
(2009). Self-regulation and its relations to adaptive
functioning in low income youths. American Journal
of Orthopsychiatry, 79(1), 19–30. doi:10.1037/
a0014796.
Burrage, M. J., Ponitz, C. C., McCready, E. A., Shah, P., Sims,
B. C., Jewkes, A. M., & Morrison, F. J. (2008). Age- and
schooling-related effects on executive functions in young
children: A natural experiment. Child Neuropsychology,
14(6), 510–524. doi:10.1080/09297040701756917.
Calfas, K. J., Sallis, J. F., Zabinski, M. F., Wilfley, D. E.,
Rupp, J., Prochaska, J. J., et al. (2002). Preliminary
evaluation of a multicomponent program for nutrition
and physical activity change in primary care: PACE
for adults. Preventive Medicine, 34, 153–161.
Cameron, C. E., & Morrison, F. J. (2011). Teacher activity orienting predicts preschoolers’ academic and selfregulatory skills. Early Education & Development,
22(4), 620–648.
293
Cameron Ponitz, C. E., Rimm-Kaufman, S. E., Grimm,
K. J., & Curby, T. W. (2009). Kindergarten classroom
quality, behavioral engagement, and reading achievement. School Psychology Review, 38(1), 102–120.
Carlson, S. M. (2005). Developmentally sensitive measures of executive function in preschool children.
Developmental Neuropsychology, 28(2), 595–616.
doi:10.1207/s15326942dn2802_3.
Carlson, S. M., Zelazo, P. D., & Faja, S. (2013). Executive
function. In P. D. Zelazo (Ed.), The Oxford handbook
of developmental psychology, Body and mind (Vol. 1,
pp. 706–743). New York, NY: Oxford University Press.
Center on the Developing Child. (2011). Building the
brain’s “Air Traffic Control” system: How early experiences shape the development of executive function.
(Working Paper No. 11). Retrieved from http://www.
developingchild.harvard.edu.
Chang, F., Kidd, J. R., Livak, K. J., Pakstis, A. J., &
Kidd, K. K. (1996). The world-wide distribution of
allele frequencies at the human dopamine D4 receptor
locus. Human Genetics, 98(1), 91–101. doi:10.1007/
s004390050166.
Christakis, D. A., Zimmerman, F. J., DiGiuseppe, D. L.,
& McCarty, C. A. (2004). Early television exposure
and subsequent attentional problems in children.
Pediatrics, 113(4), 708–713.
Colombo, J. (2001). The development of visual attention in
infancy. Annual Review of Psychology, 52, 337–367.
Committee on Psychosocial Aspects of Child and Family
Health, Committee on Early Childhood, Adoption,
Dependent Care, Section on Developmental Behavioral
Pediatrics, Garner, A. S., Shonkoff, J. P., Siegel, B. S.,
et al. (2012). Early childhood adversity, toxic stress,
and the role of the pediatrician: Translating developmental science into lifelong health. Pediatrics, 129(1),
e224–e231. doi:10.1542/peds.2011-2662.
Connor, C., Ponitz, C., Phillips, B. M., Travis, Q., Glasney,
S., & Morrison, F. J. (2010). First graders’ literacy and
self-regulation gains: The effect of individualizing student instruction. Journal of School Psychology, 48(5),
433–455.
Diamond, A. (2002). Normal development of prefrontal cortex from birth to young adulthood: Cognitive
functions, anatomy, and biochemistry. In D. T. Stuss
& R. T. Knight (Eds.), Principles of frontal lobe
function (pp. 466–503). London, England: Oxford
University Press.
Diamond, A., Barnett, W. S., Thomas, J., & Munro,
S. (2007). Preschool program improves cognitive
control. Science, 318, 1387–1388. doi:10.1126/
science.1151148.
Diamond, A., & Lee, K. (2011). Interventions shown to
aid executive function development in children 4 to
12 years old. Science, 333, 959–964. doi:10.1126/
science.1204529.
Domitrovich, C. E., Cortes, R. C., & Greenberg, M. T.
(2007). Improving young children’s social and emotional competence: A randomized trial of the preschool
‘PATHS’ curriculum. Journal of Primary Prevention,
28(2), 67–91.
294
Duckworth, A. L., & Allred, K. M. (2012). Temperament
in the classroom. In R. L. Shiner & M. Zentner (Eds.),
Handbook of temperament (pp. 627–644). New York,
NY: Guilford Press.
Duckworth, A. L., & Seligman, M. E. P. (2005). Selfdiscipline outdoes IQ in predicting academic performance of adolescents. Psychological Science, 16(12),
939–944.
Duckworth, A. L., Tsukayama, E., & Geier, A. B. (2010a).
Self-controlled children stay leaner in the transition to
adolescence. Appetite, 54(2), 304–308.
Duckworth, A. L., Tsukayama, E., & May, H. (2010b).
Establishing causality using longitudinal hierarchical linear modeling: An illustration predicting achievement from self-control. Social
Psychology and Personality Science, 1(4), 311–317.
doi:10.1177/1948550609359707.
Duckworth, A. L., Quinn, P. D., & Tsukayama, E. (2012).
What no child left behind leaves behind: A comparison of standardized achievement test score and report
card grades. Journal of Educational Psychology,
104(2), 439–451. doi:10.1037/a0026280.
Duncan, G. J., Dowsett, C. J., Claessens, A.,
Magnuson, K., Huston, A. C., Klebanov, P., et al.
(2007). School readiness and later achievement.
Developmental. Psychology, 43(6), 1428–1446.
doi:10.1037/0012-1649.43.6.1428.
DuPaul, G. J., & Kern, L. (2011). Promotion of academic
skills Young children with ADHD: Early identification and intervention (pp. 107–125). Washington, DC:
American Psychological Association.
DuPaul, G. J., Kern, L., Gormley, M. J., & Volpe, R. J.
(2011). Early intervention for young children with
ADHD: Academic outcomes for responders to behavioral treatment. School Mental Health, 3(3), 117–126.
doi:10.1007/s12310-011-9053-x.
Eisenberg, N., Smith, C. L., Sadovsky, A., & Spinrad,
T. L. (2004). Effortful control: Relations with emotion
regulation, adjustment, and socialization in childhood.
In R. F. Baumeister & K. D. Vohs (Eds.), Handbook
of self-regulation: Research, theory, and applications
(pp. 259–282). New York, NY: Guilford.
Eisenberg, N., Duckworth, A. L., Spinrad, T. L., &
Valiente, C. (2014). Conscientiousness: Origins
in childhood? Developmental Psychology, 50(5),
1331–1349. http://dx.doi.org/10.1037/a0030977.
Evans, G. W., Fuller-Rowell, T. E., & Doan, S. N. (2012).
Childhood cumulative risk and obesity: The mediating role of self-regulatory ability. Pediatrics, 129(1),
e68–e73. doi:10.1542/peds.2010-3647.
Farran, D., Wilson, S. J., & Lipsey, M. (2013). Effects
of a curricular attempt to improve self- regulation
and achievement in prekindergarten children. Paper
presented at the biennial meeting for the Society for
Research in Child Development in Seattle, WA.
Fortsmeier, S., Drobetz, R., & Maercker, A. (2011). The
delay of gratification test for adults: Validating a behavioral measure of self-motivation in a sample of older
people. Motivation and Emotion, 35(2), 118–134.
Francis, L. A., & Susman, E. J. (2009). Self-regulation and
rapid weight gain in children from age 3 to 12 years.
M. McClelland et al.
Archives of Pediatrics & Adolescent Medicine, 163(4),
297–302. doi:10.1001/archpediatrics.2008.579.
Fuller, B., Bein, E., Bridges, M., Halfon, N., Jung, S.,
Rabe-Hesketh, S., et al. (2010). Maternal practices
that influence Hispanic infants’ health and cognitive
growth. Pediatrics, 125(2), e324–e332. doi:10.1542/
peds.2009-0496.
Garstein, M. R., & Rothbart, M. K. (2003). Studying
infant temperament via the revised infant behavior
questionnaire. Infant Behavior and Development,
26(1), 64–86.
Gathercole, S. E., Pickering, S. J., Ambridge, B., &
Wearing, H. (2004). The structure of working memory
from 4 to 15 years of age. Developmental Psychology,
40(2), 177–190. doi:10.1037/0012-1649.40.2.177.
Geldhof, G. J., Little, T. D., & Colombo, J. (2010). Selfregulation across the life span. In The handbook of
life-span development, Social and emotional development (Vol. 2, pp. 116–157). Hoboken, NJ: Wiley.
Gershoff, E. T., Aber, J. L., Raver, C. C., & Lennon, M. C.
(2007). Income is not enough: Incorporating maternal hardship into models of income associations with
parenting and child development. Child Development,
78(1), 70–95.
Gestsdottir, S., & Lerner, R. M. (2008). Positive development in adolescence: The development and role of
intentional self-regulation. Human Development, 51,
202–224.
Gestsdottir, S., Lewin-Bizan, S., von Eye, A., Lerner,
J. V., & Lerner, R. M. (2009). The structure and function of selection, optimization, and compensation in
adolescence: Theoretical and applied implications.
Journal of Applied Developmental Psychology, 30(5),
585–600.
Gollwitzer, P. M. (1990). Action phases and mind-sets. In
E. T. Higgins & R. M. Sorrentino (Eds.), Handbook
of motivation and cognition: Foundations of social
behavior (Vol. 2, pp. 53–92). New York, NY: Guilford
Press.
Gollwitzer, P. M. (1996). The volitional benefits of planning. In P. M. Gollwitzer & J. A. Bargh (Eds.), The
psychology of action (pp. 287–312). New York, NY:
Guilford.
Gollwitzer, P. M. (1999). Implementation intentions:
Strong effects of simple plans. American Psychologist,
54(7), 493–503.
Grolnick, W. S., Kurowski, C. O., Dunlap, K. G., &
Hevey, C. (2000). Parental resources and the transition
to junior high. Journal of Research on Adolescence,
10(4), 465–488. doi:10.1207/SJRA1004_05.
Halfon, N. (2012). Addressing health inequalities in the
US: A life course health development approach. Social
Science & Medicine, 74(5), 671–673. doi:10.1016/j.
socscimed.2011.12.016.
Halfon, N., & Forrest, C. B. (2017). The emerging theoretical framework of life course health development. In
N. Halfon, C. B. Forrest, R. M. Lerner, & E. Faustman
(Eds.), Handbook of life course health-development
science. Cham: Springer.
Halfon, N., & Inkelas, M. (2003). Optimizing the health
and development of children. JAMA: The Journal of
Self-Regulation
the American Medical Association, 290(23), 3136–
3138. doi:10.1001/jama.290.23.3136.
Hofmann, W., Adriaanse, M., Vohs, K. D., & Baumeister,
R. F. (2014). Dieting and the self-control of eating
in everyday environments: An experience sampling
study. British Journal of Health Psychology, 19(3),
523–539. doi:10.1111/bjhp.12053.
Hooper, C. J., Luciana, M., Conklin, H. M., & Yarger,
R. S. (2004). Adolescents’ performance on the Iowa
gambling task: Implications for the development of
decision making and ventromedial prefrontal cortex.
Developmental Psychology, 40(6), 1148–1158.
Hsu, C., Soong, W., Stigler, J. W., Hong, C., & Liange, C.
(1981). The temperamental characteristics of Chinese
babies. Child Development, 52(4), 1337–1340.
doi:10.2307/1129528.
Hymel, S., LeMare, L., & McKee, W. (2011). The
early development Instrument: An examination
of convergent and discriminant validity. Social
Indicators Research, 103(2), 267–282. doi:10.1007/
s11205-011-9845-2.
Jackson, C., Lawton, R., Knapp, P., Raynor, D. K.,
Conner, M., Lowe, C., & Closs, S. J. (2005). Beyond
intention: Do specific plans increase health behaviours in patients in primary care? A study of fruit and
vegetable consumption. Social Science & Medicine,
60(10), 2383–2391.
James, W. (1890). The principles of psychology.
New York, NY: Henry Holt and Company.
James, W. (1899). Talks to teachers on psychology and to
students on some of life’s ideals. New York, NY: Holt
and Company.
Janus, M., & Offord, D. R. (2007). Development and
psychometric properties of the early development
Instrument (EDI): A measure of children’s school
readiness. Canadian Journal of Behavioural Science/
Revue canadienne des sciences du comportement,
39(1), 1–22. doi:10.1037/cjbs2007001.
Jones, S. M., & Bouffard, S. M. (2012). Social and emotional learning in schools: From programs to strategies. Society for Research in Child Development
Social Policy Report, 26, 1–33. http://files.eric.ed.gov/
fulltext/ED540203.pdf.
Jones, S. M., Brown, J. L., & Aber, J. L. (2011). Two-year
impacts of a universal school-based social- emotional
and literacy intervention: An experiment in translational
developmental research. Child. Development, 82(2),
533–554. doi:10.1111/j.1467-8624.2010.01560.x.
Kail, R. V. (2003). Information processing and memory.
In M. H. Bornstein, L. Davidson, C. L. M. Keyes, &
K. A. Moore (Eds.), Crosscurrents in contemporary
psychology (pp. 269–279). Mahwah, NJ: Lawrence
Erlbaum.
Kishiyama, M. M., Boyce, W. T., Jimenez, A. M., Perry,
L. M., & Knight, R. T. (2009). Socioeconomic disparities affect prefrontal function in children. Journal of
Cognitive Neuroscience, 21(6), 1106–1115.
Kopp, C. (1982). Antecedents of self-regulation: A developmental perspective. Developmental Psychology,
18(2), 199–214.
295
Larson, R. W. (2011). Adolescents’ conscious processes
of developing regulation: Learning to appraise challenges. New Directions for Child and Adolescent
Development, 2011(133), 87–97.
Lengua, L. J., Honorado, E., & Bush, N. R. (2007).
Contextual risk and parenting as predictors of effortful
control and social competence in preschool children.
Journal of Applied Developmental Psychology, 28(1),
40–55.
Lerner, R. M. (1982). Children and adolescents as producers of their own development. Developmental Review,
2(4), 342–370.
Lerner, R. M. (1985). Adolescent maturational changes
and psychosocial development: A dynamic interactional perspective. Journal of Youth and Adolescence,
14(4), 355–372.
Lerner, R. M., & Steinberg, L. D. (2009). The scientific
study of adolescent development: Historical and contemporary perspectives. In R. M. Lernerog & L. D.
Steinberg (Eds.), Handbook of adolescent psychology
(pp. 3–15). Hoboken, N.J: Wiley.
Lerner, R. M., Lerner, J. V., von Eye, A., Bowers, E. P.,
& Lewin-Bizan, S. (2011). Individual and contextual
bases of thriving in adolescence: A view of the issues.
Journal of Adolescence, 34(6), 1107–1114.
Levack, W. M. M., Taylor, K., Siegert, R. J., Dean, S. G.,
McPherson, K. M., & Weatherall, M. (2006). Is goal
planning in rehabilitation effective? A systematic
review. Clinical Rehabilitation, 20(9), 739–755.
Lewin-Bizan, S., Bowers, E. P., & Lerner, R. M. (2010).
One good thing leads to another: Cascades of positive youth development among American adolescents.
Development and Psychopathology, 22(4), 759–770.
doi:10.1017/S0954579410000441.
Luszczynska, A. (2006). An implementation intentions
intervention, the use of a planning strategy, and physical activity after myocardial infarction. Social Science
& Medicine, 62(4), 900–908.
Massey, E. K., Gebhardt, W. A., & Garnefski, N. (2008).
Adolescent goal content and pursuit: A review of
the literature from the past 16 years. Developmental
Review, 28(4), 421–460.
McClelland, M. M., Acock, A. C., & Morrison, F. J.
(2006). The impact of kindergarten learning-related
skills on academic trajectories at the end of elementary school. Early Child Research Quarterly, 21(4),
471–490.
McClelland, M. M., & Cameron, C. E. (2012). Selfregulation in early childhood: Improving conceptual
clarity and developing ecologically-valid measures.
Child Development Perspectives, 6(2), 136–142.
doi:10.1111/j.1750–8606.2011.00191.x.
McClelland, M. M., Cameron, C. E., Connor, C. M.,
Farris, C. L., Jewkes, A. M., & Morrison, F. J.
(2007). Links between behavioral regulation and
preschoolers’ literacy, vocabulary and math skills.
Developmental
Psychology,
43(4),
947–959.
doi:10.1037/0012-1649.43.4.947.
McClelland, M. M., Cameron Ponitz, C. C., Messersmith,
E., & Tominey, S. (2010). Self-regulation: The integration
296
of cognition and emotion. In R. Lerner (Series Ed.) &
W. Overton (Vol. Ed.), Handbook of life-span development (Cognition, biology and methods) (Vol. 1,
pp. 509–553). Hoboken, NJ: Wiley.
McClelland, M. M., Acock, A. C., Piccinin, A., Rhea,
S. A., & Stallings, M. C. (2013). Relations between
preschool attention span-persistence and age 25 educational outcomes. Early Childhood Research Quarterly,
28, 314–324. doi:10.1016/j.ecresq.2012.07.008.
McClelland, M. M., Cameron, C. E., Duncan, R., Bowles,
R. P., Acock, A. C., Miao, A., & Pratt, M. E. (2014).
Predictors of early growth in academic achievement:
The Head-toes-knees-shoulders task. Frontiers in
Psychology, 5, 1–14. doi:10.3389/fpsyg.2014.00599.
Metcalfe, J., & Mischel, W. (1999). A hot/cool-system
analysis of delay of gratification: Dynamics of willpower. Psychological Review, 106(1), 3–19.
Miller, D. C., & Byrnes, J. P. (2001). Adolescents’ decision
making in social situations. A self-regulation perspective. Journal of Applied Developmental Psychology,
22(3), 237–256.
Mischel, W. (1968). Personality and assessment. Hoboken,
NJ: Wiley.
Mischel, W., & Ayduk, O. (2004). Willpower in a
cognitive-affective processing system: The dynamics
of delay of gratification. In R. F. Baumeister & K. D.
Vohs (Eds.), Handbook of self-regulation: Research,
theory, and applications (pp. 99–129). New York, NY:
Guilford.
Mischel, W., & Ebbesen, E. B. (1970). Attention in delay
of gratification. Journal of Personality and Social
Psychology, 16(2), 329–337.
Mischel, W., Ebbesen, E. B., & Zeiss, A. R. (1972).
Cognitive and attentional mechanisms in delay of
gratification. Journal of Personality and Social
Psychology, 21(2), 204–218.
Mistry, R. S., Benner, A. D., Biesanz, J. C., Clark,
S. L., & Howes, C. (2010). Family and social risk,
and parental investments during the early childhood years as predictors of low-income children’s
school readiness outcomes. Early Childhood
Research Quarterly, 25(4), 432–449. doi:10.1016/j.
ecresq.2010.01.002.
Moffitt, T. E., Arseneault, L., Belsky, D., Dickson, N.,
Hancox, R. J., Harrington, H., et al. (2011). A gradient
of childhood self-control predicts health, wealth, and
public safety. Proceedings of the National Academy
of Sciences, 108(7), 2693–2698. doi:10.1073/
pnas.1010076108.
Moilanen, K. L. (2007). The adolescent self-regulatory
inventory: The development and validation of a
questionnaire of short-term and long-term self-regulation. Journal of Youth and Adolescence, 36(6),
835–848.
Moilanen, K. L., Shaw, D. S., & Fitzpatrick, A. (2010).
Self-regulation in early adolescence: Relations with
mother-son relationship quality and maternal regulatory support and antagonism. Journal of Youth
and Adolescence, 39(11), 1357–1367. doi:10.1007/
s10964-009-9485-x.
M. McClelland et al.
Molina, B. S. G., Hinshaw, S. P., Swanson, J. M., Arnold,
L. E., Vitiello, B., Jensen, P. S., Epstein, J. N., Hoza,
B., Hechtman, L., Abikoff, H. B., Elliott, G. R.,
Greenhill, L. L., Newcorn, J. H., Wells, K.C., Wigal,
T., Gibbons, R. D., Hur, K., Houck, P. R., Houck, P. R.
(2009). The MTA at 8 years: Prospective follow-up
of children treated for combined-type ADHD in a
multisite study. Journal of the American Academy of
Child & Adolescent Psychiatry, 48(5), 484–500. doi:
10.1097/CHI.0b013e31819c23d0.
Moore, B., Mischel, W., & Zeiss, A. (1976). Comparative
effects of the reward stimulus and its cognitive representation in voluntary delay. Journal of Personality
and Social Psychology, 34(3), 419–424.
Morrison, F. J., Ponitz, C. C., & McClelland, M. M.
(2010). Self-regulation and academic achievement in
the transition to school. In S. D. Calkins & M. Bell
(Eds.), Child development at the intersection of emotion and cognition (pp. 203–224). Washington, DC:
American Psychological Association.
Nikkelen, S. W. C., Valkenburg, P. M., Huizinga, M.,
& Bushman, B. J. (2014). Media use and ADHDrelated behaviors in children and adolescents: A
meta-analysis. Developmental Psychology, 50(9),
2228–2241. doi:10.1037/a0037318.
Norman, D. A., & Shallice, T. (1986). Attention to
action: Willed and automatic control of behavior. In
R. J. Davidson, G. E. Schwartz, & D. Shapiro (Eds.),
Consciousness and self-regulation: Advances in
research and theory (Vol. 4, pp. 1–18). New York, NY:
Plenum Press.
Obradovic, J. (2010). Effortful control and adaptive functioning of homeless children: Variablefocused and person-focused analyses. Journal of
Applied Developmental Psychology, 31(2), 109–117.
doi:10.1016/j.appdev.2009.09.004.
Orbell, S., & Sheeran, P. (2000). Motivational and volitional processes in action initiation: A field study
of the role of implementation intentions. Journal of
Applied Social Psychology, 30(4), 780–797.
Orbell, S., Hodgkins, S., & Sheeran, P. (1997).
Implementation intentions and the theory of planned
behavior. Personality and Social Psychology Bulletin,
23(9), 945–954.
Overton, W. F. (2010). Life-span development: Concepts
and issues. In W. F. Overton (Ed). Cognition, biology, and methods across the lifespan. In R. M. Lerner
(Ed.), Handbook of life-span development (pp. 1–29).
Hoboken, NJ: Wiley.
Overton, W. F. (2013). Relationism and relational developmental systems: A paradigm for developmental
science in the post-Cartesian era. In R. M. Lerner
& J. B. Benson (Eds.), Advances in child development and behavior, Embodiment and epigenesis:
Theoretical and methodological issues in understanding the role of biology within the relational
developmental system (Vol. 44, pp. 21–64). San
Diego, CA: Academic Press.
Posner, M. I., & Rothbart, M. K. (1998). Attention,
self-regulation, and consciousness. Philosophical
Self-Regulation
Transactions of the Royal Society of London B,
353(1377), 1915–1927.
Posner, M. I., & Rothbart, M. K. (2000). Developing
mechanisms of self-regulation. Development and
Psychopathology, 12(3), 427–441.
Quinn, P. D., & Fromme, K. (2010). Self-regulation as
a protective factor against risky drinking and sexual
behavior. Psychology of Addictive Behaviors, 24(3),
376–385. doi:10.1037/A0018547.
Radesky, J. S., Kistin, C. J., Zuckerman, B., Nitzberg, K.,
Gross, J., Kaplan-Sanoff, M., et al. (2014). Patterns
of mobile device use by caregivers and children during meals in fast food restaurants. Pediatrics, 133(4),
e843–e849. doi:10.1542/peds.2013-3703.
Raver, C. C., Jones, S. M., Li-Grinning, C., Zhai, F., Bub, K.,
& Pressler, E. (2011). CSRP’s impact on low- income
preschoolers’ preacademic skills: Self-regulation as
a mediating mechanism. Child Development, 82(1),
362–378. doi:10.1111/j.1467-8624.2010.01561.x.
Reid, R., Trout, A. L., & Schartz, M. (2005). Selfregulation interventions for children with attention
deficit/hyperactivity disorder. Exceptional Children,
71, 361–377.
Rimm-Kaufman, S. E., Curby, T. W., Grimm, K. J.,
Nathanson, L., & Brock, L. L. (2009). The contribution of children’s self-regulation and classroom
quality to children’s adaptive behaviors in the kindergarten classroom. Developmental Psychology, 45(4),
958–972.
Robertson, L. A., McAnally, H. M., & Hancox, R. J.
(2013). Childhood and adolescent television viewing
and antisocial behavior in early adulthood. Pediatrics,
131(3), 439–446. doi:10.1542/peds.2012-1582.
Rothbart, M. K., & Bates, J. E. (1998). Temperament.
In W. Damon (Series Ed.) & N. Eisenberg (Ed.),
Handbook of child psychology (Social, emotional,
and personality development) (Vol. 3). Hoboken, NJ:
Wiley.
Rothbart, M. K., Ahadi, S. A., Hersey, K. L., & Fisher,
P. (2001). Investigations of temperament at three to
seven years: The Children’s behavior questionnaire.
Child Development, 72(5), 1394–1408.
Sabbagh, M. A., Xu, F., Carlson, S. M., Moses, L. J., &
Lee, K. (2006). The development of executive functioning and theory of mind: A comparison of Chinese
and U.S. preschoolers. Psychological Science, 17(1),
74–81.
Schmitt, S. A., McClelland, M. M., Tominey, S. L., &
Acock, A. C. (2015). Strengthening school readiness
for head start children: evaluation of a self-regulation
intervention. Early Childhood Research Quarterly
30(A),
20–31.doi:
http://dx.doi.org/10.1016/j.
ecresq.2014.08.001.
Scholz, U., Sniehotta, F. F., Burkert, S., & Schwarzer,
R. (2007). Increasing physical exercise levels:
Age-specific benefits of planning. Journal of Aging
and Health, 19(5), 851–866.
Sektnan, M., McClelland, M. M., Acock, A. C., &
Morrison, F. J. (2010). Relations between early family
risk, children’s behavioral regulation, and academic
297
achievement. Early Childhood Research Quarterly,
25(4), 464–479. doi:10.1016/j.ecresq.2010.02.005.
Sheeran, P., & Orbell, S. (2000). Using implementation
intentions to increase attendance for cervical cancer
screening. Health Psychology, 19(3), 283–289.
Sheese, B. E., Rothbart, M. K., Posner, M. I., White,
L. K., & Fraundorf, S. H. (2008). Executive attention and self-regulation in infancy. Infant Behavior &
Development, 31(1), 501–510.
Shonkoff, J. P. (2011). Protecting brains, not simply
stimulating minds. Science, 333(6045), 982–983.
doi:10.1126/science.1206014.
Shonkoff, J. P., & Bales, S. N. (2011). Science does not
speak for itself: Translating child development research
for the public and its policymakers. Child Development,
82(1), 17–32. doi:10.1111/j.1467-8624.2010.01538.x.
Shonkoff, J. P., Garner, A. S., The Committee on
Psychosocial Aspects of Child Family Health,
Committee on Early Childhood, Adoption, Dependent
Care, Section on Developmental Behavioral Pediatrics,
Siegel, B. S., et al. (2012). The lifelong effects of
early childhood adversity and toxic stress. Pediatrics,
129(1), e232–e246. doi:10.1542/peds.2011-2663.
Sniehotta, F. F., Schwarzer, R., Scholz, U., & Schüz,
B. (2005). Action planning and coping planning
for long-term lifestyle change: Theory and assessment. European Journal of Social Psychology, 35(4),
565–576.
Sniehotta, F. F., Scholz, U., & Schwarzer, R. (2006).
Action plans and coping plans for physical exercise: A longitudinal intervention study in cardiacrehabilitation. British Journal of Health Psychology,
11(1), 23–37.
Stansbury, K., & Zimmermann, L. K. (1999). Relations
among child language skills, maternal socializations
of emotion regulation, and child behavior problems.
Child Psychiatry & Human Development, 30(2),
121–142.
Steinberg, L. (2010). A behavioral scientist looks at the
science of adolescent brain development. Brain and
Cognition, 72(1), 160–164. doi:10.1016/j.bandc.2009.
11.003.
von Suchodoletz, A., Gestsdottir, S., Wanless, S. B.,
McClelland, M. M., Birgisdottir, F., Gunzenhauser, C.,
et al. (2013). Behavioral self-regulation and relations to
emergent academic skills among children in Germany
and Iceland. Early Childhood Research Quarterly,
28(1), 62–73. doi:10.1016/j.ecresq.2012.05.003.
Tominey, S. L., & McClelland, M. M. (2011). Red light,
purple light: Findings from a randomized trial using
circle time games to improve behavioral self-regulation
in preschool. Early Education & Development, 22(3),
489–519. doi:10.1080/10409289.2011.574258.
Tsukayama, E., Toomey, S. L., Faith, M., & Duckworth,
A. L. (2010). Self-control as a protective factor against
overweight status in the transition to adolescence.
Archives of Pediatrics and Adolescent Medicine,
164(7), 631–635.
Wachs, T. D., Gurkas, P., & Kontos, S. (2004). Predictors
of preschool children’s compliance behavior in early
298
childhood classroom settings. Journal of Applied
Developmental Psychology, 25(4), 439–457.
Wanless, S. B., McClelland, M. M., Tominey, S. L., &
Acock, A. C. (2011). The influence of demographic
risk factors on children’s behavioral regulation in
prekindergarten and kindergarten. Early Education &
Development, 22(3), 461–488.
Zelazo, P. D., & Lyons, K. E. (2012). The potential benefits of mindfulness training in early childhood: A
developmental social cognitive neuroscience perspective. Child Development Perspectives, 6(2), 154–160.
doi:10.1111/j.1750–8606.2012.00241.x.
Zelazo, P. D., Anderson, J. E., Richler, J., Wallner-Allen,
K., Beaumont, J. L., & Weintraub, S. (2013). II. NIH
toolbox cognition battery (CB): Measuring executive
function and attention. Monographs of the Society
for Research in Child Development, 78(4), 16–33.
doi:10.1111/mono.12032.
Ziegelmann, J. P., Lippke, S., & Schwarzer, R. (2006).
Adoption and maintenance of physical activity:
Planning interventions in young, middle-aged, and
older adults. Psychology and Health, 21(2), 145–163.
M. McClelland et al.
Ziegelmann, J. P., Luszczynska, A., Lippke, S., &
Schwarzer, R. (2007). Are goal intentions or implementation intentions better predictors of health
behavior? A longitudinal study in orthopedic
rehabilitation. Rehabilitation Psychology, 52(1),
97–102.
Ziegelmann, J. P., & Lippke, S. (2007). Planning and
strategy use in health behavior change: A life span
view. International Journal of Behavioral Medicine,
14(1), 30–39. doi:10.1007/BF02999225.
Zimmerman, B. J. (2002). Becoming a self-regulated
learner: An overview. Theory Into Practice, 41(2),
64–70.
Zimmerman, B. J., & Schunk, D. H. (2001). Self-regulated
learning and academic achievement: Theoretical perspectives (2nd ed.). Mahwah, NJ: Erlbaum.
Zimmermann, L. K., & Stansbury, K. (2003). The influence of temperamental reactivity and situational context on the emotion-regulatory abilities of 3-year-old
children. Journal of Genetic Psychology, 164(4),
389–409.
Open Access This chapter is licensed under the terms of the Creative Commons Attribution 4.0 International License
(http://creativecommons.org/licenses/by/4.0/), which permits use, sharing, adaptation, distribution and reproduction in
any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license and indicate if changes were made.
The images or other third party material in this chapter are included in the chapter’s Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the chapter’s Creative Commons license
and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
A Life Course Health Development
Perspective on Oral Health
James J. Crall and Christopher B. Forrest
1
Introduction
1.1
Life Course Health
Development Concepts
and Principles
Life Course Health Development(LCHD) is a
conceptual framework that helps to explain how
health develops over an individual’s lifetime and
emphasizes the need to treat health development
as a long-term investment, beginning early and
continuing throughout life (Halfon and Hochstein
2002). LCHD provides a powerful approach to
understanding diseases and conditions and how
risk factors, protective factors, critical life experiences, and environments affect long-term health
and disease outcomes. LCHD also can help
examine and explain how health and disease patterns, particularly health disparities, develop
across populations and over time (Halfon and
Hochstein 2002; Ben-Shlomo and Kuh 2002;
Keating and Hertzman 1999).
J.J. Crall (*)
Division of Public Health and Community Dentistry,
University of California Los Angeles (UCLA) School
of Dentistry, Los Angeles, CA, USA
e-mail: jcrall@dentistry.ucla.edu
C.B. Forrest
Applied Clinical Research Center, Children’s
Hospital of Pennsylvania, Philadelphia, PA, USA
Efforts to relate the life course perspective or
life course theory to the field of maternal and
child health (Fine and Kotelchuck, 2010) have
emphasized the following key concepts:
Pathways or Trajectories Health pathways or
trajectories are constructed and modified throughout the life span. While individual trajectories
vary, general patterns can be predicted for populations and communities based on social, economic,
and environmental exposures and experiences. A
life course does not reflect a series of discrete
steps or stages, but rather an integrated, dynamic,
and continuous set of exposures and experiences.
The set of possible trajectories that a person can
experience is constrained by evolutionary forces
and is highly determined by exposure to various
environmental contexts.
Importance of Early Life Exposures Early
experiences can markedly influence an individual’s future health development. Of particular
salience are exposures that occur prenatally (i.e.,
exposures in utero) and intergenerationally (i.e.,
factors related to the health of the mother prior to
conception). While adverse events and exposures
can have an impact at any point in a person’s life
course, the impact may be greatest at specific
critical or sensitive periods when developing biological systems are most readily modified (e.g.,
during fetal development, in early childhood,
during adolescence).
© The Author(s) 2018
N. Halfon et al. (eds.), Handbook of Life Course Health Development,
DOI 10.1007/978-3-319-47143-3_13
299
J.J. Crall and C.B. Forrest
300
Cumulative Impact Cumulative experiences
also can influence an individual’s future health
and development, even though any individual
experience may not impact health development.
For example, individuals may adapt with minimal impact to each episode of stress; however,
the cumulative impact of multiple stresses over
time (also referred to as “weathering” or “allostatic load”) may have a profound direct impact
on health development by altering biological
function and human behavior.
Risk and Protective Factors Throughout the
life span, protective factors improve health and
contribute to health development, while risk factors diminish health and make it more difficult to
reach full health potential. Moreover, pathways
are changeable, and risk and protective factors
are not limited to individual behavioral patterns
or receipt of health-care and social services, but
also include factors related to family, neighborhood, community, and social policy.
The LCHD theoretical framework (presented
more thoroughly in Chap. 2) incorporates and
expands these concepts into a robust explanatory
synthesis of how health in individuals and populations is produced and modified. In this chapter, we
use the LCHD principles to guide our review of
oral health development. The Table 1 illustrates
each LCHD principle’s relevance to oral health.
1.2
Previous Efforts to Apply Life
Course Concepts to Oral
Health
The multifactorial etiology of oral conditions,
their chronic nature and occurrence over the life
span, and inherent features of their expression
make oral health well suited to studies which
seek to apply life course concepts. For example,
Nicolau and colleagues (Nicolau et al. 2007)
have highlighted several features that make oral
conditions amenable to life course epidemiological studies, the first of which is their observability—i.e., once these conditions develop, they are
readily detectable during assessments as opposed
to conditions that resolve or go unnoted if study
participants fail to report them. A second feature
relates to their being cumulative conditions,
which allows for comparisons of the degree of
disease development among individuals so that,
rather than all who develop the disease being
enumerated together, distinctions can be made
among them with respect to the extent or severity
of their disease. Third, oral conditions can be
reliably measured and validly diagnosed without
sophisticated or costly technology. Fourth, these
conditions are moderately prevalent; thus, the
required sample size for cohort studies is manageable. And, finally, oral health conditions have
a public health importance that makes their study
justifiable on both ethical and economic grounds.
Despite growing interest in using life course
concepts as the basis for various oral healthrelated scientific studies, robust applications have
been rather limited. A PubMed search conducted
as part of the literature review for this chapter
using the terms “life course and oral health”
yielded over 500 citations. However, a substantial portion of the publications examined in this
and other literature searches were found to have
one or more of the following limitations: studies
focused on a single condition, used crosssectional data, or analyzed a relatively narrow
portion of the life span, studies were based on
relatively small sample sizes, and studies were
predicated on retrospective analyses that frequently relied on recall of rather distant events.
Furthermore, efforts to examine oral health
from the perspective of LCHD concepts and principles have not been pursued. Therefore, the
goals of this chapter are to (a) examine the concept of oral health and major oral diseases and
conditions from a LCHD perspective using the
key concepts and principles noted in the table, (b)
assess what is (and what is not) known about the
development of oral health and its impact on general health and well-being across the life course,
and (c) offer recommendations for future
research. In light of the evidence-based literature
shortcomings noted in the preceding paragraph,
assessments of what is known about applications
of life course research approaches to studies of
oral health are limited to research publications
consistent with the life course epidemiology
1
Principle
Health Development
Brief description
Health development integrates the concepts of health and developmental
processes into a unified whole. Health enables adaptation to the environment,
task execution, participation in desired activities, and attainment of thriving.
Development refers to the mechanisms by which health changes over time.
Health development, then, comprises trajectories of health assets and
integrated sets of health resources that emerge as a result of a person’s
continuous and relational interactions with their environments.
2
Unfolding
3
Complexity
Health development unfolds continuously over the lifespan, from conception
to death, and is shaped by prior experiences and environmental interactions.
For human health development, there are four major functional phases: (1)
generativity, preconception and prenatal period; (2) acquisition of capacity,
the early years; (3) maintenance of capacity, the middle years; and (4)
managing decline, the later years. Each is conceptually distinct but can be
overlapping.
Health development results from adaptive, multilevel, and reciprocal
interactions between individuals and their physical, natural, and social
environments. There is a dynamic interplay between individual and multiple
physical, biochemical, psychological, social, and cultural networks which
enable adaptation and individual robustness. Each level is both a part and a
whole, nested and hierarchically aligned to optimize health development.
Oral health illustrations
Oral health is not merely the absence of oral
diseases. Instead, oral health refers to the
capacity to adapt to ubiquitous environmental
and microbial challenges, maintaining the
robustness of teeth and other structures in the
mouth, and oral health development refers to the
pathways and trajectories of change of these
assets over the life course.
Prenatal conditions can predispose developing
teeth to elevated risk of early childhood caries,
which is in turn a risk factor for caries later in
life, which can lead to tooth loss and associated
negative impacts on oral and overall health.
Oral health is strongly influenced by nutritional
intake and changes in the microbiome. These
effects in turn impact health development of
individuals as well as their energy levels,
healing, behavior, and cognitive development.
A Life Course Health Development Perspective on Oral Health
Table 1 Life Course Health Development principles
(continued)
301
302
Table 1 (continued)
Principle
Timing
Brief description
Health development is sensitive to the timing and social structuring of
environmental exposures and experiences. Time-specific health development
pathways refer to biological embedding that occurs during sensitive or critical
periods, when developing systems are most alterable, and exogenous and
endogenous influences can result in different adaptive responses. Socially
structured pathways link experiences and exposures in ways that create
recursive and mutually reinforcing patterns of risk, protection, and promotion.
Health development phenotypes are systematically malleable and enabled and
constrained by evolution to enhance adaptability to diverse environments.
Heredity transmits evolutionary signals through genetic, epigenetic,
behavioral, and symbolic dimensions, which establish health phenotypes that
depending on environmental circumstances may or may not be selected and
optimized to produce desirable outcomes. At the microlevel, a range of
strategies introduce variable types and levels of plasticity to optimize
adaptability from the molecular to the behavioral level. At a macrolevel,
strategies organize health development phases and life stages into functional
entities.
5
Plasticity
6
Thriving
Optimal health development promotes survival, enhances well-being, and
protects against disease. Health development can also be perturbed to create
suboptimal states that are precursors to fully formed disease—so-called
endophenotypes that represent intermediate phase transitions toward a fully
manifest phenotypic expression of a disease or disorder.
7
Harmony
Health development results from the balanced interactions of molecular,
physiological, behavioral, cultural, and evolutionary processes. Genetic
modulations that occur in molecular time frames measured in nanoseconds are
linked to biochemical modulation measured in milliseconds, homeostatic
mechanisms measured in seconds to days, social norms that evolve over years
and decades, cultural processes that change from years to centuries, and
ecological processes that until recently took millennia. Harmonization of these
processes produces rhythms and variability that characterize health. Loss of
coordination of these processes results in less robustness of the human system
with negative consequences.
Oral health illustrations
Oral cancers result from the dynamic interaction
of genetic, physical, biochemical, psychological,
behavioral, social, and cultural factors. Risk for
these conditions is highest for individuals who
use alcohol, use tobacco, are exposed to human
papilloma virus or Epstein-Barr virus, or are
exposed to x-radiation before the age of 50 years
The human jaw developed over millennia among
our human ancestors who spent hours each day
chewing their food. The interaction between
chewing of tough food and bone development
may have been responsible for the lack of
malocclusion observed in the hominid fossil
record. Today a softer diet has contributed to
smaller jaws and smaller teeth in modern
humans, leading to the large numbers of
individuals with malocclusion and need for
orthodontic treatment.
Diabetes perturbs oral health development by
increasing risk for periodontal disease, which
when present causes pain, chewing difficulties,
and tooth loss. Dry mouth, often a symptom of
undetected diabetes, can cause sores, ulcers,
infections and tooth decay. Smoking makes
these problems worse. Conversely, reducing
diabetes risk and reducing or eliminating
smoking can promote oral health.
Analysis of the DNA from ancient humans has
demonstrated shifts in oral bacterial species
ecology following dietary pattern changes and
reductions in the diversity of the oral ecosystem,
a possible factor in the rise of dental caries and
periodontal disease in modern humans whose
oral health may be better matched to
circumstances in our evolutionary history than
to modern environments.
J.J. Crall and C.B. Forrest
4
A Life Course Health Development Perspective on Oral Health
standard and LCHD framework. Furthermore,
because of the diverse nature of the major oral
health-related conditions noted above, assessments are largely limited to “dental conditions”
(i.e., dental caries, periodontal diseases, and
tooth loss), with limited additional examples pertaining to craniofacial abnormalities, oral cancers, or temporomandibular joint dysfunction.
2
Conceptualizing Oral Health
and Overview of Major
Conditions
2.1
Conceptualizing Oral Health
Concepts and definitions of oral health have varied over time, often paralleling broader conceptualizations of health. Concepts of how oral health
changes over time also often parallel more general paradigms. For example, many recent concepts of oral health have been based on the
biopsychosocial model, which focuses on how
the integration of various aspects of biological,
psychological, and social domains influence individuals’ health in different contexts and over time
(Borrell-Carrio et al. 2004).
The US Surgeon General’s Report on Oral
Health (SGROH), issued in 2000, and ensuing
initiatives have emphasized two major themes
regarding the concept of oral health—i.e., oral
health means much more than healthy teeth, and
oral health is integral to general health (US
Department of Health and Human Services 2000).
Interestingly, the SGROH embraced the World
Health Organization (WHO) definition of oral
health as “a state of being free from mouth and
facial pain, oral and throat cancer, oral infection
and sores, periodontal (gum) disease, tooth decay,
tooth loss, and other diseases and disorders that
limit an individual’s capacity in biting, chewing,
smiling, speaking, and psychosocial wellbeing.”
This definition espoused by both the WHO and
SGROH is deficient in that it does not positively
define what oral health is, but rather presents the
concept as the absence of detrimental conditions
or disorders. Conflating oral health and oral disorders is contrary to the WHO definition of overall
303
health which states that health is more than just
the absence of disease and represents a state of
complete physical, mental, and social well-being.
Researchers working to develop measures related
to oral health and its impact on quality of life also
have concluded that health and disease are different domains of human experience, which vary
both over time and among individuals, and are
dependent on context (Locker and Slade 1994;
Gregory et al. 2005).
A recent development in the conceptualization
and definition of oral health has emerged from
the FDI World Dental Federation in late 2016.
According to the new FDI definition, “Oral health
is multifaceted and includes the ability to speak,
smile, smell, taste, touch, chew, swallow, and
convey a range of emotions through facial expressions with confidence and without pain, discomfort, and disease of the craniofacial complex. The
core elements of oral health in the FDI framework are as follows: disease and condition status
refers to a threshold of severity or a level of progression of disease, which also includes pain and
discomfort; physiological function refers to the
capacity to perform a set of actions that include,
but are not limited to, the ability to speak, smile,
chew, and swallow; and psychosocial function
refers to the relationship between oral health and
mental state that includes, but is not limited to,
the capacity to speak, smile, and interact in social
and work situations without feeling uncomfortable or embarrassed.” (Glick et al. 2016).
LCHD theoretical principles conceptualize
health as an emergent set of integrated assets that
enable adaptation and pursuit of meaning and
happiness, reflect broader notions of the impact
of various health determinants over time, and
consider health in a more positive context (see
Table 1). Applying LCHD concepts, individuals
with disease (e.g., diabetics or persons living
with AIDS) can also be considered healthy if
they have developed assets which enable adaptation to their environment, task execution, participation in desired activities, and an ability to
thrive; health and disease are distinct, albeit interrelated concepts.
LCHD builds and expands upon the basic
tenets of the biopsychosocial and other ecologi-
J.J. Crall and C.B. Forrest
304
cal models and establishes a conceptual foundation for a future era of health care that moves
beyond systems focused on management of acute
and chronic diseases and conditions to systems
whose focus is on optimizing lifelong health for
individuals and populations (Halfon et al. 2014).
Using LCHD concepts and principles to conceptualize and define oral health has the potential
to move practitioners, researchers, policy makers, and the public beyond their historical limited focus on the physical form and function of
structures in and surrounding the oral cavity and
the clinical consequences of oral/dental diseases
to a new paradigm—one which fully embraces
LCHD tenets and the development of more robust
LCHD-based policies and systems for optimizing
the oral health of individuals and populations.
2.2
Major Oral Health-Related
Diseases and Conditions
Clinical conditions that have major significance
with respect to oral health in terms of prevalence,
impact on health, development and well-being,
and economic considerations include dental
diseases and their consequences (dental caries,
periodontal diseases, tooth loss), craniofacial
developmental disorders (e.g., cleft lip, cleft palate, malocclusion), oral cancers (including pharyngeal and salivary gland cancers), and facial
pain (distinct from dental pain) generally associated with temporomandibular joint and muscular
disorders. Brief overviews of these conditions are
provided below.
Dental Conditions Despite significant progress
during the latter portion of the twentieth century,
dental caries (tooth decay) remains the most
common chronic disease of childhood and a
major cause of tooth loss in children and adults.
In the United States, approximately 25% of
2–5-year-olds and over 50% of 6–8-year-old
children experience dental caries in their primary
teeth, and over 50% of 12–15-year-olds and 67%
of 16–19-year-olds experience caries in their permanent teeth (Dye et al. 2011a). Although substantial declines in the average number of teeth
and tooth surfaces affected by caries have been
documented in recent decades, particularly in
developed countries, according to the WHO, the
prevalence of tooth decay in school-age children
globally ranges from 60% to 90% (WHO, The
World Oral Health Report 2008).
Significant disparities in childhood caries
experience also persist. On average, US children
from lower-income households and children of
color are three to five times more likely than their
white more affluent counterparts to experience
caries and exhibit more severe forms of tooth
decay (Dye et al. 2011a; Vargas et al. 1998). Of
interest from a LCHD perspective, many children
from high-risk population groups do not exhibit
severe forms of dental caries, presumably because
the balance between individual’s risk and protective factors (e.g., diet, toothbrushing habits, fluoride exposure, oral microflora or other unidentified
characteristics) is conducive to maintaining a
healthy dentition (Feathersone 2000).
Caries affects dentate individuals across the
life span (Fig. 1), with over 90% of US adults
with natural teeth experiencing dental caries in
their permanent teeth (Dye et al. 2011a, b).
According to the WHO (WHO, The World Oral
Health Report, 2008) nearly 100% of dentate
adults globally are affected by dental caries,
making it the most prevalent chronic condition of
people worldwide. From a life course perspective, early childhood caries can have a profound
deleterious lifelong effect on an individual’s dentition status, as ECC often is a precursor of caries
and its consequences in adults.
Periodontal disease mainly results from infections and inflammation of the gingiva (gums) and
bone that surround and support the teeth. In its
early stage, called gingivitis, the gums can
become swollen and red and may bleed. In its
more serious form, called periodontitis, the gums
can pull away from the teeth, bone can be lost,
and teeth may loosen or even fall out. Thirty percent of US adults 30+ years of age have moderate
periodontal disease, and 8.5% have severe periodontitis (Thornton-Evans 2013). Males, older
adults, Black and Hispanic adults, current smokers, and those with lower incomes and less education are more likely to have moderate or severe
A Life Course Health Development Perspective on Oral Health
305
94%
97%
96%
97%
50-64
65-74
75+
82%
67%
56%
50%
29%
23%
2-5
6-8
9-11
12-15
16-19
20-34
34-49
Fig. 1 Proportion of the US population with caries experience (percentages for 2–5- and 6–8-year-olds reflect the
percentage of individuals with caries experience in primary teeth; for ages 9 and above, percentages reflect the
percentage with caries experience in permanent teeth) by
age (years) (Data Source: National Health and Nutrition
Examination Survey, 2011–2012. National Center for
Health Statistics, CDC)
periodontal disease (Thornton-Evans 2013).
Globally, severe periodontal disease is found in
15–20% of adults aged 35–44 years and increases
in prevalence with advancing age (WHO, The
World Oral Health Report 2008).
Dental caries and periodontal disease are the
primary reasons for tooth loss (Dye et al. 2011b;
Thornton-Evans 2013) which in its more severe
forms (loss of multiple teeth) can have negative
psychological, social, nutritional, and physical
effects. A person’s quality of life is diminished as
a result of tooth loss due to reductions in their
ability to chew and speak and reduced social
interactions and self-esteem (US Department of
Health and Human Services 2000; Hollister &
Weintraub 1993; Brennan et al. 2008). National
survey data indicate that approximately 50% of
US adults have lost at least one tooth.
Approximately 5% of US adults overall and 25%
of adults aged 65+ are completely edentulous
(have no natural teeth) (Dye et al. 2011b).
Worldwide, about 30% of people aged 65–74
have no natural teeth, with considerable disparities across countries (WHO, The World Oral
Health Report 2008).
Total US spending for dental services has
been relatively flat at approximately $111 billion
since 2010 (Wall 2013). The bulk of dental care
spending is related to diagnosis, prevention and
treatment of dental caries and periodontal disease, and treatment related to removal and
replacement of lost teeth.
Craniofacial Conditions Craniofacial defects
such as cleft lip and cleft palate (CLP) are among
the most common of all birth defects. The US incidence of cleft palate is 6 per 10,000 live births, and
the incidence for cleft lip with or without cleft palate is 11 per 10,000 live births (Center for Disease
Control and Prevention (CDC) 2014). CLP can
occur as an isolated condition or may be a component of an inherited disease or syndrome. Cleft lip
and cleft palate are thought to be caused by a combination of genes and other factors, such as things
the mother comes in contact with in her environment, what the mother eats or drinks, or certain
medications she uses during pregnancy. Maternal
smoking, alcohol use, steroid use, and anticonvulsants are associated with increased risk for cleft lip
and palate (Kohli and Kohli 2012). The incidence
of CLP also varies by race, with Asians and Native
Americans having higher rates and AfricanAmericans having lower rates. According to the
Centers for Disease Control and Prevention
J.J. Crall and C.B. Forrest
306
(CDC), in the United States, cleft lip and palate is
the third most common birth defect, and health
expenditures are approximately eight times higher
in the first 10 years of life for children with clefting than for those without (Boulet et al. 2009).
Malocclusion Misalignment or abnormal positioning of the teeth or an incorrect relation
between the teeth in the upper and lower jaws—is
neither a disease nor a life-threatening condition.
Nevertheless, growing numbers of people seek
and undergo orthodontic treatment, often because
of esthetic concerns and other quality of life
issues (Liu et al. 2009). Although children make
up the majority of orthodontic patients, adults
increasingly are seeking treatment for malocclusion and now comprise one-fifth of all orthodontic patients in the US. Orthodontic services
account for approximately one-eighth of US
dental care expenditures (Agency for Healthcare
Research and Quality 2015).
Oral
Cancers Approximately
45,000
Americans and over 450,000 people worldwide
are diagnosed each year with cancers that affect
the mouth and/or pharynx. Oral cancers comprise
85% of all head and neck cancers and have relatively high mortality rates (5-year survival = 63%)
(The Oral Cancer Foundation 2015; American
Cancer Society 2015). Oral cancer occurrence
rates are significantly higher for males than for
females (except in American Indians/Alaska
Natives) and higher for black males than for
white males up to age 70. Oral cancer rates
increase with age, increase more rapidly after age
50, and peak between ages 70 and 80 (Fig. 2)
(Ram et al. 2011). Extensive case-control and
longitudinal studies have implicated tobacco and
alcohol as major risk factors for oral cancer.
Human papillomavirus, syphilis, oro-dental factors, dietary deficiencies, chronic candidiasis,
and viruses also have been shown to be significantly associated with oral cancer (Ram et al.
2011). Findings of a recent study (Jacobson et al.
2012) suggest that oral cancers may be among
the most costly forms of cancer to treat in the
United States.
Facial Pain Associated with Temporoman dibular Joint Disorders The most common cause
of facial pain is a group of conditions called temporomandibular joint and muscle disorders
(TMJD). These disorders cause recurrent or
chronic pain and dysfunction in the jaw joint and
its associated muscles and supporting tissues.
TMJD are the second most commonly occurring
musculoskeletal condition resulting in pain and
disability (after chronic low back pain), affecting
50
45
40
35
30
25
20
15
10
5
0
All Ages
0 to 19
20 to 29
All Races
Black
30 to 39
AI/AN
40 to 49
API
50 to 59
Hispanic
60 to 69
70 to 79
80+
White Non-Hispanic
Fig. 2 Incidence of oral cancer cases per 100,000 among different age and racial groups (Data Source: Oral Cancer
Incidence by Age, Race and Gender, 2009. National Institute of Dental and Craniofacial Research)
A Life Course Health Development Perspective on Oral Health
approximately 5–12% of the population, with an
annual cost estimated at $4 billion. About half to
two-thirds of those with TMJD seek treatment;
among those seeking treatment, approximately
15% develop chronic TMJD (NIDCR 2003).
3
Evolution of Oral Health
Paradigms
The evolution of concepts regarding oral health
and associated analytical models designed to
measure oral health in the aggregate and its
various components have paralleled the evolution of general health concepts, albeit frequently
with a variable time lag. Ancient theories and
hypotheses have given way to more scientifically
grounded concepts reflecting changing scientific
paradigms, initially based on biomedical models
of disease causation, followed by biopsychosocial models that attempted to incorporate a wider
range of factors influencing health and, more
recently, Life Course Health Development. The
following section provides a broad overview of
that progression, focusing primarily on concepts
regarding dental conditions.
307
on Pasteur’s discovery that bacteria can ferment
sugars into lactic acid, formulated the chemoparasitic theory of caries. This theory held that
tooth decay is caused by acids, produced by oral
bacteria following fermentation of ingested sugars, which lead to loss of mineral from teeth
(demineralization) (Miller 1890). Miller’s second
major contribution was the focal infection theory,
which hypothesized that oral microorganisms or
their products have a role in the development of a
variety of diseases in sites removed from the oral
cavity, including brain abscesses, pulmonary diseases, and gastric problems (Miller 1891). Later
work by Keyes in the 1960s led to explanations
of the mechanisms by which dental caries develops based on the interaction of cariogenic bacteria in biofilm/dental plaque and dietary substrates
that lead to acid production, which in turn leads
to demineralization of tooth structure (illustrated
in the Venn diagram in Fig. 5).
Tooth Worm Theory The concept of a tooth
worm which according to prevailing popular
beliefs caused caries and periodontitis existed
in diverse cultures across the ages and, despite
being labeled by medical doctors as superstition during the time of the Enlightenment, persisted in some cultures into the twentieth century.
Numerous popular “therapies” were applied to
eradicate the tooth worm, including fumigations
with henbane seeds, magical formulas, and oaths
(Gerabek 1999).
Concept of Caries as a Specific and
Transmissible Infection The concept of dental
caries being infectious and transmissible is based
on the biomedical model of disease causation,
and grew out of well-designed rodent studies performed by Keyes (Keyes 1960) showing that caries only developed in rodents when they were
caged with or ate the fecal pellets of groups of
caries-active rodents. Further proof emerged
when certain streptococci isolated from caries
lesions in hamsters, unlike other types of streptococci, caused rampant decay in previously cariesinactive animals (Fitzgerald and Keyes 1961).
The bacteria, later identified as Streptococcus
mutans (SM), gave rise to the concept of caries
being due to a specific infection with mutans
streptococci (MS), a concept that has gained
wide support within the field of caries microbiology (Fejerskov 2004). Subsequent studies conducted from the mid-1970s through the 1990s
demonstrated that infants acquire MS from their
mothers and that MS can colonize the mouths of
infants even before teeth erupt (Berkowitz 2006).
Chemo-Parasitic
and
Focal
Infection
Theories In 1890, W.D. Miller (an American
dentist and the first oral microbiologist), building
Conceptualizing Caries as a Complex, Chronic
Disease More recently, Featherstone (Feathersone
2000) used a modification of the chronic disease
3.1
Changing Concepts of Dental
Disease: From Worms
to Germs to Chronic Disease
Terms
J.J. Crall and C.B. Forrest
308
model, referred to as the “caries balance,” to
describe caries as a dynamic process that depends
on the balance between constellations of risk factors (e.g., high levels of cariogenic bacteria,
reduced salivary function, cariogenic dietary practices, tooth structure anomalies) and protective factors (e.g., salivary components and flow, exposure
to fluorides, anti-cariogenic dietary components).
Although the caries balance concept highlights the
importance of risk factors and protective factors
and the potential for dynamic changes over time,
published examples generally depict factors related
to biomedical models rather than broader biopsychosocial models.
The shift from conceptualizing dental caries
based on biomedical models to models based on
biopsychosocial theory represents a relatively
recent paradigm change. These newer models
recognize that caries expression not only depends
on the complex interplay between saliva, dietary
habits, and many biological determinants related
to biofilm composition and metabolism; it also
depends on those factors acting in concert with
innumerable other biological, behavioral, and
social factors acting at the level of individuals
and populations. In summarizing the implications of this paradigm shift, Fejerskov (Fejerskov
2004) highlighted the following:
By appreciating that dental caries belongs to the
group of common diseases considered as ‘complex’ or ‘mulifactorial’ such as cancer, heart diseases, diabetes, and certain psychiatric illnesses,
we have to realize that there is no simple causation
pathway. It is not a simplistic problem such as
‘elimination of one type of microorganism’, or a
matter of improving ‘tooth resistance’. Complex
diseases cannot be ascribed to mutations in a single
gene or to a single environmental factor. Rather
Fig. 3 Common risk
factor model of oral
disease and other
chronic diseases
(Source: Tomar 2012)
they arise from the concerted action of many
genes, environmental factors, and risk-conferring
behaviors. … [This concept also] explains why
dental caries has to be controlled lifelong if a functional dentition is to be maintained.
These new concepts explain why . . . several of the
‘old’ recommended preventive programs are no
longer effective. It is of course not because the
agents we used in prevention are no longer efficacious. They just become ineffective because the
caries incidence rate has changed as the environment has changed.
This brief overview highlights changes in popular and scientific concepts regarding the etiology
of common dental diseases and methods for preventing or minimizing their consequences over
time. Nevertheless, the persistence of relatively
high levels of dental disease in sizeable segments
of the population and substantial disparities within
and across populations serve to highlight the limitations of traditional concepts and approaches.
3.2
Common Risk Factors, Social
Determinants, and Ecological
Models
Common Risk Factors As noted above, there is
growing recognition that major oral healthrelated conditions are complex (multifactorial),
chronic conditions whose occurrence and severity across the lifespan depend on interactions of a
broad array of biological, behavioral, social, and
environmental factors, which also are implicated
in other chronic diseases and conditions as indicated in Fig. 3 (Fejerskov 2004; Sheiham and
A Life Course Health Development Perspective on Oral Health
Watt 2000; Larson et al. 2008; Tomar 2012). We
now realize that different types of organisms are
involved in the etiology of dental caries and periodontal diseases and that the ecology of the oral
microbiome is influenced by an array of common
biological and biopsychosocial factors. We recognize that craniofacial disorders are caused by
developmental disturbances that occur in utero
and are influenced by genetic and environmental
factors. We know that oral cancers result from
mutations in genes that control cell behavior and
are influenced by exposure to carcinogens (most
notably tobacco) and biological factors, including viruses and fungi. Evidence also indicates
that risk factors implicated in TMJD include poor
alignment of teeth, stress, parafunctional habits,
arthritis, trauma, and structural developmental
abnormalities.
Cumulative Impact of Multiple Social
Determinants Larson et al. (Larson et al.
2008) highlighted the cumulative impact of
multiple social risk factors (limited parental
education, low family income, single-parent
309
household, race, being uninsured, family conflict, poor maternal mental health, living in an
unsafe neighborhood) on various aspects of
children’s health, including their oral health.
As shown in Fig. 4, the percentage of parents
who reported that their children had suboptimal
oral health and other indicators of poor health
rises with increases in the number of prevailing social risk factors, with the relationship
between social risk factors and poor oral health
demonstrating the steepest linear gradient. The
proportion reporting less than very good teeth
ranged from 14% for children with no social
risk factors to 64% for children with ≥6 social
risk factors. Analyses that controlled for child
age, gender, and number of children in the
household showed an almost 11-fold increase
in the odds for less than very good teeth in children with ≥6 versus no social risks, underscoring the importance of social structure to healthy
development.
Ecological Models of Caries and Periodontal
Disease Figure 5 depicts a multidimensional
Fig. 4 Percentage of children in worse health by number of social risk factors (Source: Larson et al. 2008)
310
J.J. Crall and C.B. Forrest
Fig. 5 Influences on children’s oral health: a conceptual model (Source: Fisher-Owens et al. 2007)
ecological model for childhood dental caries,
enumerating an array of biopsychosocial influences (including six child-level, eight familylevel, and eight community-level influences
identified in the literature) in addition to the
Keyes classic biological triad model and the
influence of environmental factors and time
(Fisher-Owens et al. 2007). The bulk of published studies underlying ecological models such
as this have study design limitations similar to
those identified in conducting the literature
review for this chapter. Accordingly, their utility
with respect to LCHD studies lies in helping to
identify disease correlates and generate hypotheses, not in establishing oral disease or health
development pathways.
Ecological models like the one shown in Fig. 5
have been used to design a variety of caries-risk
assessment instruments. However, most cariesrisk assessment instruments include only a subset
of the broad range of influences depicted in the
Fisher-Owens model and generally have not
undergone extensive validity testing (Quinonez
and Crall 2009).
A Life Course Health Development Perspective on Oral Health
311
Social Determinants
Familial Determinants
Health care
system
Political
system
Employment
situation
Economic
system
Public policies
Educational
system
Family structure
Behaviors
Culture
Individual Determinants
Sex
Smoking
Diabetes
Social
structure
Host and Genetic Factors
Public health
infrastructure
Family health
status
Family
norms
Oral hygiene
Stress
Use of dental
services
Periodontal status
Bacterial biofilm
Social
inequalities
SES
SES
Physical
environment
Social
norms
Immune and inflammatory
factors
Fig. 6 Social determinants of oral health and disease in US men (Source: Tomar 2012)
Figure 6 depicts an ecological conceptual
model for periodontal disease using a framework
similar to the Fisher-Owens et al. model for
childhood caries (Tomar 2012). In this case, the
biological model emphasizes the interactions
among host and genetic factors, bacteria associated with periodontal disease (different and distinct from cariogenic bacteria), and immune and
inflammatory factors—reflecting a different
pathogenic mechanism for periodontal disease
than for dental caries. Constellations of individual, familial, and social influences are identified
within categories of social, behavioral, and contextual factors in this model.
Ecological models like those shown in Figs. 5
and 6 are useful for depicting the complex array
of etiological factors that potentially influence
disease development, but are of limited value for
explaining disease mechanisms or pathways as
they do not explicitly incorporate developmental
mechanisms or time into the frameworks. LCHD
principles address these gaps by building on ecological models, adding time dimensions and
developmental pathways to the models.
4
Research Applications of Life
Course Concepts to Oral
Health
4.1
Life Course Epidemiology
Life course epidemiology adds the time dimension to disease causation models, moving beyond
the limitations of models such as those shown in
the preceding section which depict an unspecified, essentially cross-sectional interplay among
nested layers of environmental and individual
factors. According to Kuh and Ben-Shlomo
(Ben-Shlomo and Kuh 2002), “Life course epidemiology is defined as ‘the study of long-term
effects on chronic disease risk of physical and
social exposures during gestation, childhood,
adolescence, young adulthood and later adult
life. It seeks to understand causal links between
exposures and outcomes, taking into consideration the importance of time (duration) and timing in the disease development.” In a recent paper
on concepts and theoretical models of life course
epidemiology and its relevance to chronic oral
J.J. Crall and C.B. Forrest
312
conditions, Nicolau et al. (Nicolau et al. 2007)
noted that “The life course approach to studying
chronic disease etiology is not merely a collection of longitudinal data or the use of a particular
study design or analytical method. Rather, the
unique feature of this approach is a theoretical
framework which assumes and tests a temporal
ordering of exposure variables and their interrelationship with a specific outcome.” The following
section provides several examples of applications
of life course epidemiology to studies of chronic
oral diseases and conditions.
4.2
Oral Health Trajectories:
Dunedin Cohort Studies
A number of studies published since the turn
of the twenty-first century have begun to more
fully embrace life course research methodologies and examine a broader array of life course
influences. Foremost among them from the
standpoint of assessing changes in oral health
status over time and the influence of socioeconomic influences beginning in early childhood
is the Dunedin Multidisciplinary Health and
Development Study—a longitudinal investigation of health and behavior in a complete birth
cohort of study members born in Dunedin,
New Zealand, between April, 1972 and March,
1973. Findings of several Dunedin studies are
summarized below.
Poulton et al. (Poulton et al. 2002) assessed a
number of health outcomes, including dental caries, dental plaque scores, gingival bleeding, and
periodontal disease status in a cohort of 1000
26-year-old Dunedin Study subjects whose
socioeconomic status (SES) had been documented at birth and at ages, 3, 5, 7, 9, 11, 13, and
15 years. All dental health measures at age
26 years showed a graded relation with childhood
SES. As SES increased, the amount of plaque
and gingival bleeding and the proportion of individuals with periodontal disease and decayed
surfaces decreased. The adverse influence of low
childhood socioeconomic status was seen after
controlling for infant health and contemporaneous adult SES. Additionally, the results showed
that low adult SES had a significant effect on
poor adult dental health after controlling for low
childhood SES. The authors concluded that their
“findings document that the social gradient in
health—which has been amply documented
among middle-aged and older adults—actually
emerges in childhood. Whereas clinical and
research interest in the social gradient has been
generated mostly by studies of adults, the findings from this study suggest that the social gradient can be scrutinised in paediatric and adolescent
populations as well. Further, whereas most studies of the social gradient have narrowed their
attention to specific diseases, such as
cardiovascular diseases, we document that the
social gradient is far more ubiquitous and troubling. Low social class adversely affects many
areas of people’s health, including their physical,
dental and mental health” (Poulton et al. 2002).
This finding concerning children’s dental health
is highly noteworthy because there are few pediatric conditions that show a substantial SES gradient, which raises the possibility that body
systems that develop rapidly and dramatically
during childhood, such as the structures and
physiological systems of the oral cavity, are more
vulnerable to, and perhaps dependent on, environmental exposures.
Thomson et al. (2004) analyzed data on 789
Dunedin Study subjects at ages 5 and 26 years to
investigate whether adult oral health status is
influenced by (a) childhood socioeconomic
advantage or disadvantage (controlling for childhood oral health) or (b) oral health in childhood
(controlling for childhood socioeconomic advantage or disadvantage) and whether oral health in
adulthood is affected by changes in SES. With
respect to the question of whether poor adult oral
health is predicted by socioeconomic disadvantage in childhood, after controlling for childhood
oral health, analyses revealed that oral health
inequalities present at age 5 years were also
apparent at age 26 years when the early childhood SES categories were used, suggesting that
early socioeconomic inequalities in a number of
important oral health indicators do persist well
into the third decade of life. Concerning the question of whether poor adult oral health is predicted
A Life Course Health Development Perspective on Oral Health
interest from the standpoint of LCHD is the possibility that oral health may represent a sentinel
system, with early manifestations of disease serving as a harbinger for elevated risk of other
chronic diseases (e.g., obesity, diabetes).
Broadbent et al. (2008) investigated longitudinal patterns of caries experience in a birth cohort
of 955 Dunedin Study subjects in order to identify and describe developmental trajectories of
caries experience in the permanent dentition
using data collected from dental examinations at
ages 5, 9, 15, 18, 26, and 32 years. Three caries
experience trajectory classes were identified:
“high” (15% of subjects), “medium” (43%), and
“low” (42%) with respect to decayed, missing,
and filled (tooth) surfaces (DMFS). All trajectories were relatively linear, although the higher
trajectories were more “S-shaped” (Fig. 7). This
effect disappeared following adjustment for the
number of unaffected surfaces remaining at each
age, suggesting that, among individuals following a similar caries trajectory, the caries rate is
relatively constant across time. This finding is
40
50
by poor oral health in childhood, after controlling
for childhood socioeconomic status, the evidence
was unequivocal regarding dental caries: having
high caries disease experience early in life predicted having greater disease experience in adulthood, other factors being equal. The pattern was
not as clear with respect to periodontal disease,
however. Finally, in examining the impact of
individuals’ SES at ages 5 and 26, Thomson and
colleagues (Thomson et al. 2004) found that, for
nearly all oral health indicators, a clear gradient
for disease severity and prevalence was observed
across socioeconomic trajectory groups in the
following ascending order: “high (at age 5)–high
(at age 26),” “low–high” (upwardly mobile),
“high–low” (downwardly mobile), and “low–
low.” These findings led the authors to conclude
that adult oral health is predicted by not only
childhood socioeconomic advantage or disadvantage but also by oral health in childhood. Changes
in socioeconomic advantage or disadvantage
between ages 5 and 26 were associated with differing levels of oral health in adulthood. Also of
313
10
Medium trajectory
N = 427 (44.7%)
0
20
DMFS
30
High trajectory
N = 144 (15.1%)
Low trajectory
N = 384 (40.2%)
5
9
15
18
Age (years)
26
32
Bars represent inter quartile range, dotted lines represent 95% CI
DMFS: Decayed, Missing, and Filled surfaces
Fig. 7 Trajectory patterns of dental caries experience in the permanent dentition to the fourth decade of life (Source:
Broadbent et al. 2008)
314
consistent with observations by Fejerskov (2004)
that the caries incidence rate in a group of individuals appears fairly constant throughout life if
no special efforts to control lesion progression
are made. Broadbent et al. (2008) concluded that
these results did not support the commonly held
belief among dentists that childhood and adolescence are periods of special risk for dental caries
or that caries “immunity” may be acquired during late adolescence or early adulthood. In fact,
other studies have reported that ECC is one of the
most reliable predictors of elevated risk of caries
experience later in life.
Crocombe et al. (2011) also analyzed data
from 833 participants in the Dunedin Study to
assess the impact of dental visiting trajectory
patterns on clinical oral health and oral healthrelated quality of life measures. Information on
the use of dental services was collected at ages
15, 18, 26, and 32. Dental visiting trajectories
were significantly associated with both measures
of self-reported oral health and three of the four
dental clinical indicators. The regular dental
attendance group had less than half the mean
number of missing teeth, a lower mean DMFS
score, and a lower mean decayed surfaces (DS)
than those defined as “opportunists” and “decliners.” The regular group had the lowest mean Oral
Health Impact Profile (OHIP-14) score and was
nearly twice as likely as opportunists and decliners to report that they had better-than-average
oral health. Low SES and being dentally anxious
were associated with worse health outcomes for
all outcome variables, while “poor plaque trajectory” and smoking were associated with worse
outcomes for all variables except filled surfaces
(FS). The influence of dental visiting trajectory
on clinical oral health outcomes was much
higher at lower SES levels than higher SES levels, leading the authors to conclude that improving dental visiting behavior among people of
lower SES would improve clinical oral health
outcomes more and have a greater reduction of
oral health impacts than it would for people of
higher SES.
A number of additional studies of shorter
duration and/or employing retrospective meth-
J.J. Crall and C.B. Forrest
ods of data collection (which may be subject to
some degree of recall bias) have been conducted in other countries, most notably Brazil,
Norway, the United Kingdom, and Hong Kong
(Lu et al. 2011; Holst; Peres et al. 2011; Mason
et al. 2006). Collectively, these studies generally affirm the findings from analyses conducted as part of the Dunedin Study, with some
variation that likely reflects varying levels of
SES (particularly poverty) and differences in
the types of oral health-care systems and programs available across countries. Many of these
studies are limited to adolescents (starting at
age 15) or adults. An exception is the work of
(Holst and Schuller 2011) which examined different birth cohorts in Norway and found that
the health environment in childhood was important for adults’ oral health during most of the
twentieth century. Attention from parents and
the local environment lead to better oral health
outcomes in adulthood. Social status measured
by length of education also was associated with
choices leading to better oral health. Regular
dental visits were important especially for the
eldest birth cohort. Good oral health behaviors
early and during adulthood were important for
oral health.
The studies summarized in this section represent advances in our understanding of the
impacts of various influences on oral health
over the life course. Interest in using a life
course perspective to study various aspects of
oral health has grown steadily in recent times.
Main lines of pursuit include investigations of
critical periods and accumulation of risk models, which have shed light on the importance of
early life experiences, oral health trajectories,
and the importance of social determinants to
oral and general health (Heilman et al. 2015).
However, it is worth noting that life course epidemiology only reflects the principles of LCHD
(see Table 1) in a very limited way, primarily
with respect to LCHD Principle 3 (Complexity:
multidimensional person-environmental interactions) and LCHD Principle 5 (Timing: health
is highly sensitive to timing and social structuring of environmental exposures).
A Life Course Health Development Perspective on Oral Health
4.3
Relating Oral Health, General
Health, and Quality of Life
Summarizing what is known about the influences
of oral health and oral health problems on life
course outcomes, general health and overall quality of life is a daunting challenge that goes beyond
the scope of this chapter. In lieu of attempting
such a synopsis, the reader is referred initially to
the US Surgeon General’s Report on Oral Health
(US Department of Health and Human Services
2000) for a broad introduction to the literature.
Chapters 5 and 6 of that extensively researched
and widely vetted document are summarized
below.
Chapter 5 of the SGROH examines multiple
linkages between oral and general health, noting
that the mouth and the face reflect signs and
symptoms of health and disease that can serve as
an adjunct for diagnosis for some conditions.
Diagnostic tests using oral cells and fluids—
especially saliva—are available to detect drug
abuse, hormonal changes, and specific diseases,
and more are being developed. The mouth also is
a portal of entry for pathogens and toxins, which
can affect the mouth and, if not cleared by the
many defense mechanisms that have evolved to
protect the oral cavity, may spread to the rest of
the body. Recent epidemiologic and experimental
animal research provides evidence of possible
associations between oral infections—particularly periodontal disease—and diabetes, cardiovascular disease, and adverse pregnancy
outcomes. The review in Chapter 5 of the
SGROH also highlights the need for an aggressive research agenda to better delineate the specific nature of these associations and the
underlying mechanisms of action.
Chapter 6 of the SGROH looks at the impact
of oral health problems on quality of life and
includes examples of the kinds of questionnaires
used to measure oral-health-related quality of
life. Oral health generally is highly valued by
society and individuals, and the chapter begins
with a brief description of the reflections of those
values in myth and folklore concerning facial
appearance and the meaning of teeth. It then
explores dimensions beyond the biological and
315
the physical to examine how oral diseases and
disorders can interfere with the functions of daily
living, including participation in work or school,
and what is known about their psychosocial
impacts and economic costs. The deleterious
effects of facial disfigurement and tooth loss may
be magnified in modern societies that celebrate
youth and beauty. Self-reported impacts of oral
conditions on social functions include limitations
in communication, social interactions, and intimacy. Additional research on oral-health-related
quality of life is recommended to permit further
exploration of the dimensions of oral health and
well-being.
Sheiham (2005) also has provided a commentary on the relationships among oral health, general health, and quality of life. Many of the points
highlighted in this commentary relate to LCHD
Principle 1 (Health Development) that health is an
emergent property and that oral health is a component of and contributes to overall health. Sheiham’s
summary ultimately reiterates the SGROH’s call
for cessation of “the compartmentalization
involved in viewing the mouth separately from the
rest of the body, noting that oral health affects general health by causing considerable pain and suffering and by altering what people eat, their
speech, and their quality of life and well-being.”
Additional excerpts from Sheiham’s commentary include the following. “Oral health also has
an effect on other chronic diseases, and failure to
tackle social and material determinants and
incorporate oral health into general health promotion means that millions suffer intractable
toothache and poor quality of life, and end up
with few teeth.
Oral diseases are the most common of the
chronic diseases and are important public health
problems because of their prevalence, their
impact on individuals and society, and the
expense of their treatment. The general determinants of oral diseases are known and include risk
factors common to a number of chronic diseases:
diet, inadequate hygiene practices, smoking,
alcohol, risky behaviors causing injuries, and
stress. Moreover, effective methods are available
to prevent or reduce the impact of major oral
diseases.
J.J. Crall and C.B. Forrest
316
Oral health affects people physically and psychologically and influences how they grow, enjoy
life, look, speak, chew, taste food and socialize,
as well as their feelings of social well-being.
Severe caries detracts from children’s quality of
life: they experience pain, discomfort, disfigurement, acute and chronic infections, and eating
and sleep disruption as well as higher risk of hospitalization, high treatment costs and loss of
school days with the consequently diminished
ability to learn. Caries affects nutrition, growth
and weight gain. Children of three years of age
with nursing caries weigh about 1 kg less than
control children because toothache and infection
alter eating and sleeping habits, dietary intake
and metabolic processes. Disturbed sleep affects
glucocorticoid production. In addition, there is
suppression of hemoglobin from depressed
erythrocyte production.
Chronic diseases such as obesity, diabetes and
caries are increasing in developed and developing
countries, with the implication that quality of life
related to oral health, as well as general quality of
life, may deteriorate. Because oral and other
chronic diseases have determinants in common,
more emphasis should be on the common risk
factor approach. The key concept underlying
future oral health strategies is integration with
this approach, a major benefit being the focus on
improving health conditions in general for the
whole population and for groups at high risk,
thereby reducing social inequities.”
5
Relating Oral Health
and LCHD Concepts
5.1
Relating Oral Health Research
Findings to Key LCHD
Concepts
Pathways and Trajectories Analyses based on
the Dunedin prospective, longitudinal birth
cohort studies have distinguished three oral
health trajectory groupings (high, medium, low)
that emerge as early as age 5, remain relatively
constant over time, and exhibit increasing divergence through the third decade of life. Authors of
the Dunedin Study acknowledge that aggregating
their findings into three trajectory ‘groups’ is an
analytical convenience that may mask the heterogeneity of individuals within each group. To that
point, LCHD suggests that a focus on intraindividual trajectories rather than on “group” means
may prove more insightful because it allows for
careful ascertainment of temporal exposure-outcome relationships and allows the analysis to
take advantage of the diversity and complexity of
these interactions rather than masking variability
by aggregation into a few classes. Classes assume
homogeneity of persons within a group, an
assumption that is generally not supported. Little
if any investigations of intraindividual change
with detailed ascertainment of exposure and outcome over time have been reported in the dental/
oral health literature.
Additional Dunedin cohort studies have examined the effect of SES in childhood and adulthood on adult oral health, and the effect of early
childhood oral health on adult oral health, and
found that the highest oral health trajectories are
exhibited by those who have higher SES and better oral health in childhood; the lowest trajectories are found in those who have low SES and
poor oral health in childhood; and those whose
SES changed (either upwardly or downwardly)
between childhood and adulthood fall between
the other two trajectories. Oral health-care utilization and other personal, family, and community factors (e.g., oral hygiene, diet, dental
anxiety, emphasis placed on oral health within
one’s family or community) also appear to exert
influences on oral health trajectories throughout
the life course.
Early Programming Evidence concerning the
impact of early programming on oral health over
the lifespan is less well defined, in part because
many studies focus on adolescents or adults.
Developmental disturbances that result in structural anomalies of tooth structure represent one
documented example of early programming
influences that can increase risk for dental disease (Targino et al. 2011). Findings from studies
conducted by Holst and Schuller (2011) and Alm
(Alm et al. 2008) suggest that good oral hygiene
A Life Course Health Development Perspective on Oral Health
317
habits, including the use of fluoride toothpaste,
established in early childhood provide a foundation for a low risk of proximal caries in adolescents. Evidence for the influence of less direct
influences such as low birth weight is more
equivocal, but represents an area of growing
research interest (Nicolau et al. 2007). Evidence
of intergenerational programming has been documented, particularly with respect to maternalchild oral health relationships (Shearer and
Thomson, 2010; Shearer et al. 2011).
protective factors for major dental conditions
(see Figs. 5 and 6). The influence of genetic, epigenetic, family, community, social, and environmental risk factors, in individual populations or
across populations, is less well documented and
understood.
Critical or Sensitive Periods Some evidence,
including studies cited above, supports the critical or sensitive period hypothesis with respect
to the impact of various influences during fetal,
early childhood, and later stages on life course
oral health development and the determinants
of oral health disparities. The importance and
etiological mechanisms of disturbances arising during the prenatal period on craniofacial
developmental problems such as cleft lip and/or
cleft palate have been extensively documented.
However, much remains to be elucidated with
respect to the effects of different influences, at
different times, on other oral health-related conditions (dental caries, periodontal disease, cancer, TMJD) (Hallqvist et al. 2004).
Researchers involved in studying life course
influences on oral health have identified a number of gaps in the current knowledge base. Using
the lens of the LCHD principles, we have compiled research needs for future studies on oral
health:
Cumulative Impact Substantial evidence also
exists to support the hypothesis that the impact of
major oral health-related conditions is a function
of cumulative experiences or episodes of disease
over time. Clearly, it is well established that dental caries is a chronic, cumulative disease and that
the caries status of an individual develops and is
subject to biological, behavioral, social, and environmental influences over time. Caries occurs at
any stage in life, provided that an individual has
susceptible teeth (and surfaces) remaining
(Broadbent et al. 2008). The same case can be
made for periodontal disease and tooth loss
resulting from caries and periodontal diseases
(Shearer et al. 2011; Watt 2007).
Risk and Protective Factors Considerable evidence has been compiled with respect to the biological and, to a lesser extent, behavioral risk and
5.2
Gaps in Knowledge
Concerning Oral Health
from a LCHD Perspective
• Evaluate the contribution of social isolation,
social relationships, and social support to oral
health.
• Obtain more information on the economic,
political, social, and environmental causes of
individual behaviors related to oral health.
• Increase the number of longitudinal studies
assessing the long-term effects of early childhood caries (ECC) and treatment on the health
and quality of life of preschool children.
• Longitudinal studies are needed in order to
obtain more knowledge about causative factors
and the possible relationships between dental
caries and overweight/obesity in children.
• Conceptual work that articulates a definition
of oral health motivated by the LCHD principles is needed.
• Theoretical explanations for health inequalities are limited and biased due to the types
of data collected in modern epidemiological
studies, which are geared toward identifying
and quantifying risk factors for disease and
intended as a basis for description (of disease), not explanation (of causal pathways).
This raises questions about the validity of current explanations. Current epidemiological
approaches are widely criticized for neglecting
broad social factors and failing to dig below
the surface into issues such as how differ-
J.J. Crall and C.B. Forrest
318
•
•
•
•
•
•
•
ent social class groups live their lives and
what factors influence their lifestyle decisions. Current explanations are based largely
on what epidemiological researchers can see
and measure; factors that are harder to assess
(such as culture), but which may be critically
important to advancing our understanding of
social inequalities in health, are frequently
neglected.
Epidemiologists have largely persisted in the
use of disease-based measures to assess
inequalities in health and oral health. The
most significant advance to our understanding
of social inequalities in health may come from
the creation of a data set that measures oral
health as defined by the FDI or oral-healthrelated quality of life.
To date there is no research exploring how
members of the population understand and
account for inequalities in oral health, which
remains a significant omission as it could offer
promising new insights.
Further research is needed to clarify the
apparently differing oral health beliefs, attitudes, and practices of lower and higher SES
groups.
Much work remains to be done to establish
which biological, social, and environmental
factors are determinants of oral health or disease and which are merely markers, “proxies,”
or confounders.
The process by which social stratification
translates to poor oral health beginning in earliest childhood, especially for groups at higher
risk for disease, is not well understood.
There is a need for future research to move
beyond traditional risk factors and more
closely examine the impact of the social environment on oral health beliefs, behavior, and
outcomes.
A number of oral health problems have been
shown to be associated with other health conditions (e.g., low birth weight, cardiovascular
disease, respiratory disease); however, the
demonstration of actual causal connections or
pathways, such as the bidirectional relationship between diabetes and periodontal disease, has been modest.
5.3
Recommendations
for Research and Policy
Priorities
The knowledge gaps noted above give rise to a
number of recommendations for research priorities
to better understand oral health life course influences and policy changes to help reduce the impact
of oral health problems and reduce oral health
inequalities, which can be summarized as follows:
• Greater support for longer-term longitudinal
oral health life course studies with detailed
and frequent data collection to allow for the
construction of intraindividual trajectories
• Replication of true oral health life course studies in different populations to assess whether
findings apply across diverse settings
• Greater emphasis on research delineating relationships among oral health determinants to
identify causal pathways
• Greater emphasis on identifying and addressing broader influences on oral health (i.e.,
family, community, social, and environmental
influences)
6
Summary
This chapter has outlined major concepts embodied
in the Life Course Health Development framework,
examined evidence relating various aspects of
major oral health-related conditions to this framework, and produced recommendations for advancing research and policy concerning oral health.
LCHD provides a highly useful approach for understanding oral health determinants, disparities, and
influences on general health and well-being and for
advancing knowledge, policies, and programs to
optimize health across individuals and populations.
References
Agency for Healthcare Research and Quality. 2015. Dental
Services Mean and Median Expenses per Person With
Expense and Distribution of Expenses by Source of
Payment: United States, 2012. Medical Expenditure
A Life Course Health Development Perspective on Oral Health
Panel Survey Household Component Data. Generated
interactively. (3 Mar 2015).
Alm, A., et al. (2008). Oral hygiene and parent-related factors
during early childhood in relation to approximal caries at
15 years of age. Caries Research, 42(1), 28–36.
American Cancer Society. (2015). Cancer Facts &
Figures 2015. Atlanta: American Cancer Society.
Ben-Shlomo, Y., & Kuh, D. (2002). A life course approach
to chronic disease epidemiology: Conceptual models,
empirical challenges and interdisciplinary perspectives.
International Journal of Epidemiology, 31, 285–293.
Berkowitz, R. J. (2006). Mutans steptococci: Acquistion
and transmission. Pediatric Dentistry, 28, 106–109.
Borrell-Carrio, F., Suchman, A. L., & Epstein, R. M.
(2004). The biopsychosocial model 25 years later:
Principles, practice, and scientific inquiry. Annals of
Family Medicine, 2, 576–582.
Boulet, S. L., Grosse, S. D., Honein, M. A., & CorreaVillasenor, A. (2009). Children with orofacial clefts:
Health care use and costs among a privately insured
population. Publ Health Reports, 124, 447–453.
Brennan, D. S., Spencer, A. J., & Roberts-Thomson, K. F.
(2008). Tooth loss, chewing ability and quality of life.
Quality of Life Research, 17, 227–235.
Broadbent, J. M., Thomson, W. M., & Poulton, R. (2008).
Trajectory patterns of dental caries experience in
the permanent dentition to the fourth decade of life.
Journal of Dental Research, 87, 69–72.
Center for Disease Control and Prevention (CDC).
(2014). Prevalence (number of cases) of cleft
lip and cleft palate. Available at: http://www.
nidcr.nih.gov/DataStatistics/FindDataByTopic/
CraniofacialBirthDefects/PrevalenceCleft%20
LipCleftPalate.htm.
Crocombe, L. A., et al. (2011). Dental visiting trajectory patterns and their antecedents. Journal of Public
Health Dentistry, 71(1), 23–31.
Dye, B. A., Thornton-Evans, G., Li, X., & Iafolla, T. J.
(2015a). Dental caries and sealant prevalence in
children and adolescents in the United States, 2011a–
2012. NCHS data brief, no 191. Hyattsville: National
Center for Health Statistics.
Dye, B. A., Thornton-Evans, G., Li, X., & Iafolla, T. J.
(2015b). Dental caries and tooth loss in adults in the
United States, 2011b–2012. NCHS data brief, no 197.
Hyattsville: National Center for Health Statistics.
Feathersone, J. B. D. (2000). The science and practice
of caries prevention. Journal of the American Dental
Association (Chicago, IL), 131, 887–899.
Fejerskov, O. (2004). Changing paradigms in concepts
on dental caries: Consequences for oral health care.
Caries Research, 38, 182–191.
Fine, A., & Kotelchuck, M. (2010). Rethinking MCH:
The life course model as an organizing framework.
Washington, DC: US Department of Health and
Human Services, Health Resources and Services
Administration, Maternal and Child Health Bureau.
Fisher-Owens, S. A., Gansky, S. A., Platt, L. J., et al.
(2007). Influences on children's oral health: A conceptual model. Pediatrics, 120, e510–e520.
319
Fitzgerald, R. J., & Keyes, P. H. (1961). Demonstration
of the etiologic role of streptococci in experimental
caries in the hamster. Journal of the American Dental
Association (Chicago, IL), 61, 9–19.
Gerabek, W. E. (1999). The tooth-worm: Historical
aspects of a popular medical belief. Clin Oral Invest,
3, 1–6.
Glick, M., et al. (2016). A new definition for oral health
developed by the FDI World dental federation opens
the door to a universal definition of oral health.
Journal of the American Dental Association (Chicago,
IL), 147, 915–917.
Gregory, J., Gibson, B., & Robinson, P. G. (2005).
Variation and change in the meaning of oral health
related quality of life: A ‘grounded’ systems approach.
Social Science & Medicine, 60, 1859–1868.
Halfon, N., & Hochstein, M. (2002). Life course health
development: An integrated framework for developing
health, policy, and research. The Milbank Quarterly,
80, 433–479.
Halfon, N., Larson, K., Lu, M., Tullis, E., & Russ, S. (2014).
Lifecourse health development: Past, present and future.
Maternal and Child Health Journal, 18, 344–365.
Hallqvist, J., et al. (2004). Can we disentangle life
course processes of accumulation, critical period and
social mobility? An analysis of disadvantaged socioeconomic positions and myocardial infarction in
the Stockholm heart epidemiology program. Social
Science & Medicine, 58(8), 1555–1562.
Heilman, A., et al. (2015). Oral health over the life
course. In C. Burton-Jeangros, S. Cullati, & A. Sacker
(Eds.), A life course perspective on health trajectories and transitions [Internet]. Cham (CH): Springer.
doi:10.1007/9783319204840_3.
Hollister, M. C., & Weintraub, J. A. (1993). The association of oral status with systemic health, quality of
life, and economic productivity. Journal of Dental
Education, 57(12), 901–912.
Holst, D., & Schuller, A. A. (2011). Equality in adults’ oral
health in Norway. Cohort and cross-sectional results
over 33 years. Community Dent Oral Epidemiol, 39(6),
488–497. doi:10.1111/j.1600-0528.2011.00624.x.
Jacobson, J. J., et al. (2012). The cost burden of oral, oral
pharyngeal, and salivary gland cancers in three groups:
Commercial insurance, Medicare, and Medicaid.
Head & Neck Oncology, 4, 15.
Keating, D. P., & Hertzman, C. (1999). Developmental
health and the wealth of nations : Social, biological,
and educational dynamics. New York, NY: Guilford
Press.
Keyes, P. H. (1960). The infectious and transmissible
nature of experimental dental caries. Archives of Oral
Biology, 1, 304–320.
Kohli, S. S., & Kohli, V. S. (2012). A comprehensive review of the genetic basis of cleft lip and
palate. J Oral Maxillofac Pathol., 16, 64–72.
doi:10.4103/0973-029X.92976.
Larson, K., Russ, S. A., Crall, J. J., & Halfon, N. (2008).
Influence of multiple social risks on children's health.
Pediatrics, 121, 337–344.
320
Liu, Z., et al. (2009). The impact of malocclusion/orthodontic treatment need on the quality of life: A systematic review. The Angle Orthodontist, 79, 585–591.
Locker, D., & Slade, G. (1994). Association between clinical and subjective indicators of oral health status in an
older adult population. Gerodontology, 11, 108–114.
Lu, H. X., et al. (2011). Trends in oral health from childhood to early adulthood: A life course approach.
Community Dentistry and Oral Epidemiology, 39(4),
352–360.
Mason, J., et al. (2006). How do factors at different stages
of the lifecourse contribute to oral-health-related quality of life in middle age for men and women? Journal
of Dental Research, 85(3), 257–261.
Miller, W. D. (1890). The micro-organisms of the human
mouth. Philadelphia: S.S. White Publishing Co..
Miller, W. D. (1891). The human mouth as a focus of
infection. Dental Cosmos, 33, 689–706.
Nicolau, B., et al. (2007). Life-course epidemiology:
Concepts and theoretical models and its relevance to
chronic oral conditions. Community Dentistry and
Oral Epidemiology, 35(4), 241–249.
NIDCR. (2003). Prevalence of TMJD and its signs and
symptoms; Available from: http://www.nidcr.nih.
gov/DataStatistics/FindDataByTopic/FacialPain/
PrevalenceTMJD.htm.
Peres, M. A., et al. (2011). The influence of family income
trajectories from birth to adulthood on adult oral
health: Findings from the 1982 Pelotas birth cohort.
American Journal of Public Health, 101(4), 730–736.
Poulton, R., et al. (2002). Association between children’s experience of socioeconomic disadvantage and
adult health: A life-course study. Lancet, 360(9346),
1640–1645.
Quinonez, R., & Crall, J. J. (2009). Caries-risk assessment in
early childhood. In J. H. Berg & R. Slayton (Eds.), Early
childhood oral health. Hoboken: Wiley-Blackwell.
Ram, H., Kumar, H., Konwar, R., Bhatt, M. L. B., &
Mohammad, S. (2011). Oral cancer: Risk factors and
molecular pathogenesis. J Maxillofac Oral Surg, 10,
132–137.
Shearer, D. M., & Thomson, W. M. (2010). Intergenerational
continuity in oral health: A review. Community Dentistry
and Oral Epidemiology, 38(6), 479–486.
Shearer, D. M., et al. (2011). Maternal oral health predicts their children's caries experience in adulthood.
Journal of Dental Research, 90(5), 672–677.
J.J. Crall and C.B. Forrest
Sheiham, A. (2005). Oral health, general health and quality of life. Bulletin of the World Health Organization,
83(9), 644–644.
Sheiham, A., & Watt, R. G. (2000). The common risk
factor approach: A rational basis for promoting oral
health. Community Dentistry and Oral Epidemiology,
28, 399–406.
Targino, A. G. R., et al. (2011). The relationship of enamel
defects and caries: A cohort study. Oral Diseases,
17(4), 420–426.
The Oral Cancer Foundation. (2015). Oral cancer facts.
Available at: http://www.oralcancer.org/facts/.
Thomson, W. M., et al. (2004). Socioeconomic inequalities in oral health in childhood and adulthood in a birth
cohort. Community Dentistry and Oral Epidemiology,
32, 345–353.
Thornton-Evans, et al. (2013). Periodontitis among adults
aged > United States, 2009–2010. MMWR, 62(3),
129–135.
Tomar, S. L. (2012). Social determinants of oral health
and disease in US men. Journal of Mens Health, 9,
113–119.
U.S. Department of Health and Human Services.
(2000). Oral Health in America: A Report of the
Surgeon General. Rockville: U.S. Department of
Health and Human Services, National Institute
of Dental and Craniofacial Research, National
Institutes of Health.
Vargas, C. M., Crall, J. J., & Schneider, D. A. (1998).
Sociodemographic distribution of pediatric dental caries: NHANES III, 1988-1994. Journal of the American
Dental Association (1939), 129(9), 1229–1238.
Wall, T., & Vujicic M. (2014). No growth in U.S. dental spending in 2013. Health policy Institute research
brief. American Dental Association. December
2014. Available from: http://www.ada.org/~/media/
ADA/Science%20and%20Research/HPI/Files/
HPIBrief_1214_4.ashx.
Watt, R. G. (2007). From victim blaming to upstream
action: Tackling the social determinants of oral
health inequalities. Community Dentistry and Oral
Epidemiology, 35(1), 1–11.
WHO, The World Oral Health Report 2003. (2008).
Continuous improvement of oral health in the 21st
century–the approach of the WHO global oral health
Programme. Community Dent Oral Epidemiol, 31(s1),
3–24.
Open Access This chapter is licensed under the terms of the Creative Commons Attribution 4.0 International License
(http://creativecommons.org/licenses/by/4.0/), which permits use, sharing, adaptation, distribution and reproduction in
any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to
the Creative Commons license and indicate if changes were made.
The images or other third party material in this chapter are included in the chapter’s Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the chapter’s Creative Commons license
and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
Life Course Health Development
Outcomes After Prematurity:
Developing a Community, Clinical,
and Translational Research
Agenda to Optimize Health,
Behavior, and Functioning
Michael E. Msall, Sarah A. Sobotka,
Amelia Dmowska, Dennis Hogan,
and Mary Sullivan
Definitions Late preterm infants (LPIs) 340/7–
366/7 week infants; moderate preterm infants
(MPI) 320/7–336/7 week infants; very preterm
infants (VPI) 280/7–316/7 weeks; extreme preterm infants (EPI) <280/7 weeks; cerebral palsy
(CP); normal birth weight (NBW) ≥2500 g;
low birth weight (LBW) 1500–2499 g; very
low birth weight (VLBW) 1000–1499 g;
extremely low birth weight (ELBW) <1000 g;
IQ = intelligence quotient; ID = intellectual
disability
M.E. Msall, MD (*)
Developmental and Behavioral Pediatrics University
of Chicago, Comer and LaRabida Children’s
Hospitals, Chicago, IL, USA
JP Kennedy Research Center on Intellectual and
Developmental Disabilities, University of Chicago
Comer Children’s Hospital, Section of
Developmental and Behavioral Pediatrics,
950 East 61St Street, SSC Room 207,
Chicago, IL 60637, USA
e-mail: mmsall@peds.bsd.uchicago.edu
1
Introduction
National vital statistics from 2013 estimate that
the rate of preterm birth (<37 weeks of gestation)
involves approximately 1 in 10 infants in the
United States each year (Hamilton et al. 2015).
An estimated 68 per 1000 US live births, over
270,000 annually, are born at late preterm gestation (34–36 weeks). Currently, 28 per 1000 US
live births, more than 100,000 infants annually,
S.A. Sobotka, MD, MS • A. Dmowska, BA
Section of Developmental and Behavioral Pediatrics,
University of Chicago Comer Children’s Hospitals,
Chicago, IL, USA
D. Hogan, PhD
Sociology and Demography, Population Research and
Training Center, Brown University,
Providence, RI, USA
M. Sullivan, PhD, RN
University of Rhode Island, College of Nursing,
Women and Infants Hospital, Providence, RI, USA
© The Author(s) 2018
N. Halfon et al. (eds.), Handbook of Life Course Health Development,
DOI 10.1007/978-3-319-47143-3_14
321
322
are born <34 weeks of gestation, and 56,000 are
born at very low birth weight status (<1500 g).
These infants have high risks for long-term neurodevelopmental disabilities such as intellectual
disability (ID), blindness, sensorineural hearing
loss, and cerebral palsy (CP). There are several
biomedical causes for these high rates of preterm
birth, such as the role of assisted reproductive
technologies increasing the rate of multiple births
(twins, triplets, or quadruplets) and maternal age.
Teenage pregnancy, also a risk factor for prematurity, has instead been decreasing over the past
decade (Hamilton et al. 2015; The March of
Dimes Data Book for Policy Makers 2012).
However, it is very critical to remember that both
very preterm infants (VPI) and extreme preterm
infants (EPI) are heterogeneous, and our understanding of causal pathways that lead to successful interventions is at an early scientific stage
(Rubens et al. 2014). For example, the same factors that create the biological risk in the mothers
for prematurity may be the same factors that
influence the inflammatory, hormonal, and neurochemical regulators that influence the mothers’
caregiving capacity once their premature infant is
born. Therefore, ascribing causality to biomedical, behavioral, or neuro-regulatory factors is difficult and does not allow for the complex systems
understanding required to advance life courseoriented prevention and intervention strategies.
In addition, despite increased access to prenatal
care, there still remain limitations on our ability
to prevent all moderate and late preterm birth
(Requejo et al. 2013).
Estimates of the economic impact of prematurity typically factor in acute care medical costs,
early childhood intervention expenditures, longterm special education, special health care, and
disability costs. A report released by the Institute
of Medicine in 2007 estimated that the economic
burden associated with preterm birth was at least
$26.2 billion in 2005, equivalent to $51,600 per
infant born preterm. These estimates included
$16.9 billion in medical care costs, $1.9 billion in
maternal delivery costs, $611 million for early
intervention services, $1.1 billion for special
education services, and $5.7 billion in lost household and labor market productivity (Preterm
M.E. Msall et al.
Birth: Causes, Consequences, and Prevention
2007). Importantly, each premature/LBW baby
costs employers an average of $54,149 in payments for newborn medical care during the first
year of life, about twelve times that of an uncomplicated newborn (Preterm Birth: Causes,
Consequences, and Prevention 2007). In addition
to medical care, children born preterm are more
likely to experience learning and behavior disabilities later in life, often resulting in poor test
scores, grade repetition, and increased utilization
of special education services. The Institute of
Medicine estimates the costs of special education
at $2200 per year per child (Preterm Birth:
Causes, Consequences, and Prevention 2007),
though for children with major neurodevelopmental disabilities, the special cost per child can
exceed $50,000 per academic year.
High-quality early childhood intervention
programs, however, may minimize the later need
for special education and drastically reduce these
additional costs (Aron and Loprest 2012). Model
programs like the Infant Health Development
Program (IHDP) are successful in improving
educational, behavioral, and life course outcomes
for preterm infants experiencing poverty, thereby
decreasing children’s need for special education
(Aron and Loprest 2012). Economic modeling
estimates the impact of comprehensive early
intervention services on long-term special education costs as having a direct savings of $2.60 for
every dollar invested in early intervention, early
childcare services, and Early Head Start
(Dmowska et al. 2016). When considering costs
incurred from school dropouts, reentry to school,
and additional weight on mental health-care services and the criminal justice system, the savings
would likely double (Dmowska et al. 2016).
Increased access to comprehensive familycentered early intervention services, therefore,
may substantially reduce special education costs
for children born preterm as well as long-term
behavioral and social health costs.
There have been dramatic improvements in
long-term survival for infants born premature and
low birth weight, but developmental outcomes are
far from optimized. Survival has increased dramatically with the regionalization of neonatal
Life Course Health Development Outcomes After Prematurity: Developing a Community, Clinical…
intensive care and the application of obstetrical
and neonatal biomedical interventions for optimizing growth, lung maturity, cardiopulmonary
functioning, and infection control. Overall, these
efforts resulted in decreased rates of major neurodevelopmental disabilities in survivors in the
1980s and 1990s from 25–40% to 15–25%. For
those born with extreme prematurity (<28 weeks
of gestation) and extremely low birth weight status (<1000 g), survival has increased dramatically
from <50% to >80%. However, there remain high
rates of cognitive impairment and intellectual disability (ID), with over 50% of survivors requiring
special education services.
Growing inequality in the United States hits
families with young children very hard. Close to
50% of children younger than 5 are growing up
in households living below 200% of the poverty
level. Research shows that families with this
level of income do not have the economic, social,
educational, and community assets they need to
support optimal health development (Crouter and
Booth 2014). So many of the 8% premature
babies, who start off life with higher levels of
inherent vulnerabilities, are increasingly facing a
family, social, and community landscape that
cannot support their optimal development.
In addition, there is increased recognition that
both moderate and late preterm births (32–
36 weeks of gestation), which account for over
8% of live births or approximately 325,000 children yearly, have increased risk for long-term
health, developmental, and behavioral challenges
(Table 1). In spite of these biomedical advances,
environmental and social conditions that can deleteriously affect the health and well-being of
these already vulnerable children have not only
improved in the United States; there are some
indications that they have in fact worsened.
323
Additionally, there are major gaps in accessing
comprehensive family supports and quality
health, early childhood, and educational and community experiences for recent cohorts of children
at the highest biomedical and social risks.
The purpose of this paper is to apply a life
course health development perspective in order
to identify factors that promote more optimal
health and developmental trajectories and the
mechanisms that underlie resilience for children
with prematurity from a life course health development perspective. We will describe some
research findings about the role of social and
environmental factors among VPI and EPI survivors in aggravating or moderating neonatal risks
for suboptimal developmental and behavioral
outcomes. We will also discuss available evidence from longitudinal studies of preterm children at psychosocial disadvantage and what
lessons can be learned from the bidirectional
impacts of prematurity and poverty. Longitudinal
studies will be evaluated for health, disability,
and community outcome trajectories for children
with prematurity across preschool, middle childhood, adolescent, and young adult epochs. Our
review will highlight important opportunities on
a community level for systematically optimizing
population-based prevention strategies for individuals with the double jeopardy of prematurity
and social adversity and the long-term impact of
failing to optimize these outcomes.
1.1
Approach
Two frameworks will inform our analysis. The
first framework will be the International
Classification of Functioning, Disability, and
Health (ICF) model (World Health Organization
Table 1 Life course impact of prematurity
Weeks gestation
<28
28–31
32–36
37+
Children special
health-care needs
50%
40%
30%
20%
Major neurodevelopmental
disability
20%
15%
10%
5%
Educational supports
50%
40%
25%
15%
Behavioral disorders
20%
15%
10%
5%
Morse et al. (2009), Allen et al. (2011), Stephens and Vohr (2009), B. Vohr (2013), Saigal and Doyle (2008)
M.E. Msall et al.
324
2007), which we will use to comprehensively
describe the diverse outcomes occurring in preterm survivors. This framework goes beyond
dichotomous classification of impairments (e.g.,
cerebral palsy, yes or no; intellectual disability,
yes or no) and instead describes a spectrum of
functioning at body structure and body function
levels. For example, activities in whole-person
tasks include running, reading, and dancing and
participation in children’s roles with peers including being on a team; participating in church, temple, or mosque; or meeting friends for a movie.
The second framework will be the life course
health development (LCHD) model, which holds
that the trajectories of children are influenced by
the dynamic interaction of multiple risk, protective, and promoting factors, especially during
sensitive periods of health development. From
the standpoint of fetal development, due to critical human brain development in the second and
third trimesters, a focus on premature infants
must consider complex maternal, placental, and
fetal dynamic interactions. Likewise, infant, toddler, and childhood periods of development are
indelibly influenced by multilevel, multidirectional, transactional, and long-lasting interactions and critically emphasize the importance of
timing. Using a LCHD framework to analyze the
origins and impact of prematurity and the opportunities to optimize health development outcomes suggest the following considerations:
• Children who are born prematurely are
assumed to be more developmentally vulnerable and are potentially more sensitive to a
wide range and nested array of dynamic interacting influences.
• Because the alterations in evolutionarily influenced and developmentally determined adaptive mechanisms are well documented, lags in
developmental processes, as well as catch-up
and feed-forward processes that are specific to
premature infants, may influence the nature
and dynamic of their health and developmental trajectories.
• Understanding how the caregiving environment of premature infants interacts with
emerging developmental capacities and how
different types of exposures, levels of support,
and adversity influence these emergent developmental trajectories is important if specific
and targeted interventions are to be designed
to modify developmental pathways based on
specific risk profiles to shift the health and
developmental curves for the entire population of premature infants.
• In order to implement a broader approach to
improve the health and developmental outcomes of diverse preterm populations, it is
important to determine what is known about
the special development vulnerabilities of premature infants, how that vulnerability manifests (timing, context, specific risks), and
whether the mechanisms involved are phase
or period specific, modifiable, or one of cumulative risk.
2
Framing Our Inquiry
and Agenda
On a population level, it is important to acknowledge the diversity of both the underlying causes
and life course effects of prematurity and the
gaps in proactive and comprehensive medical,
developmental, and behavioral supports. This is
occurring within the context of recognizing that
current community systems are under-resourced
to systematically audit barriers and facilitators to
home visiting, medical homes, early interventions, parenting supports, and coordinated services for children with special health-care needs
(CSHCN). In particular, we must go from a
crisis-oriented response system for the few with
the severest impairments to an optimization system for all at risk for less than optimal health
development.
To address these systemic needs, the following themes will be highlighted that require
increased research and policy efforts:
• We must better understand the role of and need
to engage mothers and other caregivers in
developmentally optimizing interactions from
the neonatal period through school entry. This
includes how they gain and utilize their knowledge about their child’s health and development, as well as how to optimally serve as their
Life Course Health Development Outcomes After Prematurity: Developing a Community, Clinical…
•
•
•
•
child’s first teacher and advocate. This requires
understanding ways that early parent involvement and developmental activities can be part
of everyday childcare tasks.
We must explore how social environments are
best tailored to maximally support positive
growth, child regulatory behaviors, and developmental competencies. This includes how all
children can access comprehensive preschool
services and how health, developmental, and
behavioral competencies are measured, monitored, and accounted for from birth to kindergarten entry. In keeping with a two-generation
model of optimizing health development,
close attention to caregiver physical and
behavioral health and supports that help vulnerable children access quality childcare and
early child education have the potential to
ensure that children’s social-emotional, communicative, and cognitive competencies are
supported so that children enter kindergarten
healthy and ready to learn.
We must clarify the roles that preconception
maternal, physical, and behavioral health play
on maternal and child vulnerabilities and on
epigenetic programming. This includes a better
understanding of how maternal mental health
stressors (depression, anxiety, isolation, and
violence) increase vulnerability of children
when they do not receive quality early childhood social, learning, and behavioral supports.
We must determine the optimum and appropriate role and function of community outreach strategies that promote parental physical
and behavioral health, child development, and
social competencies.
We should pursue evidence-based strategies to
promote resiliency and positive adult health
trajectories which include independent living,
employment, and family formation while minimizing physical risk factors that increase
early-onset adult chronic diseases (e.g., cardiac, pulmonary, mental illness, substance
abuse) (Figs. 1 and 2).
Our cases illustrate how the interaction of positive home, preschool, and educational supports
can increase thriving and reduce adverse longterm adult health conditions.
3
Risk Factors for Poor Birth
Outcomes
3.1
Social Risks
325
An important determinant of preterm birth is
social risk. Social risk factors include suboptimal
home and community environments. Poverty,
domestic violence, drug addiction, crime, hunger, and poor-quality housing are some of the
features of social risks (Holzmann and Jørgensen
2001). Mothers who live in adverse environments
often experience multiple stressors and are prone
to nutritional deficiency, suboptimal prenatal
care, single parenthood, and frequent tobacco
and alcohol use compared to mothers from nonpoor backgrounds (Jiang 2015). Several studies
have also shown that rates of marijuana, cocaine,
tobacco, and alcohol use are higher for women
who are unmarried, unemployed, and have less
than a college education, indicating that substance abuse and poverty are closely related
(Huston 1991). It has been suggested that the
prevalence of substance abuse, illicit drug use,
and smoking among women from impoverished
or low SES background is largely due to the sense
of helplessness, low self-esteem, difficulties coping with stress, and pressure from coping with
difficult financial situations in everyday living
(Huston 1991; Weitzman et al. 2002).
When mothers receive late prenatal care (or
not at all), the opportunity to identify and intervene on maternal reproductive complications or
health problems that jeopardize fetal growth is
limited. Also, late or no prenatal care decreases
the chances of maternal access to educational and
support services (such as counseling, community
health, and education services) (The March of
Dimes Data Book for Policy Makers 2012).
3.2
Race and Social Disadvantage
Race in the United States is closely related to
SES; thus, it is not surprising to see racial discrepancies in preterm births (Table 2). AfricanAmerican infants are more than 1.5 times likely
than whites to be born preterm and 2.5 times
likely to be very premature than their white peers
M.E. Msall et al.
326
Body Function & Structure
- 3rd grade: diagnosed with
verbal learning
disability/ADHD
- Exercise-induced asthma,
controlled well with inhaler
- Long-acting stimulant
medication for ADHD
Activities
- Challenges with
impulsivity in preschool
- Challenges with reading
and attentiveness in early
grade school
- Excelled in math
Environmental Factors
- Special education pull-out
services for reading in early
grade school
- Tutoring and subjectspecific education supports
in high school
Participation
- Graduated from college
with mathematics degree
- Will attend graduate
computer science
engineering program
Personal Factors
- At birth, parents married,
mom had law degree
- Parents focused on
positive successes in high
school
Fig. 1 Case 1. James was born late preterm at 34 weeks
of gestation due to preeclampsia. His parents were married and mother completed law school. She had some
challenges with impulsivity in preschool, and reported
significant job-related stressors during pregnancy. The
immediate newborn period was complicated by immature
lungs leading to respiratory distress syndrome; however,
ultimately James was discharged at 38 weeks of gestational age without a need for oxygen in the home. He was
enrolled in full-day daycare and preschool since the age
of 2 years. James was found to have some challenges
with impulsivity in preschool, which his parents
addressed with occupational and behavioral therapies. He
entered kindergarten without an Individualized Education
Program and in early grade school was found to struggle
with reading and inattentiveness. He received a formal
diagnosis of verbal learning disability and ADHD in third
grade. James received special education pull-out services
for reading and language arts and was starting on a longacting stimulant medication for ADHD with the guidance
of a Developmental and Behavioral Pediatrician. James
received tutoring and subject-specific special education
supports throughout high school. His parents focused on
his positive successes, such as his strong performance in
math. James went to college and pursued a math and
engineering program. His adult health is complicated by
exercise-induced asthma, well-controlled on an inhaler
(The March of Dimes Data Book for Policy
Makers 2012). These data on preterm birth rates
correlate with disparities in wealth distribution,
with African-American families experiencing the
lowest 3-year average median income (2003–
2005) among US racial groups (Income, Poverty,
and Health Insurance Coverage in the United
States: 2005–2006). These data on higher rates of
prematurity in women experiencing social disadvantage from poverty and minority status also
hold across both developing and developed
countries. The role of maternal health, educational, behavioral, and social competencies during her own childhood and how they impact on
her child’s health and development require systematically measuring both maternal and child
allostatic load. These data can help understand
what factors promote positive adaptations.
4
Prematurity and
Developmental Outcomes
Prematurely born infants have long-term vulnerabilities on multiple outcomes, including physical and developmental health, behavioral and
adaptive well-being, as well as social functioning. Over the past decade, much has been learned
Life Course Health Development Outcomes After Prematurity: Developing a Community, Clinical…
Body Function & Structure
- Ages 2-4:
asthma exacerbations
- Adult: obesity,
asthma, and depression
-
Activities
- 2 years: neurotypical
performance on Bayley
Motor and Cognitive Skills
- Repeated kindergarten:
poor regulatory behaviors
- Continued to struggle in
school
Environmental Factors
- Inconsistent home daycare
enrollment
- Exposure to secondary
smoke, household mold,
cockroaches: asthma
complications
- IEP not until 5 years old
327
Participation
- Dropped out of high
school junior year
- Unemployed
As Adult: No health
insurance
Self-Medicating with
alcohol and street drugs.
Personal Factors
- At birth, mom unmarried,
had not finished high school
- Mom had to work 2 jobs
to care for 3 kids
- Child home life dominated
by TV
Fig. 2 Case 2. Michael was born at 28 weeks due to preeclampsia and preterm labor. His mother was unmarried and
did not finish high school. His NICU course was complicated by intubation for the first 4 weeks of life, but he did not
experience additional medical complications and was able
to be slowly transitioned to room air and oral feedings. At
2 years of age, he tested within age appropriate range on the
Bayley Scales of Toddler Development. Between ages 2 and
4, he experienced asthma exacerbations complicated by
environmental exposures to secondary tobacco smoke,
household mold, and cockroach infestation and was hospitalized about 2–3 times a year. Until the age of 5 years,
Michael was enrolled in inconsistent home daycare programs and did not receive early intervention services
because he was considered less than 30% delayed and
thereby was deemed not eligible for services. He enrolled in
kindergarten at age 5 years. However, he had to repeat kindergarten due to poor regulatory behaviors, which interfered
with learning. Throughout his early childhood education, he
continued to struggle in school. His home life was largely
dominated by television watching while his mom worked
two jobs to help care for him and his two siblings. In school
he was supported by an Individual Education Plan, but did
not access pharmacotherapies for ADHD. He dropped out
of high school in his junior year. His adult health is complicated by obesity, asthma, and depression. Because of unemployment, he does not have health insurance. His state has
not expanded Medicaid access
in the management of preterm infants and the
interventions required to stabilize their immature
organ system functioning.
4.1
Table 2 Rate (%) of preterm birth by maternal race and
trimester of first prenatal visit
Trimester of
first prenatal
visit
First
Second
Third
None
NonHispanic
black
14.7
17.6
16.0
33.4
Lang and Iams (2009)
NonHispanic
white
8.3
10.2
10.0
21.7
Hispanic
9.7
10.0
10.0
19.8
Cerebral Palsy
and Neurosensory
Impairment
Table 3 highlights rates of CP and neurodevelopmental disabilities in early childhood among
extremely preterm cohorts born into the medical
era, which had both prenatal maternal corticosteroid and surfactant replacement interventions
available. These studies demonstrate that, despite
common misconceptions, the overwhelming
majority of children who survive extreme prematurity do not experience CP. These data
M.E. Msall et al.
328
Table 3 Major neurodevelopmental disabilities at age 2 years among survivors of ELBW in the 1990s
Study
Schmidt et al.
(1996–1998)
Mikkola et al.
(1996–1998)
Vohr et al.
(1993–1998)
Wood et al. (1995)
Doyle et al. (1997)
Mestan et al.
(1998–2001)
Shankaren et al.
(1993–1999)
Wilson-Costello et al.
(1990–1998)
Fily et al. (1997)
Sample
N = 944
500–999 g
N = 206
<1000 g
N = 2291
22–26 weeks
N = 1494
27–32 weeks GA
N = 283
22–25 weeks GA
N = 170
500–999 g
N = 138
27.4 weeks GA
N = 246; ≤750 g
≤24 weeks; APl ≤3
N = 417
500–999 g
N = 545
<33 weeks GA
% CP
12
% DD
27
% HI
2
% VI
2
14
9
4
2.6
19
30
2
2
11.6
26
1
0.7
16
30
2
2
11
22
1.8
2.4
9.5
27.3
0.8
1.5
30
46
5
5
14
26
7
1
9
4.7
0.8
0.2
DD developmental disability as defined as mental developmental index < 70, HI hearing impairment, VI visual
impairment
AP1 Apgar 1 min
GA gestational age
complement population registries, which dramatically highlight the increased rate of CP among
preterm survivors with decreasing gestational
age. In Sweden, EPI have rates of CP at 71 per
1000, VPI 40 per 1000, and LPI (32–36 weeks)
6.4 per 1000, compared to term births at 1.1 per
1000 (Himmelmann and Uvebrant 2014).
Although there are many types of CP with
varying degrees of difficulty with motor control
and higher cortical function, the two most common types among preterm survivors are spastic
diplegia and spastic hemiplegia. These CP syndromes affect the motor control of lower extremities (diplegia) or one side of the body (hemiplegia)
and increase risks for communicative, perceptual, learning, and attention disorders (Allen
et al. 2011). However, an examination of these
broader outcomes for recent VPI and EPI cohorts
at ages 5–8 years has not occurred in large
United States regional or multicenter cohorts.
Furthermore, functional measures of gross motor,
manual ability, and communication activities
offer more detailed descriptions of performance
in everyday activities than topographical classifications of CP.
Over the past decade, there has been an
increased appreciation of a wider range of motor
challenges that have been characterized as developmental coordination disorders. A developmental coordination disorder is defined by acquisition
and execution of coordinated motor skills below
what would be expected at a given chronologic
age. Difficulties are manifested as clumsiness,
slowness, and inaccuracy of performance of
motor skills. These motor skill deficits persistently interfere with activities of daily living
appropriate to the chronologic age (Diagnostic
and Statistical Manual of Mental Disorders:
DSM-5 2013). A systematic analysis of 14 studies of children <33 weeks of gestation and
<1501 g demonstrated 41% had mild developmental coordination disorder (characterized by
motor standard scores 1–2 standard deviations
below the mean) and 19% had moderate developmental coordination disorder (≥2 standard deviations below the mean) (Williams et al. 2010).
Life Course Health Development Outcomes After Prematurity: Developing a Community, Clinical…
The second column in Table 3 reviews rates of
developmental disability. These studies demonstrated rates of cognitive disability ranging
between 9 and 46% among those EPI and 5%
when VPI and EPI were combined. It must be
emphasized that their early cognitive disabilities
do not fully capture long-term educational
impact.
Among ELBW survivors, there are high rates
of neurosensory impairments. Severe hearing
impairment ranges from 1% to 7%. This is also
substantially higher than the 1 per 1000 rate in
term infants. Another neurodevelopmental disability after prematurity is visual loss or blindness caused by retinopathy of prematurity (ROP).
This disorder primarily affects premature infants
weighing 1250 g or less and those born before
31 weeks of gestation. ROP in the current era of
neonatology reaches its severest stages in those
who are most immature and medically fragile.
Severe visual impairment occurs in 1–2% of
<27-week preemies, 20–40 times higher than in
term infants. In a multicenter study on cryosurgery for ROP, increased severity of ROP was
linked to motor, self-care, and communicative
disability at kindergarten entry (Msall et al.
2000). Children with severe ROP, but with favorable visual acuity, had a motor disability rate of
5% compared to 43% of children with severe
ROP and unfavorable visual acuity (eyesight
worse than 6.4 cycles per degree on Teller Cards
includes legal and total blindness). In this cohort,
neonatal risk factors for severe disability involving multiple motor, self-care, and communicative
domains included severe ROP, gestational
age <27 weeks, birth weight <750 g, and poverty
as reflected by the absence of private health
insurance. A protective factor associated with a
significant risk reduction for severe disability
was African-American race. In middle childhood, children with severe ROP had substantial
differences in cognitive and educational outcomes (Msall et al. 2004). Children with severe
ROP and unfavorable visual status had a 3 in 5
chance of ID (57%) and a 3 in 5 chance of needing special education services. Children with
severe ROP and favorable visual skills had a 1 in
5 chance of ID (22%) and a 1 in 4 chance of need-
329
ing special education. More than 4 in 5 children
with unfavorable vision (84%) but less than half
of children with favorable vision (48%) were
below grade level in school performance. Lower
socioeconomic status (SES) and minority status
were associated with lower-grade performance
and utilization of special education services
across both visual outcome groups. Table 3 also
highlights that over 95% of extremely preterm
survivors do not experience blindness.
4.2
Cognitive Outcomes
The assessment of cognitive outcomes can focus
on three related domains: intelligence testing,
achievement testing, and neuropsychological
testing. The latter includes evaluation of specific
higher cortical processes such as executive function, working memory, and information processing. Assessment of toddlers and young children
combines precursors of verbal and nonverbal
intelligence and cognitive processes (e.g., object
permanency, symbolic play), whereas schoolage assessment and beyond require more complex testing to assess problem-solving, literacy,
and numeracy. There is increased recognition
that Bayley II and III scores <70 predict complex
cognitive and learning challenges among schoolage children. However, among children with
2-year Bayley scores of 71–84 (1–2 standard
deviations below the mean), they may be
uniquely vulnerable to the lack of quality early
childhood education and preschool experiences,
especially in socially disadvantaged families
(Patrianakos-Hoobler et al. 2009). Therefore,
one needs to be cautious in using developmental
assessments in the first 2 years of life to fully
assess the spectrum of cognitive and learning
disorders in all children, not only those preterm
and growing up in vulnerable circumstances.
The limitations in Bayley assessments are due to
the dynamic processes of higher cortical functioning in childhood. Additionally, the assessment of
developmental delay is intertwined with perceptual and sensory skills as well as early learning
experiences and neuroplasticity. Learning,
communication, and social challenges may not
330
be apparent until there is increased complexity
of tasks for academic skills and behavioral/
attention regulatory capacity required by school
environments.
In order to address higher-level skills at school
entry, one strategy is to examine children’s status
at kindergarten entry. EPI cohorts born in the
1980s from Hamilton, Ontario, Melbourne,
Australia, Buffalo, New York, and Chicago,
Illinois, have demonstrated that 44–56% require
special education resources and 21–29% have
major neurodevelopmental impairments (Baek
et al. 2002; Vohr and Msall 1997). These studies
indicate that, in addition to planning for major
developmental disabilities, resources are required
to ensure success with peers in the classroom.
Gross and colleagues followed infants born
very preterm and found that 41% of preterm
infants were performing at grade level versus
70% of term children. These preterm children
were more likely to receive special education services and three times as likely to be diagnosed
with learning disabilities. In this cohort, parental
marital status and educational attainment were
significantly related to educational outcomes
(Gross et al. 2001). Nearly three times as many
preterm children achieved grade-level performance if parents were married as compared to
children from single-parent homes. Thus, preterm survivors with limited family resources are
further disadvantaged and vulnerable.
In Cleveland, Litt and colleagues prospectively followed 219 surviving ELBW children
born between 1992 and 1995 through middle
childhood and adolescence. Surviving children
had a mean birth weight of 815 g, a mean GA of
26.4 weeks; and almost 1 in 5 was from multiple
birth gestations. Neonatal morbidities included
bronchopulmonary dysplasia in 41%, sepsis in
29%, and sonographic parenchymal brain injury
in 24% (IVH3–IVH4/PVL by cranial ultrasound).
Importantly, 115 term controls from the extremely
preterm survivor’s community classroom were
assessed at 14 years. Both the extremely preterm
survivors and a prospectively recruited term control group underwent a comprehensive neuropsychological battery that included reading and
mathematics and neuropsychological skills of
M.E. Msall et al.
processing speed, attention, visual memory,
working memory, and planning. In addition,
social capital as determined by maternal education and median household income as well as
gender and minority status was used as covariates when analyzing predictors of high school
functioning (Litt et al. 2012).
Though major neurosensory disabilities
occurred in approximately 1 in 6 of ELBW survivors (CP 15%, blindness in 0.5%, and hearing loss
requiring amplification in 1.7%), a far greater number of children experienced ID and cognitive
impairments. More than 1 in 6 (18%) had ID
(IQ <70), a 4.5-fold greater risk than for term peers.
More than 1 in 3 (37%) were cognitively impaired
(IQ 71–84), a 2.3-fold greater risk than for term
peers. Most importantly, almost 1 in 2 (49%) of
ELBW survivors required an individualized educational plan, a fivefold higher rate than term peers.
The educational resources required for managing
these highly prevalent disorders would require an
additional $50,000 per child during their high
school years for tutoring, smaller class size, and
specific curriculum modifications (Msall 2012).
Of concern were the achievement challenges
of extremely preterm survivors who experienced
socioeconomic adversity. These children not only
demonstrated a 6.6-point lower IQ but had standard scores that were 10 points lower in reading
and 8 points lower in mathematics. Importantly,
both executive function and visual memory challenges were associated with academic struggles
in reading and mathematics (Johnson et al. 2009,
2011; Simms et al. 2015; Wolke et al. 2015).
4.2.1 What Does This Mean?
The first lesson brings optimism: the large
majority of ELBW survivors (83%) are free of
neurosensory disability. Thus, unprecedented
survival without major neurosensory disability
can be expected. However, despite antenatal corticosteroids, surfactant replacement, improved
nutrition, and infection control, the rates of CP
are over 100-fold greater than for term infants.
Thus, ongoing efforts in neuroprotection remain
a scientific, clinical, and political priority.
The second lesson is sobering: the majority
of ELBW survivors experience ongoing and
Life Course Health Development Outcomes After Prematurity: Developing a Community, Clinical…
serious challenges in learning, coordination,
and executive function. These children benefit
from quality and comprehensive interventions
and special educational accommodations with
continued review of how these services promote
literacy, numeracy, and life skills. Litt and colleagues’ outcomes suggest that evidence-based
psychoeducational management strategies for
executive function and specific learning disorders may be key areas for maintaining children
on positive developmental trajectories (Litt
et al. 2012). An important challenge in the
upcoming decade is to comprehensively implement, at a population level, appropriately targeted
developmental
and
educational
optimization strategies for all preterm survivors.
With more developmentally appropriate individual- and population-level supports, their neurodevelopmental risks that currently result in
costly and adverse long-term physical, behavioral, educational, and social health outcomes
could be dramatically reduced.
4.2.2
Behavioral/Emotional
Disorders
Behavioral and emotional disorders are more
prevalent among children born premature compared to their term birth peers. Meta-analysis of
behavioral outcomes by Bhutta and colleagues
revealed that children born preterm have a 2.6fold risk for developing attention-deficit hyperactivity disorder (ADHD) during school age
(Bhutta et al. 2002). These children are also at
high risk for externalization problems such as
aggression, oppositionality, and disruptive
behaviors, which also are major obstacles in
establishing friendships or other social relationships (e.g., lack of patience in waiting for their
turn in group play) (Farooqi et al. 2007; Saigal
et al. 2003). In these studies, birth weight, family
function, gender, and SES predicted the behavioral adjustment of adolescents who survived
ELBW status in the 1980s or the middle childhood behavioral and childhood behavioral and
social adjustment of children who survived
23–25 weeks of gestation in the 1990s.
An important discovery occurred over the
past decade whereby it was noted that very and
331
extremely preterm infants were at high risk for
delays in social-emotional behavioral regulation
that manifested as hyperactivity, impulsivity,
and difficulty with social competencies in the
preschool years (Jones et al. 2013). Importantly,
those children receiving appropriate quality preschool education experiences were more ready
for kindergarten both cognitively and behaviorally than those who did not access these services
(Arpi and Ferrari 2013; Treyvaud et al. 2013).
There has also been increased recognition that
late preterm survivors are at risk for regulatory
and social challenges. Recent comparative
investigations of toddler outcomes for LPI versus other premature infants demonstrate more
externalizing, oppositional, and aggressive
behaviors, suggesting a unique vulnerability for
LPIs (Shah et al. 2013). Infants more prone to
distress who also experienced more critical parenting styles in infancy were more likely to
demonstrate externalizing behaviors at age 3,
suggesting that characteristics of both infants
and parents influence preterm vulnerability
(Poehlmann et al. 2012).
There has also been an increased awareness of
the vulnerability of adolescent and adult survivors of very and extremely preterm birth to
increased rates of behavior and mental health disorders (Gardner et al. 2004). This is highlighted
in Table 4. An important review by Johnson and
Marlow emphasized a life course health development framework and highlighted the increased
risk of attention, socio-communicative (including autism spectrum disorder), and emotional difficulties among extremely preterm survivors
(Johnson et al. 2011). Importantly they highlighted rates of autism spectrum disorder (ASD)
as high as 1 in 12 EPI survivors as they enter adolescence (Johnson et al. 2011). A New Jersey
cohort found a rate of 1 in 20 for ASD in moderate LBW at school exit (Pinto-Martin et al. 2011).
Most recently, in a large US cohort of 889 EPI
survivors born in 2002–2004, Joseph and colleagues found a rate of ASD in males at age 10 of
9% and in females of 5%. Rates of ASD were
15% at 23–24 weeks of gestation, 6.5% at
25–26 weeks of gestation, and 3.4% at 27 weeks
of gestation (Joseph et al. 2016).
M.E. Msall et al.
332
Table 4 Adolescent and adult behavioral health outcomes
Authors
Botting et al. (1997)
Cohort
1980–1983 Liverpool,
England
138 VLBW and 108 matched
controls
Age at assessment
12 years
Dahl et al. (2006)
1978–1989
Norway
99 VLBW
13–18 years
Grunau et al. (2004)
1981–1986
British Columbia
79 <800 g vs. 31 term
17 years
Jong et al. (2012)
Meta-analysis of moderate
and late preterm
(32–36 weeks)
28 papers reviewed
Meta-analysis of LBW and
schizophrenia prevalence;
750 with schizophrenia and
29,000 control subjects
Israel
90 VLBW and 90 NBW
Not reported
(meta-analysis)
1977–1982
Ontario, Canada
166 ELBW and 145 NBW
Ontario, Canada
149 ELBW and 133 NBW
22–25 years
Kunugi et al. (2001)
Levy-Shiff et al.
(1994)
Saigal et al. (2006)
Saigal et al. (2007)
4.2.3 What Does This Mean?
The current understanding of the vulnerabilities
of the preterm infant brain to challenges in coordination, learning, attention, and social skills
offers opportunities for understanding cumulative
risk and protective strategies linked to biomarkers
and neurometrics. The goal of understanding what
supports are needed to protect the vulnerable preterm infant brain for learning regulatory, attention,
executive function, and social competencies will
require increased attention to how active health
Developmental outcome
1. Any psychiatric disorder:
28% VLBW vs. 9% controls
had any psychiatric disorder
2. ADHD: 23% VLBW vs. 6%
controls
1. VLBW adolescents report
less externalizing behaviors
than NBW adolescents
2. Parents of VLBW
adolescents report more
externalizing behaviors and
emotional problems than NBW
adolescents.
1. No differences for focus and
attention
2. Significantly more parental
reported internalizing,
externalizing, and problem
behaviors
30% higher psychiatric
disorders
Not reported
(meta-analysis)
LBW prevalence: 9.5% of
schizophrenics and 3.9% of
controls
13–14 years
Significantly increased
hyperactive behavior among
VLBW. However paternal
involvement was as predictive
as birth weight for
hyperactivity in childhood
1.3% of ELBW had autism vs.
0% controls
23 years
14.1% ELBW vs. 6% on
antidepressants
and developmental experiences shape these developmental processes and what proactive informed
interventions support success academically,
behaviorally, and socially. Revolutionary strategies in developmental neuroscience will help
inform an evidence-based approach to early
identification and intervention so that children
stay on track in development. In many ways,
children with prematurity can help inform a shift
from categorizing children’s delays as leading to
low learning expectations. We can examine how
Life Course Health Development Outcomes After Prematurity: Developing a Community, Clinical…
attention, regulatory, and social skills can be
modified and lead to pathways of resilience in
academic achievement and social-emotional
behavioral competencies.
5
Chronic and Acute Stress
Recent studies have produced a large body of evidence that stress is significantly correlated with
the occurrence of adverse birth outcomes, including prematurity and low birth weight (GraignicPhilippe et al. 2014). Prenatal stressors can
include physical and psychological stress, infections, nutritional deficiencies, and drug exposures
(Bock et al. 2015). In a prospective study,
Killingworth-Rein et al. combined three variables
into a single stress indicator comprised of state
anxiety, perceived chronic stress, and life event
distress. Higher scores on this aggregated factor
predicted a shortened gestation and lower birth
weight (Rini et al. 1999). Torche and Kleinhaus
(2012) found that females who were exposed to
earthquakes in early pregnancy had higher rates of
preterm delivery (Torche and Kleinhaus 2012). In
a study in the southern Israeli town of Sderot
(4 km away from the Gaza Strip), Wainstock et al.
hypothesized that the frequent exposure to rocket
attacks in this community would be associated
with adverse birth outcomes when compared to
Kiryat Gat, a similar town without constant rocket
attack exposure. Researchers found that women
living in Sderot gave birth to more babies born
preterm and at low birth weight, confirming the
hypothesis that stress in pregnancy increases
adverse birth outcomes (Wainstock et al. 2014).
A number of other studies have demonstrated
an association between prenatal stress and
adverse pregnancy and birth outcomes, including
effects on birth weight, gestational age, and preterm birth (Bock et al. 2015). Chronic stress and
acute stress may exert different effects on fetal
development and perinatal outcomes. Several
authors have reported lower birth weight and
shortened gestation in children born to mothers
exposed to chronic stress compared to acute
stress (Chrousos and Gold 1992; Rini et al. 1999;
Sable and Wilkinson 2000; Wadhwa et al. 1996).
333
Likewise, there is increasing evidence that
exposure to forms of toxic stress in early life alters
stress responses in adulthood. A case-controlled
longitudinal study of preterm and term infants with
and without medical complications demonstrated
dysregulation of the hypothalamic-pituitaryadrenal axis in adult survivors of prematurity, particularly survivors also exposed to socioeconomic
disadvantage (Winchester et al. 2016).
There is also a strong association between specific early pregnancy events, such as complications in the first trimester, and increased risk of
preterm delivery and very preterm delivery (van
Oppenraaij et al. 2009). An increased risk of low
birth weight and very low birth weight was associated with first-trimester bleeding in a large prospective study (M. A. Williams et al. 1991). These
adverse infant outcomes were associated with
threatened miscarriage in the first trimester in
another large retrospective study (Wijesiriwardana
et al. 2006).
6
Epigenetic Mechanisms
Although the mechanisms of prenatal stress are
not precisely known, it has been proposed that
the hypothalamic-pituitary-adrenal (HPA) axis
and sympathetic nervous system stress responses
could lead to complications in pregnant women
during pregnancy and delivery (GraignicPhilippe et al. 2014). In particular, McLean et al.
(1995) suggest that high levels of neurohormones
in the HPA axis as well as placental neurohormones could induce prematurity. A growing body
of evidence suggests that epigenetic mechanisms
induce transient and permanent alterations in
gene function, resulting in altered developmental
processes in the brain that result in long-term
changes in emotional and cognitive behaviors
(Bock et al. 2015).
The main function of the epigenome is to regulate gene transcription and compaction of DNA
into the cell nucleus through mechanisms such as
DNA methylation and hydroxymethylation,
histone modifications, ATP-dependent chromatin
remodeling, and noncoding RNAs (Provencal
and Binder 2015). Prenatal stress has been shown
M.E. Msall et al.
334
to reduce the expression and activity of the
enzyme 11-β-hydroxysteroid dehydrogenase
type 2, an enzyme that converts glucocorticoids
into inactive metabolites (Bock et al. 2015). An
alteration in placental permeability may lead to
excess exposure to maternal stress hormones,
thereby promoting developmental dysregulation.
Maternal intake of steroid hormones (Marciniak
et al. 2011), maternal depression (O’Connor et al.
2014), and maternal anxiety (Kane et al. 2014)
have been shown to alter maternal glucocorticoid
levels, with a high correlation between maternal
and fetal plasma glucocorticoids (Provencal and
Binder 2015). In addition to affecting birth outcomes, stressful experiences in utero or during
early life may also increase the risk of neurodevelopmental and behavioral disorders later in life
due to alterations in epigenetic regulation
(Babenko et al. 2015).
Pregnant mothers who have anxiety disorders,
post-traumatic stress disorder (PTSD), depressive disorders, or major psychoses are at higher
risk of adverse birth outcomes including preterm
birth, low birth weight, and small-for-gestationalage infants. Mothers with bipolar disorder are at
twice the risk for these adverse outcomes
(MacCabe et al. 2007). Importantly, pregnant
mothers with both PTSD and major depressive
episode have four times the risk of having a preterm birth (Yonkers et al. 2014).
These risks occur because the brain and placenta are closely linked by a number of peptides
and proteins, including oxytocin, somatostatin,
neurotensin, encephalin, cortisol, insulin-like
growth factor 1, vascular endothelial growth factor, and cyclic AMP response element binding
factor. Individuals with mental illness often have
a tendency toward coagulation and low activity
of tissue plasminogen activator, potentially contributing to placental insufficiency that may lead
to increased exposure of the fetus to maternal
hormones. Hyperemesis gravidarum, a condition
more common in women with eating disorders
and anxiety than in controls, is another risk factor for pregnancy complications which may
increase the risk of miscarriage, low birth weight
infants, and preterm infants (Hoirisch-Clapauch
et al. 2015).
7
Prenatal Stress Reduction
Strategies
Intervention programs aimed at reducing stress
should be considered to potentially lower the
rates of unfavorable pregnancy outcomes
(Wainstock et al. 2014). Feinberg et al. examined
the effects of family foundation (FF), a transition
to parenthood program, through a randomized
control study of 148 expectant mothers.
Researchers showed that the intervention significantly reduced the negative impact of maternal
cortisol on birth weight, gestational age, and days
in hospital in infants, thereby decreasing the risk
for adverse birth outcomes (Feinberg et al. 2015).
Follow-up at age 5–7 years demonstrated
improved outcomes in behavior regulation and
children’s school adjustment (Feinberg et al.
2014). It is important to recognize the extent to
which a child is affected by a mother’s stress, and
mental health heavily depends on moderating
factors that include quality of parenting, social
support, and the length and severity of the parental disorder (Howard et al. 2014). In this respect,
early identification and interventions are essential (Stein et al. 2014). A recent report estimated
that the long-term economic costs of perinatal
mental disorders for each annual cohort exceed 8
billion euros in the UK alone (Bauer et al. 2014).
Early interventions for perinatal mental disorders
could produce great economic benefits and
improve maternal and child physical and mental
health (Howard et al. 2014) via impacting on
child’s regulatory behaviors and learning
opportunities.
8
The Impact of Fetal
Environment
and Prematurity on Adult
Health Outcomes
Exposure to an unfavorable environment in early
life is closely associated with an increased tendency to develop adult disease. In 1992, David
Barker hypothesized that the period of pregnancy and the intrauterine environment have a
profound impact on risk of developing diseases
Life Course Health Development Outcomes After Prematurity: Developing a Community, Clinical…
like hypertension; diabetes; cardiac, pulmonary,
and renal diseases; and mental illness (Capra
et al. 2013). Intrauterine environment and early
postnatal life are now generally accepted as
important factors that may lead to increased risk
for disease in adulthood (Hofman et al. 2004). In
particular, low birth weight, a marker of poor
fetal growth, is linked to vascular disease, hypertension, obesity, and insulin resistance (Calkins
and Devaskar 2011).
Although several studies have reported an
association between low birth weight and poor
adult health outcomes, it remains unclear whether
this association exists for children with poor fetal
growth born small for gestational age as well as
premature babies whose weight is appropriate for
gestational age. In a retrospective study, Kaijser
et al. identified subjects born preterm or with a
low birth weight at four major delivery units in
Sweden from 1925 to 1945. Researchers found
that future risk for ischemic heart disease was
most closely mediated by fetal growth restriction
instead of preterm birth without growth restriction. Of note, these results came from a 1925 to
1949 birth cohort, a period when infants did not
benefit from modern neonatal care. During more
recent decades, infant survival has improved substantially for children born very preterm. It may
not be appropriate, therefore, to generalize these
results to preterm and growth-restricted infants
being born today (Kaijser et al. 2008).
Babies born prematurely, whether or not they
have intrauterine growth restriction (IUGR), are
also at risk as adults for poor cardiovascular
health outcomes (Irving et al. 2000). In a study of
mothers of 72 LBW and 54 NBW infants born in
Edinburgh between November 1973 and
February 1975, LBW premature babies had
higher adult blood pressure and fasting plasma
glucose than NBW controls born at term. Babies
born prematurely also had trends for an adverse
metabolic profile and were at risk for hypertension and hyperglycemia as adults. Infants with
IUGR were not measurably more disadvantaged
than preterm infants with birth weight appropriate for gestational age (Irving et al. 2000).
A prospective follow-up study of 458 adults at
age 30 in New Zealand also aimed to distinguish
335
the relative contributions of gestation length and
fetal growth to cardiovascular risk factors in
adulthood. Preterm birth, rather than poor fetal
growth, was shown to be the major determinant
of the association between early environment and
adult health outcomes; adults born preterm had
increased systolic blood pressure and insulin
resistance at age 30. Birth weight, independent of
gestational age, was not associated with increased
adult systolic blood pressure and insulin resistance, suggesting that length of gestation may be
the major contribution to cardiovascular risk in
adulthood (Dalziel et al. 2007).
Adult-onset insulin resistance, an early marker
for type 2 diabetes, is also associated with premature birth (Hofman et al. 2004). Exposure to an
adverse environment may be responsible for this
reduction to insulin sensitivity, whether during
intrauterine life in infants small for gestational
age or a primarily adverse postnatal environment
in premature infants (Hofman et al. 2004).
For the most part, mechanisms to explain the
associations between intrauterine and postnatal
environments and adult health outcomes are
unknown. It is hypothesized that alterations to
the HPA axis may explain the link between low
birth weight and later increased blood pressure
(Dalziel et al. 2007). Furthermore, it is theorized
that earlier maturation of organ system exposure
to nutritional, metabolic, hormonal, sensory, and
respiratory environments at earlier time points in
premature babies may also lead to abnormal
development of organ systems involved in cardiovascular health (Curhan et al. 1996). Preterm
infants and small-for-gestational-age infants
often experience a period of “catchup growth” in
the postnatal period, which may influence
changes in their metabolism that are related to
increased risk for these diseases (Smith and
Ryckman 2015).
Increasing preterm infant survival has critical
implications on the burden of adult diseases such
as diabetes and cardiovascular disease. In fact,
the historic trends in preterm survival and birth
may be playing a critical, yet underappreciated,
role in the population trend toward increasing
prevalence of diabetes and cardiovascular disease (Dalziel et al. 2007).
M.E. Msall et al.
336
9
Postnatal and Social Risk
Despite knowledge that supports that intervention
in early childhood can positively impact disability
trajectories, large gaps in services exist and disproportionally impact disadvantaged families. A
1997–2000 study looked at access to early intervention services among a population of infants
aged birth to 3 years. Even in the highest risk neonates, access to community and early intervention
supports was problematic and fragmented at best
(>40% not enrolled in early intervention). Those
toddlers with higher rates of disability were more
likely to receive more services, yet a significant
unmet need for services was documented among
milder cases (Hintz et al. 2008).
Second, a Chicago-based cohort was followed
after NICU discharge and demonstrated that less
than 60% of VLBW infants living in extreme
poverty (<50% federal poverty level) were
receiving early intervention (EI) services despite
having access to a medical home and legal advocacy. Of the 415 infants deemed not automatically eligible by EI, 95% had child and family
impairments (e.g., gastrostomy, feeding delays,
income <$10 K per year, parental mental or
developmental disorder) that met EI inclusion
criteria. This data confirms that a substantial
number of infants with multiple medical and
social risks do not receive ongoing developmental surveillance or early intervention services
(Weiss et al. 2007).
Third, in a recent analysis of the Early
Childhood Longitudinal Survey-Birth Cohort, it
was found that eligibility for Part C varied widely
between states (2–78%), yet the proportion of children receiving services remained consistently low
(1.5–7%). There is significant state-by-state variability between eligibility criteria, yet there is also
a clear national trend of insufficient enrollment for
children with qualifying delays (Rosenberg et al.
2008). Given that in elementary school years
10–20% of children nationally have Individual
Educational Plans, early intervention rates less
than 5% reflect significant missed opportunities
for secondary and tertiary prevention.
Among both VLBW and ELBW survivors,
disproportionate numbers live in communities
with high rates of school dropouts and poorly per-
forming schools. This will result in barriers to
accessing quality early and middle childhood
educational experiences. The combination of
developmental vulnerability due to LBW and preterm birth with social and family distress has
shown to have cumulative impacts (Hille et al.
1994). In an 8–10-year follow-up study of infants
who were the sickest and tiniest and had the most
medical complications in the newborn period,
Msall and colleagues found that not only were
favorable vision and functional motor status at
kindergarten entry associated with significantly
lower rates of special education and below-gradelevel educational achievement, but higher SES
was also associated with positive academic and
developmental outcomes. Factors strongly associated with increased risk for special education services included minority status, poverty, lack of
access to a car, and Supplemental Social Security
Income because of disability and poverty (Msall
et al. 2004).
9.1
Adolescent and Adult
Outcomes
Clinical research to date on the outcomes for preterm infants during adolescence and adulthood
reveals a spectrum of strengths and challenges in
physical and behavioral health, educational
achievement and supports, and community participation. However, the social, educational/vocational, and independent adaptive life skills
required for adulthood are inherently more quantitatively and qualitatively complex than basic
developmental milestones in an infant or school
achievement in middle childhood. A few crude
measures of adult success have been used to
examine adolescent and adult outcomes, but much
work remains to unveil more meaningful multidimensional measures of adult physical and behavioral health, daily functioning, social participation,
family formation, and economic well-being.
Educational attainment is a frequently examined adult outcome in the literature. The studies
highlighted in Table 5 demonstrate lower rates of
academic achievement, high school completion,
and postsecondary education among preterm survivors. Premature survivors show high rates of
Life Course Health Development Outcomes After Prematurity: Developing a Community, Clinical…
337
Table 5 Cognitive, executive function and academic achievement in adolescence and adulthood
Authors
Botting et al. (1998)
Saigal et al. (2000)
Cohort
1980–1983 Liverpool,
England
138 VLBW
108 matched controls
1977–1982
Ontario, Canada
141 ELBW
124 matched controls
Assessment age
12 years
Developmental outcome
1. VLBW lower IQ
2. Lower math and reading
comprehension
12–16 years
1. 28% reported neurosensory
impairments
2. 25% of ELBW vs. 6% repeated a
grade
3. 49% of ELBW vs. 10% required
special education services
4. 22% ELBW required full-time
educational assistance (vs. 0%)
5. Lower mean WISC-R DQ
6. Lower mean WRAT-R
1. No differences for tests of executive
function, verbal memory, attention
2. Preterm group had impaired verbal
fluency
1. 56.1% ELBW vs. 84.6% controls
completed HS
2. 33% vs. 9% required special
education
3. Significant differences in low IQ
(<85)
1. 74% VLBW graduated HS vs. 83%
NBW
2. 30% pursued secondary education vs.
53% NBW
3. 40% repeated grade vs. 27% NBW
4. Scored 1/3 SD lower on WAIS-R
71% vs. 78.6% completed 12 or more
years of school
1979–1980
London, England
<33 weeks; 75
premature and 53 FT
1976–1981
Montreal, Canada
57 ELBW and 44 NBW
14–15 years
Hack et al. (2002)
1977–1979
Cleveland, Ohio
242 VLBW and 233
controls
20 years
Lindstrom et al.
(2009)
1973–1979
Sweden
24–28 weeks vs. FT
1976–1983
Norway
325 preterm (23–
27 weeks) vs. 828,227
FT
1960–1965
Baltimore
226 near-term and 1393
FT
1987–2002 national
registry
Rushe et al. (2001)
Lefebvre et al. (2005)
Moster et al. (2008)
Nomura et al. (2009)
Johns Hopkins
Collaborative
Perinatal Study
Saigal et al. (2006)
1977–1982
Ontario, Canada
166 ELBW and 145
controls
18 years
20–36 years
1. 67.7 preterm vs. 75.4% completed HS
2. 4.4% preterm vs. 0.1% fullterm with
ID
27–33 years
1. Near-term birth associated with lower
adult educational attainment only for
those living below poverty line
2. SGA had no association with
educational attainment
No significant difference in:
1. % graduation from high school
2. Pursuit of postsecondary education
22–25 years
HS high school, SD standard deviation, WAIS-R Wechsler Adult Intelligence Scale-Revised, WISC-R Wechsler
Intelligence Scale for Children-Revised, DQ deviation quotient, WRAT-R Wide Range Achievement Test-Revised, FT
full term
need for special education supports, both in general special education services and with individual assistants, and have higher rates of grade
repetition than their normal birth weight peers
(Saigal et al. 2003). Furthermore, preterm survivors are less likely to graduate high school and
pursue secondary education (Hack et al. 2002;
Moster et al. 2008).
338
However, this outcome is also influenced by
contextual factors. When high school graduation
rates are adjusted for parental SES, there is evidence of mitigation of the difference in educational attainment. In a study by Nomura et al., it
was found that in near-term survivors, only those
living at incomes close to poverty line differed
from full-term peers on educational attainment.
The relationship between cognitive testing in
adulthood and gestational age has also been significantly mitigated by socioeconomic factors
(Ekeus et al. 2010). There is compelling evidence
that the same risk factors disproportionately distributing premature birth to poorer, less educated
mothers are also at play with the long-term outcomes of low birth weight progeny, compounding the disadvantage of the disadvantaged.
IQ outcomes have been repeatedly tested and
show consistently lower scores for ELBW adults
compared to matched peers. However, many
studies are unable to match for socioeconomic
status or parental IQ which are highly influential
mediating factors. Maternal education and
income have been shown consistently to predict
IQ within these cohorts, indeed often with stronger correlation coefficients than LBW status
(Drillien 1961; Drillien et al. 1980). There is evidence that IQ in middle childhood is strongly
predictive of IQ at adolescence, suggesting that,
on this measure, there is neither increasing gap
nor improved catch-up to normal birth weight
peers. Alternatively, one study highlighted a widening IQ gap in later childhood (Botting et al.
1998), which may be due to the complexity of the
involved intellectual task in testing at older ages
or the additional components of social/emotional
intelligence, which are not entirely captured by
younger measures of IQ. Furthermore, studies
that evaluate both educational and professional
attainment do not consistently demonstrate
strong correlation; 50% of preterm adults in the
1983 Dutch cohort who reported poor educational attainment during the school years held
full-time employment as young adults (Hille
et al. 2007). How this translates to US cohorts in
the ongoing economic crisis for those without
college degrees is yet unknown. Other educational outcomes, such as reading and math
M.E. Msall et al.
achievement, have shown significant gaps in core
educational outcomes between VLBW and agematched peers. However given that many term
children in low-income urban or rural communities in the United States also are struggling with
reading, mathematics, science, and social studies
performance, it is difficult to make international
comparisons.
Additional evidence demonstrates the power of
socioeconomic status in modifying the effect of
prematurity; Lefebvre et al. assessed the cognitive
and academic achievement outcomes in early
adulthood of a cohort of 82 ELBW subjects in
Montreal (Lefebvre et al. 2005). There were significant differences between ELBW and NBW
groups in full-scale IQ (94 versus 108), verbal IQ
(93 versus 106), and performance IQ (97 versus
109). However, father’s low socioeconomic status
contributed significantly to the prevalence of
IQ <85 (19% versus 2%, p = 0.012), schooling in
mainstream education with a regular curriculum
for age (36% versus 68%), requirement for special
classes or schools (33% versus 9%), and high
school graduation for those 18 years or older (56%
versus 85%).
Adolescent/adult self-perceptions of health,
quality of life, and overall functioning are important outcomes, which may guide our clinical
understanding and facilitate improved anticipatory guidance to families of premature neonates.
A socioeconomically diverse cohort of adolescents born in 1983–1984 at <801 g (Brown et al.
2003) reported themselves and was reported by
their parents as having lower functional status
than normal birth weight peers; however, overall
these health concerns did not significantly interfere with tasks of daily living. On rating scales of
externalizing and internalizing problems, however, (The Behavioral Assessment System for
Children- BASC), there were no differences
between preterm and comparison groups.
Additionally, using a self-perception profile,
which included domains of scholastic competence, social acceptance, athletic competence,
and global self-worth, responses did not significantly differ. In a similar cohort of adults (born at
weight <1250 g in Zurich), preterm and term
controls did not differ on overall physical health
Life Course Health Development Outcomes After Prematurity: Developing a Community, Clinical…
339
Table 6 Adult outcomes after very preterm birth
Authors
Baumgardt et al.
(2012)
Hack et al. (2007)
Hille et al. (2008)
Moster et al. (2008)
Saigal et al. (2006)
Cohort
1983–1985
Zurich, Switzerland
52 preterm (<1250 g)
75 controls
1977–1979
Cleveland, Ohio
241 VLBW
232 NBW
1983
Netherlands
959 adult survivors of
prematurity (<32 weeks
or VLBW)
1976–1983; Norway
325- 23–27 weeks
1608- 28–30 weeks
6363- 31–33 weeks
31,169- 34–36 weeks
828,227- full term
1977–1982
Ontario, Canada
166 ELBW and 145
controls
Age at assessment
23 years
Developmental outcome
No difference in overall self-reported
quality of life
20 years
1. No difference on self-reported
health satisfaction
2. No difference on self-reported
comfort (physical or emotional)
3. Decreased self-reported resiliency
4. Increased self-reported risk
avoidance
1. 11.4% had moderate/severe
problem with profession
2. ½ of individuals with moderate/
severe problems in education had
full-time employment
1. 10.6% vs. 1.7% receiving
disability pension
2. Lower gestation less likely to have
found life partner
3. Lower gestation less likely to have
children
No significant difference in rates of:
1. Employment
2. Independent living
3. Married/cohabitation
4. Parenthood
19 years
20–36 years
22–25 years
VLBW very low birth weight (<1500 g), ELBW extremely low birth weight (<1000 g), NBW normal birth weight
(>2500 g), CPT Conners’ continuous performance task
or mental health scores on the Short Form 36
Health Survey (Baumgardt et al. 2012) (Table 6).
Adult males who survived prematurity rated
themselves significantly lower than male term
adults on the “physical functioning domain,” but
females did not differ from matched controls in
any subset. Patterns of high risk and protective
health habits varied: the use of marijuana was
significantly lower among adults who survived
prematurity, yet significantly more control adults
practiced sports and more males exercised. The
overall trend appears that adolescents and adult
survivors of prematurity, by and large, consider
themselves similar to normal term survivors on
self-ratings of physiologic and psychological
functioning. Some studies suggest that females
may be more similar to full-term peers than
males, suggesting that males may be somewhat
more vulnerable to the long-term effects of prematurity and LBW. It deserves mentioning that
the discrepancy between self-reported health and
external measures of health has been reported in
other health conditions; overall persons with a
health condition rate the impact of that condition
more positively than health professionals may
surmise (Groot 2000).
Research on long-term consequences of low
birth weight has focused on adolescent and early
adult life – especially the negative impact of low
birth weight on academic achievement, high
school graduation and years of school completion, and college attendance. Low birth weight
has been linked to these outcomes through the
effects of low birth weight on kindergarten preparedness and weak performance in the first few
years of schooling. Poor educational performance in subsequent years is negatively linked
to both poor school readiness and accomplishment on subsequent school performance and the
lingering health effects of low birth weight. The
340
latter often includes not only growth and respiratory
sequelae but increased vulnerability to a spectrum of
neurobehavioral disorders that impact attention and
mood. Importantly, buffers for these disorders
include attention to caregiver mental health and
child behavior health in the context of increasing
school and community successes. While longerterm effects of low birth weight, mediated by
poor educational performance and continued
weak health, are hypothesized, little research has
examined the midlife consequences of low birth
weight and what factors contribute to thriving.
An additional complexity of measuring adolescent and adult outcomes is the shift of health
status reporting from the parent of a premature
survivor to the adolescent or adult himself/herself.
In an adolescent follow-up of 99 very low birth
weight babies born in Norway, there was found to
be a significant discrepancy between adolescent
and parental reporters, with the overall trend
being parents reporting significantly more emotional and behavior problems than the adolescents
reported in themselves (Dahl et al. 2006). In these
later stages of follow-up, investigators must consider the inherent bias in asking parents vs. adolescents to report on health and behavior.
This research limitation can be remedied
through analysis of data from two population
datasets that include information on weight at
birth and current health and well-being – the
Wisconsin Longitudinal Study (WLS) and the
Midlife in the United States: A National Study of
Health and Well-Being (MIDUS). Additionally,
utilizing these cohorts from the United States
may help remedy the current data gap between
well-tracked international cohorts and the dearth
of longitudinal US data. International cohorts,
which have lower rates of single parenthood and
more homogenous racial ethnic and socioeconomic composition as compared to the United
States, dominate the current knowledge base of
long-term outcomes for premature babies.
Extrapolating from their long-term outcomes
likely vastly underestimates the need for social
support throughout the life course in the United
States. Additionally, international countries with
single-payer systems tend to diagnose and provide more consistent social support for persons
M.E. Msall et al.
with disabilities when compared to the fragmented nature of health care in the United States.
The Wisconsin Longitudinal Study (WLS) is
a long-term study of a random sample of 10,317
men and women who graduated from Wisconsin
high school in 1957. The WLS provides an
opportunity to study the life course, intergenerational transfers and relationships, family functioning, physical and mental health and
well-being, and morbidity and mortality from
late adolescence (in 1957) through early geriatric (in 2008). WLS data also cover social background, youthful aspirations, schooling, military
service, labor market experiences, family characteristics and events, social participation, psychological characteristics, and retirement.
Survey data were collected from the original
respondents or their parents in 1957, 1964,
1975, 1992, and 2004 (when they were approximately at age 64 or 65). The WLS includes endof-life data for members of the cohort who have
died. Sibling and twin data for the original
respondents, as well as gene-environment interactions from late adolescent to the retirement
years, will permit models that separate the
effects of family origins from the impact of low
birth weight at retirement age.
The first national representative survey of
Midlife Development in the United States
(MIDUS) was conducted in 1995–1996. MIDUS
is a 1994 national sample survey of 7189 randomly chosen adults ages 25–74 years, as well as
1914 respondents to a separate nationally representative sample of twin pairs. The wide age
range of the sample was intended to permit comparisons on persons in their early adult life to
those in midlife and old age. The survey permits
the assessment of many psychological factors
such as personality traits, sense of control, and
goal commitments and their linkages to marital
status, family structure, socioeconomic standing,
social participation, social support, employment
status, health status, and health-care utilization.
MIDUS respondent follow-ups were conducted
from 2002 to 2006, and there were a third round
of interviews from 2006 to 2011. This dataset,
including sibling data, will permit models that
separate the effects of family origins and early
Life Course Health Development Outcomes After Prematurity: Developing a Community, Clinical…
life experiences from the impact of low birth
weight at retirement age and beyond.
10
Conclusion
The interdependent challenges of prematurity
and socioeconomic risk create numerous hurdles
for achieving optimal physical, developmental,
and emotional health. With over 50,000 surviving extremely preterm and very preterm infants
per year in the United States, we have the equivalent of an ongoing annual polio crisis in the prevaccination era. Not only are many of these
children growing up in families that are strapped
for material resources, services and time, but our
current early intervention and preschool systems
are not delivering child health, developmental,
and behavioral supports on a population basis.
Therefore, there is decreased likelihood that the
325,000 late preterm survivors who annually
enter kindergarten will be healthy and ready to
learn. Nor will they stay on track during the first
three grades, achieving adequate levels of literacy, numeracy, information handling, and social
participation; they will need to succeed in school
and life. As their development lags and their academic performance falters, there is too often a
response of educational systems, especially during the middle school years, to divert children by
using grade repetition, expulsions, and
nonevidence-based remediation that only
increase the likelihood that the child will leave
school or be classified with behavioral problems.
Importantly, unlike their middle-class and affluent counterparts, low-income children with a
spectrum of neurodevelopmental dysfunction
will have less access to high-quality medical
care, appropriately targeted and responsive
parent-child interactions, quality educational
accommodations and explicit strategies for promoting basic competencies, and extracurricular
activities.
Although potentially detrimental to a child’s
future, his/her gestational age and birth weight
are rarely the major determining factors when
long-term educational, social, and behavioral
outcomes are considered. In this chapter we have
341
examined several research studies that emphasized the importance of family, social, and economic environmental factors, which can either
aggravate or moderate neonatal risks caused by
premature birth. Much is to be gained by attention to how adversity impacts allostatic load, and
through epigenetic mechanisms and complex
protective buffers of parenting, health promotion,
and social supports, leads to long-term outcomes
of thriving across physical, behavioral, and social
health outcomes. With so much to lose while living under suboptimal conditions, it is of critical
importance that both health and educational professionals create systems for enhancing access to
early childhood learning experiences, parenting
supports, quality preschool education services,
and biopsychosocial strategies in middle childhood to help children achieve basic educational,
behavioral, and social competencies and adolescent supports of mentoring, preparation for independent living, and proactive behavioral health
strategies for stress, time management, mood disorders, and self-efficacy. We need more research
about how to spread and scale successful biopsychosocial health development strategies at a
community level to insure that VLBW and
ELBW survivors do not miss out on the critical
experiences, which have potential to compensate
for birth disadvantage, and are critical for longterm adult success. A summary of the gaps and
recommendations for increasing research in life
course health development after prematurity
follows.
Research in life course health development
for children who survive prematurity has made
the following advances:
1. Medical advances in pregnancy: Over the past
decades, there has been improved management of high-risk pregnancy disorders such as
hypertension, diabetes, multiple gestation,
intrauterine growth restriction, prenatal infections, and fetal malformations. There has been
growing awareness that women’s health
involving the use of tobacco, alcohol, illicit
drugs, anticonvulsants, and antidepressants
has subtle neurobehavioral consequences on
child development. Additionally, society has
M.E. Msall et al.
342
had greater recognition that poverty, social
adversity involving basic resources as well as
lack of partner/family supports, previous preterm birth, minority status, limited education,
and low health literacy increase the risk of
low birth weight, prematurity, and early childhood health and developmental impairments.
2. Medical advances in the neonatal period:
Over the last decades, there has been remarkable improvement in the management of premature labor and delivery. In particular, the
widespread screening for and treatment of
Group B Streptococcus colonization and the
use of maternal corticosteroids when preterm
labor presents have had marked improvements
in premature survival and sequelae of neonatal infections. We have had improved resuscitation and neonatal transport to tertiary care
centers. Throughout the vulnerable neonatal
course, improved respiratory support (the use
of positive pressure and noninvasive ventilator
techniques after surfactant), evidence-based
nutritional interventions, and infection control
management have improved neonatal
survival.
Although there is increased recognition that
very and extremely preterm survivors experience
a variety of health (e.g., growth, pulmonary),
neurological (e.g., sensory impairments, seizures), neurodevelopmental (e.g., CP, ID, ASD),
and learning and behavior impairment, the
population-level risks have not been systematically measured. In order to improve life course
health development, we propose seven theme
research agenda.
11
Recommendations
for an After Prematurity Life
Course Health Disparities
Research Agenda
1. Establish national registries.
It is imperative to create national registries
for children from birth to 5 years old who are
at highest risk of CP and neurodevelopmental
disabilities such as intellectual disability, neu-
rosensory disability, and autism spectrum disorders. These registries would not only
include preterm infants but also include term
infants with history of critical illness from
cardiac, pulmonary, infectious, neurological,
or genetic disorders. Such registries would
allow for the investigation of diverse interventions and link health, early intervention, special education, and rehabilitation services to
long-term outcomes across the life course.
Most importantly attention must be paid to
maternal health, adversity, and mental health.
Registries ought to include placental samples
as well as cord blood and newborn screening
blood spots, in order to allow for the evaluation of early biomarkers of toxic stress during
early childhood.
2. Build population-based datasets.
To understand health across the life course,
it is imperative to create population-based
datasets which originate at delivery and during
newborn primary care visits. We must be able
to link information on birth certificates with
early childhood health, developmental, and
educational outcomes through collaborative
arrangements between public health, regional
neonatal follow-up, early intervention, and
district-wide educational testing. We should
use these population databases to engage in
ongoing health and developmental surveillance of all children with all degrees of prematurity requiring neonatal intensive care,
especially those with seizures, neonatal
encephalopathy, congenital heart disease, malformation requiring surgery, and abnormalities
in newborn genetic and hearing screening. We
should model our US efforts on the
Longitudinal Study of Australian Children to
understand diverse trajectories of health,
development, and behavior for those with and
without special health-care needs.
Preterm infants need the establishment of
longitudinal data collection registries that can
collect information that is similar to the
National Survey of Children’s Health and
NHIS disability follow-up studies. We must
determine the risk factors for trajectories of
positive and negative outcomes across cohorts
Life Course Health Development Outcomes After Prematurity: Developing a Community, Clinical…
of middle childhood, adolescence, and adulthood for sequential preterm survivors who
were <28 weeks of gestation, 29–31 weeks of
gestation, 32–36 weeks of gestation, and term
gestation. Only with informed, collaborative,
and comprehensive life course health development registries will we determine ways to
support resiliency among preterm offspring of
high-risk mothers with respect to health,
developmental, educational, and social outcomes over the life course.
3. Expansion and evaluation of comprehensive
health and developmental surveillance interventions on a population level.
We need to improve population surveillance for children exposed to high-risk maternal conditions such as hypertension, diabetes,
obesity, asthma, seizure disorders, depression,
ADHD, anxiety, and maternal use of tobacco,
alcohol, and illegal substances. In the setting
of these risk factors, we must evaluate what
factors promote maternal and child resiliency
and promote positive newborn and developmental outcomes. We must understand which
factors mitigate the negative effect of these
exposures on growth, obesity, developmental
delays, and regulatory disorders. We should
seek to determine the impact of positive parenting, quality early child education (early
intervention, head start preschool programming), and pediatric medical homes especially
using neonatal and epigenetic biomarkers.
Surveillance should extend beyond the
neonatal period into preschool (ages
2–4 years) to identify young children who
lack the basic numeracy, literacy, and social
and emotional maturity needed to enter kindergarten ready to learn. Kindergarten is also
a key transition for measuring health and
developmental status, yet there are no systematic screening modalities in place at kindergarten entry across diverse biomedical and
social risks.
4. Integration of twenty-first-century technologies to improve care coordination and integrated health and engage families in
time-efficient interventions.
343
Diagnostic tools such as fidgety movements are shown to have high reliability in
detection of future motor delays; texting has
been shown to improve parental engagement with early childhood development;
and telemedicine has enabled the delivery
of highly specialized care to remote or
mobility-challenged populations. These
growing technologies, increasingly available
across socioeconomic populations, must be
fully embraced and leveraged for population
surveillance and therapeutic interventions.
5. Shift research methodology from antiquated
methods studying individuals and parent-child
dyads to novel techniques examining clusters
of families and communities.
Increasingly we need to better understand
the power of parent-child interactions on
determining developmental trajectories in
conjunction with genetic and epigenetic biomarkers. Furthermore, siblings certainly
impact health and development of one another,
yet most research fails to consider the contribution of sibling factors. We must seek to
develop and improve our research modalities,
which look beyond a single child as the unit of
measure and examine interrelatedness among
family and community factors.
6. Research
on
impacting
parent-child
interventions.
A critical need is to formally evaluate the
caregiving environment of premature infants
and determine how cumulative parent-child
interactions impact the child’s developmental
capacities and parental health and well-being.
Examine developmental activities as a part of
everyday childcare tasks to promote early parent involvement and positive health and regulatory trajectories.
7. Determine what types of interventions effectively impact maternal mental health in the
setting of adversities.
It is incompletely known how maternal mental health stressors (depression, anxiety, isolation, and violence) increase vulnerability of
children when they do not receive quality early
344
childhood social learning and behavioral
supports. Strategies for integrating maternal and
child physical and behavioral health have the
potential to increase resilience for high-risk
populations.
By combining themes from this research
agenda to populations involving life course
health development and the ICF model for promoting health, functioning, and participation, we
have the opportunities to improve thriving after
prematurity and measure our impact on longterm health, education, and community costs.
Acknowledgments This work was supported in part by
NIH NINR03095 (Sullivan, Msall). Dr. Msall was also
supported in part by NIH/NICHD Grant P30 HD0544275
JP Kennedy Intellectual and Developmental Disabilities
Research Center and HRSA-DHHS T73 MC 11047
Leadership Education in Neurodevelopmental and
Related Disorders Training Program (LEND).
References
Allen, M. C., Cristofalo, E. A., & Kim, C. (2011).
Outcomes of preterm infants: Morbidity replaces
mortality. Clinics in Perinatology, 38(3), 441–454.
doi:10.1016/j.clp.2011.06.011.
Aron, L., & Loprest, P. (2012). Disability and the education system. The Future of Children, 22(1), 97–122.
Arpi, E., & Ferrari, F. (2013). Preterm birth and behaviour problems in infants and preschool-age children:
A review of the recent literature. Developmental
Medicine and Child Neurology, 55(9), 788–796.
doi:10.1111/dmcn.12142.
Babenko, O., Kovalchuk, I., & Metz, G. A. (2015). Stressinduced perinatal and transgenerational epigenetic
programming of brain development and mental health.
Neuroscience and Biobehavioral Reviews, 48, 70–91.
doi:10.1016/j.neubiorev.2014.11.013.
Baek, Y., Vohr, B. R., Alksninis, B., Cashore, W. J., Hogan,
D. P., & Msall, M. E. (2002). Kindergarten readiness
in very low birth weight (VLBW) children. Pediatric
Research, 51(4), 293A.
Bauer, A., Parsonage, M., Knapp, M., Iemmi, V., &
Adelaja, B. (2014). The costs of perinatal mental
health problems. London: Cemtre for Mental Health
and London School of Economics.
Baumgardt, M., Bucher, H. U., Mieth, R. A., & Fauchere,
J. C. (2012). Health-related quality of life of former very
preterm infants in adulthood. Acta Paediatrica, 101(2),
e59–e63. doi:10.1111/j.1651-2227.2011.02422.x.
Bhutta, A. T., Cleves, M. A., Casey, P. H., Cradock,
M. M., & Anand, K. J. (2002). Cognitive and behav-
M.E. Msall et al.
ioral outcomes of school-aged children who were born
preterm: A meta-analysis. JAMA, 288(6), 728–737.
Bock, J., Wainstock, T., Braun, K., & Segal, M. (2015).
Stress in utero: Prenatal programming of brain
plasticity and cognition. Biological Psychiatry.
doi:10.1016/j.biopsych.2015.02.036.
Botting, N., Powls, A., Cooke, R. W., & Marlow, N.
(1998). Cognitive and educational outcome of verylow-birthweight children in early adolescence.
Developmental Medicine and Child Neurology,
40(10), 652–660.
Brown, K. J., Kilbride, H. W., Turnbull, W., & Lemanek,
K. (2003). Functional outcome at adolescence for
infants less than 801 g birth weight: Perceptions of
children and parents. Journal of Perinatology, 23(1),
41–47. doi:10.1038/sj.jp.7210850.
Calkins, K., & Devaskar, S. U. (2011). Fetal origins
of adult disease. Current Problems in Pediatric
and Adolescent Health Care, 41(6), 158–176.
doi:10.1016/j.cppeds.2011.01.001.
Capra, L., Tezza, G., Mazzei, F., & Boner, A. L. (2013). The
origins of health and disease: The influence of maternal
diseases and lifestyle during gestation. Italian Journal
of Pediatrics, 39, 7. doi:10.1186/1824-7288-39-7.
Chrousos, G. P., & Gold, P. W. (1992). The concepts
of stress and stress system disorders. Overview of
physical and behavioral homeostasis. JAMA, 267(9),
1244–1252.
Crouter, A. C., & Booth, A. (Eds). (2014). Work-family
challenges for low-income parents and their children.
Routledge. Abingdon, UK.
Curhan, G. C., Willett, W. C., Rimm, E. B., Spiegelman,
D., Ascherio, A. L., & Stampfer, M. J. (1996). Birth
weight and adult hypertension, diabetes mellitus, and
obesity in US men. Circulation, 94(12), 3246–3250.
Dahl, L. B., Kaaresen, P. I., Tunby, J., Handegard, B. H.,
Kvernmo, S., & Ronning, J. A. (2006). Emotional,
behavioral, social, and academic outcomes in adolescents born with very low birth weight. Pediatrics,
118(2), e449–e459. doi:10.1542/peds.2005-3024.
Dalziel, S. R., Parag, V., Rodgers, A., & Harding, J. E.
(2007). Cardiovascular risk factors at age 30 following
pre-term birth. International Journal of Epidemiology,
36(4), 907–915. doi:10.1093/ije/dym067.
Diagnostic and Statistical Manual of Mental Disorders:
DSM-5. (2013). (5th ed.). Washington, DC: American
Psychiatric Association.
Dmowska, A., Andrews, B., Schreiber, M., & Msall,
M. E. (2016). Preterm survivors: Community support
to lessen health disparities. In J. Merrick & L. Rubin
(Eds.), Environmental health disparities: Costs and
benefits of breaking the cycle. Hauppauge, NY: Nova
Publishers.
Drillien, C. M. (1961). A longitudinal study of the growth
and development of prematurely and maturely born
children. VIII. Morbidity in the age period 2-5 years.
Archives of Disease in Childhood, 36, 515–525.
Drillien, C. M., Thomson, A. J., & Burgoyne, K. (1980).
Low-birthweight children at early school-age: A lon-
Life Course Health Development Outcomes After Prematurity: Developing a Community, Clinical…
gitudinal study. Developmental Medicine and Child
Neurology, 22(1), 26–47.
Ekeus, C., Lindstrom, K., Lindblad, F., Rasmussen, F., &
Hjern, A. (2010). Preterm birth, social disadvantage,
and cognitive competence in Swedish 18- to 19-yearold men. Pediatrics, 125(1), e67–e73. doi:10.1542/
peds.2008-3329.
Farooqi, A., Hagglof, B., Sedin, G., Gothefors, L., &
Serenius, F. (2007). Mental health and social competencies of 10- to 12-year-old children born at 23 to 25
weeks of gestation in the 1990s: A Swedish national
prospective follow-up study. Pediatrics, 120(1), 118–
133. doi:10.1542/peds.2006-2988.
Feinberg, M. E., Jones, D. E., Roettger, M. E., Solmeyer,
A., & Hostetler, M. L. (2014). Long-term follow-up
of a randomized trial of family foundations: Effects
on children's emotional, behavioral, and school adjustment. Journal of Family Psychology, 28(6), 821–831.
doi:10.1037/fam0000037.
Feinberg, M. E., Roettger, M. E., Jones, D. E., Paul, I. M.,
& Kan, M. L. (2015). Effects of a psychosocial couplebased prevention program on adverse birth outcomes.
Maternal and Child Health Journal, 19(1), 102–111.
doi:10.1007/s10995-014-1500-5.
Gardner, F., Johnson, A., Yudkin, P., Bowler, U., Hockley,
C., Mutch, L., et al. (2004). Behavioral and emotional
adjustment of teenagers in mainstream school who
were born before 29 weeks’ gestation. Pediatrics,
114(3), 676–682. doi:10.1542/peds.2003-0763-L.
Graignic-Philippe, R., Dayan, J., Chokron, S., Jacquet,
A. Y., & Tordjman, S. (2014). Effects of prenatal stress
on fetal and child development: A critical literature
review. Neuroscience and Biobehavioral Reviews, 43,
137–162. doi:10.1016/j.neubiorev.2014.03.022.
Groot, W. (2000). Adaptation and scale of reference bias
in self-assessments of quality of life. Journal of Health
Economics, 19(3), 403–420.
Gross, S. J., Mettelman, B. B., Dye, T. D., & Slagle,
T. A. (2001). Impact of family structure and stability
on academic outcome in preterm children at 10 years
of age. The Journal of Pediatrics, 138(2), 169–175.
doi:10.1067/mpd.2001.111945.
Hack, M., Flannery, D. J., Schluchter, M., Cartar, L.,
Borawski, E., & Klein, N. (2002). Outcomes in young
adulthood for very-low-birth-weight infants. The
New England Journal of Medicine, 346(3), 149–157.
doi:10.1056/NEJMoa010856.
Hamilton, B. E., Martin, J. A., Osterman, M. J., Curtin,
S. C., & Matthews, T. J. (2015). Births: Final data
for 2014. National Vital Statistics Reports, 64(12),
1–64. Retrieved from http://www.ncbi.nlm.nih.gov/
pubmed/26727629.
Hille, E. T., den Ouden, A. L., Bauer, L., van den
Oudenrijn, C., Brand, R., & Verloove-Vanhorick,
S. P. (1994). School performance at nine years of age
in very premature and very low birth weight infants:
Perinatal risk factors and predictors at five years of
age. Collaborative project on preterm and small for
345
gestational age (POPS) infants in The Netherlands.
The Journal of Pediatrics, 125(3), 426–434.
Hille, E. T., Weisglas-Kuperus, N., van Goudoever, J. B.,
Jacobusse, G. W., Ens-Dokkum, M. H., de Groot,
L., et al. (2007). Functional outcomes and participation in young adulthood for very preterm and very
low birth weight infants: The Dutch project on preterm and small for gestational age infants at 19 years
of age. Pediatrics, 120(3), e587–e595. doi:10.1542/
peds.2006-2407.
Himmelmann, K., & Uvebrant, P. (2014). The panorama
of cerebral palsy in Sweden. XI. Changing patterns in
the birth-year period 2003-2006. Acta Paediatrica,
103(6), 618–624. doi:10.1111/apa.12614.
Hintz, S. R., Kendrick, D. E., Vohr, B. R., Poole, W. K., &
Higgins, R. D. (2008). Community supports after surviving
extremely low-birth-weight, extremely preterm birth: Special
outpatient services in early childhood. Archives of Pediatrics
& Adolescent Medicine, 162(8), 748–755. doi:10.1001/
archpedi.162.8.748.
Hofman, P. L., Regan, F., Jackson, W. E., Jefferies,
C., Knight, D. B., Robinson, E. M., et al. (2004).
Premature birth and later insulin resistance. The New
England Journal of Medicine, 351(21), 2179–2186.
doi:10.1056/NEJMoa042275.
Hoirisch-Clapauch, S., Brenner, B., & Nardi, A. E.
(2015). Adverse obstetric and neonatal outcomes
in women with mental disorders. Thrombosis
Research, 135(Suppl 1), S60–S63. doi:10.1016/
s0049-3848(15)50446-5.
Holzmann, R., & Jørgensen, S. (2001). Social risk
management: A new conceptual framework for
social protection, and beyond. International Tax
and Public Finance, 8(4), 529–556. doi:10.102
3/A:1011247814590.
Howard, L. M., Piot, P., & Stein, A. (2014). No health without perinatal mental health. The Lancet, 384(9956),
1723–1724. doi:10.1016/S0140-6736(14)62040-7.
Huston, A. C. (1991). Children in poverty : Child development and public policy. Cambridge, NY: Cambridge
University Press.
Income, Poverty, and Health Insurance Coverage in the
United States: 2005 (2006). In U. S. C. Bureau (Ed.).
Irving, R. J., Belton, N. R., Elton, R. A., & Walker, B. R.
(2000). Adult cardiovascular risk factors in premature
babies. Lancet, 355(9221), 2135–2136. doi:10.1016/
s0140-6736(00)02384-9.
Jiang Y. E. M., & Skinner C. (2015). Basic facts about
low-income children: Children under 3 Years, 2013
(trans: D. o. H. P. Management). Columbia University:
National Center for Children in Poverty.
Johnson, S., Hennessy, E., Smith, R., Trikic, R.,
Wolke, D., & Marlow, N. (2009). Academic attainment and special educational needs in extremely
preterm children at 11 years of age: The EPICure
study. Archives of Disease in Childhood. Fetal and
Neonatal Edition, 94(4), F283–F289. doi:10.1136/
adc.2008.152793.
346
Johnson, S., Wolke, D., Hennessy, E., & Marlow, N.
(2011). Educational outcomes in extremely preterm
children: Neuropsychological correlates and predictors of attainment. Developmental Neuropsychology,
36(1), 74–95. doi:10.1080/87565641.2011.540541.
Jones, K. M., Champion, P. R., & Woodward, L. J. (2013).
Social competence of preschool children born very
preterm. Early Human Development, 89(10), 795–
802. doi:10.1016/j.earlhumdev.2013.06.008.
Joseph, R. M., O'Shea, T. M., Allred, E. N., Heeren, T.,
Hirtz, D., Paneth, N., Leviton, A., & Kuban, K. C.
(2016). Prevalence and associated features of autism
spectrum disorder in extremely low gestational age
newborns at age 10 years. Autism Research, 10(2),
224–232. doi:10.1002/aur.1644.
Kaijser, M., Bonamy, A. K., Akre, O., Cnattingius, S.,
Granath, F., Norman, M., et al. (2008). Perinatal risk factors for ischemic heart disease: Disentangling the roles
of birth weight and preterm birth. Circulation, 117(3),
405–410. doi:10.1161/circulationaha.107.710715.
Kane, H. S., Dunkel Schetter, C., Glynn, L. M., Hobel,
C. J., & Sandman, C. A. (2014). Pregnancy anxiety and
prenatal cortisol trajectories. Biological Psychology,
100, 13–19. doi:10.1016/j.biopsycho.2014.04.003.
Lang, C. T., & Iams, J. D. (2009). Goals and strategies for
prevention of preterm birth: An obstetric perspective.
Pediatric Clinics of North America, 56(3), 537–563.
Lefebvre, F., Mazurier, E., & Tessier, R. (2005). Cognitive
and educational outcomes in early adulthood for infants
weighing 1000 grams or less at birth. Acta Paediatrica,
94(6), 733–740. doi:10.1080/08035250510025987.
Litt, J. S., Gerry Taylor, H., Margevicius, S., Schluchter,
M., Andreias, L., & Hack, M. (2012). Academic
achievement of adolescents born with extremely low
birth weight. Acta Paediatrica, 101(12), 1240–1245.
doi:10.1111/j.1651-2227.2012.02790.x.
MacCabe, J. H., Martinsson, L., Lichtenstein, P., Nilsson,
E., Cnattingius, S., Murray, R. M., et al. (2007).
Adverse pregnancy outcomes in mothers with affective psychosis. Bipolar Disorders, 9(3), 305–309.
doi:10.1111/j.1399-5618.2007.00382.x.
Marciniak, B., Patro-Malysza, J., PoniedzialekCzajkowska, E., Kimber-Trojnar, Z., LeszczynskaGorzelak, B., & Oleszczuk, J. (2011). Glucocorticoids
in pregnancy. Current Pharmaceutical Biotechnology,
12(5), 750–757.
McLean, M., Bisits, A., Davies, J., Woods, R., Lowry, P.,
& Smith, R. (1995). A placental clock controlling the
length of human pregnancy. Nature Medicine, 1(5),
460–463.
Morse, S. B., Zheng, H., Tang, Y., & Roth, J. (2009). Early
school-age outcomes of late preterm infants. Pediatrics,
123(4), e622–e629. doi:10.1542/peds.2008-1405.
Moster, D., Lie, R. T., & Markestad, T. (2008). Long-term
medical and social consequences of preterm birth. The
M.E. Msall et al.
New England Journal of Medicine, 359(3), 262–273.
doi:10.1056/NEJMoa0706475.
Msall, M. E. (2012). Academic achievement after extreme
prematurity: Optimizing outcomes for vulnerable
children in times of uncertainty. Acta Paediatrica,
101(12), 1196–1197. doi:10.1111/apa.12038.
Msall, M. E., Phelps, D. L., DiGaudio, K. M., Dobson,
V., Tung, B., McClead, R. E., et al. (2000). Severity
of neonatal retinopathy of prematurity is predictive
of neurodevelopmental functional outcome at age
5.5 years. Behalf of the Cryotherapy for retinopathy
of prematurity cooperative group. Pediatrics, 106(5),
998–1005.
Msall, M. E., Phelps, D. L., Hardy, R. J., Dobson,
V., Quinn, G. E., Summers, C. G., et al. (2004).
Educational and social competencies at 8 years in
children with threshold retinopathy of prematurity in
the CRYO-ROP multicenter study. Pediatrics, 113(4),
790–799.
O'Connor, T. G., Monk, C., & Fitelson, E. M. (2014).
Practitioner review: Maternal mood in pregnancy and
child development--implications for child psychology and psychiatry. Journal of Child Psychology and
Psychiatry, 55(2), 99–111. doi:10.1111/jcpp.12153.
van Oppenraaij, R. H., Jauniaux, E., Christiansen, O. B.,
Horcajadas, J. A., Farquharson, R. G., & Exalto, N.
(2009). Predicting adverse obstetric outcome after
early pregnancy events and complications: A review.
Human Reproduction Update, 15(4), 409–421.
doi:10.1093/humupd/dmp009.
Patrianakos-Hoobler, A. I., Msall, M. E., Marks, J. D.,
Huo, D., & Schreiber, M. D. (2009). Risk factors
affecting school readiness in premature infants with
respiratory distress syndrome. Pediatrics, 124(1),
258–267. doi:10.1542/peds.2008-1771.
Pinto-Martin, J. A., Levy, S. E., Feldman, J. F., Lorenz,
J. M., Paneth, N., & Whitaker, A. H. (2011).
Prevalence of autism spectrum disorder in adolescents
born weighing <2000 grams. Pediatrics, 128(5), 883–
891. doi:10.1542/peds.2010-2846.
Poehlmann, J., Hane, A., Burnson, C., Maleck, S.,
Hamburger, E., & Shah, P. E. (2012). Preterm infants
who are prone to distress: Differential effects of parenting on 36-month behavioral and cognitive outcomes.
Journal of Child Psychology and Psychiatry, 53(10),
1018–1025. doi:10.1111/j.1469-7610.2012.02564.x.
Preterm Birth: Causes, Consequences, and Prevention
(2007). In R. E. Behrman, & A. S. Butler (Eds.).
Washington, DC: Committee on Understanding
Premature Birth and Assuring Healthy Outcomes,
Institute of Medicine of the Academies.
Provencal, N., & Binder, E. B. (2015). The effects of early
life stress on the epigenome: From the womb to adulthood and even before. Experimental Neurology, 268,
10–20. doi:10.1016/j.expneurol.2014.09.001.
Life Course Health Development Outcomes After Prematurity: Developing a Community, Clinical…
Requejo, J., Merialdi, M., Althabe, F., Keller, M., Katz,
J., & Menon, R. (2013). Born too soon: Care during pregnancy and childbirth to reduce preterm
deliveries and improve health outcomes of the preterm baby. Reproductive Health, 10(Suppl 1), S4.
doi:10.1186/1742-4755-10-s1-s4.
Rini, C. K., Dunkel-Schetter, C., Wadhwa, P. D., &
Sandman, C. A. (1999). Psychological adaptation
and birth outcomes: The role of personal resources,
stress, and sociocultural context in pregnancy. Health
Psychology, 18(4), 333–345.
Rosenberg, S. A., Zhang, D., & Robinson, C. C. (2008).
Prevalence of developmental delays and participation in early intervention services for young children. Pediatrics, 121(6), e1503–e1509. doi:10.1542/
peds.2007-1680.
Rubens, C. E., Sadovsky, Y., Muglia, L., Gravett, M. G.,
Lackritz, E., & Gravett, C. (2014). Prevention of preterm birth: Harnessing science to address the global
epidemic. Science Translational Medicine, 6(262),
262sr265. doi:10.1126/scitranslmed.3009871.
Sable, M. R., & Wilkinson, D. S. (2000). Impact of perceived stress, major life events and pregnancy attitudes
on low birth weight. Family Planning Perspectives,
32(6), 288–294.
Saigal, S., & Doyle, L. W. (2008). An overview of mortality and sequelae of preterm birth from infancy to
adulthood. Lancet, 371(9608), 261–269. doi:10.1016/
s0140-6736(08)60136-1.
Saigal, S., Pinelli, J., Hoult, L., Kim, M. M., & Boyle,
M. (2003). Psychopathology and social competencies
of adolescents who were extremely low birth weight.
Pediatrics, 111(5 Pt 1), 969–975.
Shah, P. E., Robbins, N., Coelho, R. B., & Poehlmann,
J. (2013). The paradox of prematurity: The behavioral
vulnerability of late preterm infants and the cognitive
susceptibility of very preterm infants at 36 months
post-term. Infant Behavior & Development, 36(1),
50–62. doi:10.1016/j.infbeh.2012.11.003.
Simms, V., Gilmore, C., Cragg, L., Clayton, S., Marlow,
N., & Johnson, S. (2015). Nature and origins of mathematics difficulties in very preterm children: A different etiology than developmental dyscalculia. Pediatric
Research, 77(2), 389–395. doi:10.1038/pr.2014.184.
Smith, C. J., & Ryckman, K. K. (2015). Epigenetic and
developmental influences on the risk of obesity, diabetes, and metabolic syndrome. Diabetes Metab Syndr
Obes, 8, 295–302. doi:10.2147/dmso.s61296.
Stein, A., Pearson, R. M., Goodman, S. H., Rapa, E.,
Rahman, A., McCallum, M., et al. (2014). Effects
of perinatal mental disorders on the fetus and
child. Lancet, 384(9956), 1800–1819. doi:10.1016/
s0140-6736(14)61277-0.
Stephens, B. E., & Vohr, B. R. (2009). Neurodevelopmental
outcome of the premature infant. Pediatric Clinics of
North America, 56(3), 631–646., Table of Contents.
doi:10.1016/j.pcl.2009.03.005.
347
The March of Dimes data book for policy makers:
Maternal, infant, and child health in the United States
(2012). Washington, DC: March of Dimes.
Torche, F., & Kleinhaus, K. (2012). Prenatal stress, gestational age and secondary sex ratio: The sex-specific
effects of exposure to a natural disaster in early
pregnancy. Human Reproduction, 27(2), 558–567.
doi:10.1093/humrep/der390.
Treyvaud, K., Ure, A., Doyle, L. W., Lee, K. J., Rogers,
C. E., Kidokoro, H., et al. (2013). Psychiatric outcomes
at age seven for very preterm children: Rates and predictors. Journal of Child Psychology and Psychiatry,
54(7), 772–779. doi:10.1111/jcpp.12040.
Vohr, B. (2013). Long-term outcomes of moderately
preterm, late preterm, and early term infants. Clinics
in Perinatology, 40(4), 739–751. doi:10.1016/j.
clp.2013.07.006.
Vohr, B. R., & Msall, M. E. (1997). Neuropsychological
and functional outcomes of very low birth weight
infants. Seminars in Perinatology, 21(3), 202–220.
Wadhwa, P. D., Dunkel-Schetter, C., Chicz-DeMet, A., Porto,
M., & Sandman, C. A. (1996). Prenatal psychosocial
factors and the neuroendocrine axis in human pregnancy.
Psychosomatic Medicine, 58(5), 432–446.
Wainstock, T., Anteby, E. Y., Glasser, S., Lerner-Geva,
L., & Shoham-Vardi, I. (2014). Exposure to lifethreatening stressful situations and the risk of preterm
birth and low birth weight. International Journal
of Gynaecology and Obstetrics, 125(1), 28–32.
doi:10.1016/j.ijgo.2013.09.035.
Weiss, E. L., Msall, M.E., & Muhammad, C. (2007).
Barriers to receipt of early intervention. Passages.
doi:E-PAS2007:616311.10.
Weitzman, M., Byrd, R. S., Aligne, C. A., & Moss, M.
(2002). The effects of tobacco exposure on children's
behavioral and cognitive functioning: Implications for
clinical and public health policy and future research.
Neurotoxicology and Teratology, 24(3), 397–406.
Wijesiriwardana, A., Bhattacharya, S., Shetty, A., Smith,
N., & Bhattacharya, S. (2006). Obstetric outcome in
women with threatened miscarriage in the first trimester. Obstetrics and Gynecology, 107(3), 557–562.
doi:10.1097/01.AOG.0000199952.82151.de.
Williams, M. A., Mittendorf, R., Lieberman, E., &
Monson, R. R. (1991). Adverse infant outcomes associated with first-trimester vaginal bleeding. Obstetrics
and Gynecology, 78(1), 14–18.
Williams, J., Lee, K. J., & Anderson, P. J. (2010). Prevalence
of motor-skill impairment in preterm children who
do not develop cerebral palsy: A systematic review.
Developmental Medicine and Child Neurology, 52(3),
232–237. doi:10.1111/j.1469-8749.2009.03544.x.
Winchester, S. B., Sullivan, M. C., Roberts, M. B., &
Granger, D. A. (2016). Prematurity, birth weight, and
socioeconomic status are linked to atypical diurnal
hypothalamic-pituitary-adrenal Axis activity in young
adults. Research in Nursing & Health, 39(1), 15–29.
348
Wolke, D., Strauss, V. Y., Johnson, S., Gilmore, C.,
Marlow, N., & Jaekel, J. (2015). Universal gestational
age effects on cognitive and basic mathematic processing: 2 cohorts in 2 countries. J Pediatr, 166(6),
1410–1416. doi:10.1016/j.jpeds.2015.02.065.
World Health Organization. (2007). ICF-CY, international classification of functioning, disability, and
M.E. Msall et al.
health: Children & Youth version. Geneva: World
Health Organization.
Yonkers, K. A., Smith, M. V., Forray, A., Epperson, C. N.,
Costello, D., Lin, H., et al. (2014). Pregnant women
with posttraumatic stress disorder and risk of preterm
birth. JAMA Psychiatry, 71(8), 897–904. doi:10.1001/
jamapsychiatry.2014.558.
Open Access This chapter is licensed under the terms of the Creative Commons Attribution 4.0 International License
(http://creativecommons.org/licenses/by/4.0/), which permits use, sharing, adaptation, distribution and reproduction in any
medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the
Creative Commons license and indicate if changes were made.
The images or other third party material in this chapter are included in the chapter’s Creative Commons license, unless
indicated otherwise in a credit line to the material. If material is not included in the chapter’s Creative Commons license
and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain
permission directly from the copyright holder.
A Life Course Approach to Hearing
Health
Shirley A. Russ, Kelly Tremblay, Neal Halfon,
and Adrian Davis
1
Introduction
Challenges to hearing health are a significant
public health problem.
At least ten million Americans have a hearing
loss that interferes with the understanding of normal speech (Mitchell 2005). If lesser degrees of
loss are included, the number rises to 28 million
(US DHHS 2010). Sensorineural hearing loss
S.A. Russ, MD, MPH (*)
UCLA Center for Healthier Children, Families and
Communities, Department of Pediatrics, David
Geffen School of Medicine, UCLA,
Los Angeles, CA, USA
e-mail: shirlyruss@cox.net
K. Tremblay
Speech & Hearing Sciences College of Arts &
Sciences, University of Washington,
Seattle, WA, USA
N. Halfon
Department of Pediatrics, David Geffen School of
Medicine, UCLA, Los Angeles, CA, USA
Department of Health Policy and Management,
Fielding School of Public Health, UCLA, Los
Angeles, CA, USA
Department of Public Policy, Luskin School of Public
Affairs, UCLA, Los Angeles, CA, USA
Center for Healthier Children, Families, and
Communities, UCLA, Los Angeles, CA, USA
A. Davis
University College London, NHS Newborn Hearing
Screening Program, London, UK
(SNHL) is the commonest sensory deficit in
more developed societies (Davis 1989). The term
“deaf” is usually reserved for individuals with
severe (>60–90 dBHL) or profound (>90 dBHL)
losses, representing half a million Americans,
while those with mild (<40 dBHL) or moderate
(40–60 dBHL) losses are referred to as “hard of
hearing” (Smith et al. 2005; Blanchfield et al.
2001). Congenital losses sufficient to adversely
affect speech and language development are seen
in at least one per thousand newborns (Fortnum
et al. 2001; Russ et al. 2003; Van Naarden et al.
1999), and if lesser degrees of loss and unilateral
losses are included, this number rises to up to five
per thousand. Only 4% cases of hearing loss
nationwide are accounted for by children under
the age of 18 years, while 50% cases occur in
persons 65 years of age or older (Russ 2001). The
cumulative prevalence of hearing loss within the
US population rises with age, as has been demonstrated in other countries (Russ 2001; Parving
and Christensen 1996), with the sharpest rise in
prevalence occurring in old age, when 50–80%
are ultimately affected (see Table 1). The relative
contributions of delayed diagnosis of congenital
losses, progression of existing losses, and acquisition of new losses to the rising prevalence of
hearing loss with age are uncertain. Improvements
to the prevention, diagnosis, and management of
hearing loss across all age groups are public
health priorities (Reavis et al. 2016; Davis et al.
2016).
© The Author(s) 2018
N. Halfon et al. (eds.), Handbook of Life Course Health Development,
DOI 10.1007/978-3-319-47143-3_15
349
S.A. Russ et al.
350
Table 1 Prevalence of hearing loss in the USA across the life span
Age
Newborn
Newborn
Newborn
Newborn
3 years
10 years
8 years
6–19 years
18–34 years
Estimated
prevalence of
hearing loss
% (95% CI) Case definition
0.14%
Bilateral loss >35
dBHL requiring
amplification
0.19%
Loss ≥35 dBHL in
one or both ears
0.31%
Unilateral and
bilateral
sensorineural and
conductive losses
0.33%
Unilateral and
bilateral
sensorineural
hearing loss
Bilateral PTA loss
0.067%
at 0.5, 1, and 2
(0.053–
KHz ≥40dBHL in
0.085)
better unaided ear
0.14%
(0.12–0.16)
0.14%
Bilateral PTA loss
0.12%
at 0.5, 1, and 2
KHz ≥40 dBHL in
better unaided ear
0.4%
Bilateral loss at
low (0.5, 1, and 2
kHZ) frequencies
PTA ≥26 dBHL
0.7%
Bilateral loss at
high (3, 4, and 6
kHZ) frequencies
PTA ≥26 dBHL
14.9%
Unilateral or
bilateral loss PTA
>16 dBHL at low
or high frequencies
3.4%
Self-reported
hearing trouble
35–44 years
6.3%
Self-reported
hearing trouble
45–54 years
10.3%
Self-reported
hearing trouble
48–59 years
70–79 years
80 years
21%
66%
90%
PTA 0.5, 1, 2, and
4 KHz >25 dBHL
in the worst ear
Author and year of
study
Mason, Herrman
1992–1997
Place of study and
sample size
Honolulu, HI,
10,372
Mehl, Thompson
1992–1999
Finitzo, Albright,
O’Neal
1994–1997
Colorado
63,590
Texas
54,228
Barsky-Firsker, Sun Livingston, New
1993–1995
Jersey, 15,749
Metropolitan
Van Naarden,
Decoufle, Caldwell Atlanta 255,742
1991–1993
Data source
Kaiser Permanente
Honolulu, HI, NHSP
Colorado NHSP
Texas Newborn
Hearing Screening
Data
St. Barnabas Medical
Center Newborn
Hearing Screening
Data
Metropolitan Atlanta
Developmental
Disabilities
Surveillance Program
(MADDSP)
Karapurkar Bhasin,
Brocksen, Avchen,
Van Naarden Braun
1996 and 2000
Niskar, Kieszak,
Holmes, et al.
1988–1994
Metropolitan
Atlanta
36,749 (1996)
43,593 (2000)
USA 5249
MADDSP
National Center for
Health Statistics
(1990 and 1991)
National Center for
Health Statistics
(1990 and 1991)
National Center for
Health Statistics
(1990 and 1991)
Cruickshanks,
Wiley, Tweed, et al.
USA 239,663
National Health
Interview Survey
(NHIS)
National Health
Interview Survey
(NHIS)
National Health
Interview Survey
(NHIS)
Epidemiology of
Hearing Loss Study
(EHLS)
USA 239,663
USA 239,663
Beaver Dam, WI,
(EHLS) 4541
NHANES III
(continued)
A Life Course Approach to Hearing Health
351
Table 1 (continued)
Age
60–90 years
73–84 years
Estimated
prevalence of
hearing loss
% (95% CI) Case definition
29%
PTA 0.5, 1, and 2
KHz >26 dBHL in
the better ear
59.9%
Hearing loss: two
averages of
thresholds 0.5, 1,
and 2 KHz >25
dBHL
76.9%
High-frequency
hearing loss >40
dBHL 2, 4, and 8
KHz
Author and year of
study
Gates, Cooper,
Kannel, et al.
1983–1985
Helzner, Cauley,
Pratt, et al.
1997–1998
Although there have been considerable
advances in understanding the etiology of hearing
loss, with genetic causes now thought to account
for up to 50% of congenital losses (Mitchell
2005), in many individual cases, the cause of
hearing loss remains unknown. Even where
genetic causes have been identified, discovery of
the abnormal gene does not necessarily lead to an
understanding of the mechanism whereby the
gene’s product exerts its effect. Similarly, genetic
and environmental causes of hearing losses that
have their onset later in life have not been well
defined. This lack of knowledge of the basic
pathophysiology of hearing difficulties hampers
prevention and treatment efforts.
Hearing health has important implications for
general health and well-being. Both children and
adults with hearing loss face significant educational and social challenges. For children who are
profoundly deaf, language and academic levels at
high school graduation have been reported historically to correspond to those of fourth grade
students with normal hearing (Holt 1993). Adults
with hearing loss are reported to have higher levels of unemployment (Parving and Christensen
1993) and lower quality of life than their hearing
peers (Appollonio et al. 1996). For older individuals, hearing disability is associated with accelerated cognitive decline, depression, increased risk
of dementia, poorer balance, falls, hospitaliza-
Place of study and
sample size
Framingham 1662
Pittsburgh,
Pennsylvania, and
Memphis,
Tennessee, 2052
Data source
Framingham Heart
Study
Health, Aging, and
Body Composition
(ABC) Study
tions, and early mortality (for a review, see Davis
et al. 2016). In addition to these medical consequences, there are also social functioning implications including social isolation due to reduced
communication, loss of autonomy, and financial
decline. Traditionally, hearing losses in childhood
and in adult life have been considered as separate
issues.
Growing interest in life course theory has led
to suggestions that it could prove useful to apply
a life course lens to the study of hearing loss, and
of hearing health, throughout the life span. The
early years of life, especially the period from
conception through to 3 years of age, are now
understood to impact lifelong health. Childhood
conditions and early experiences can become
“embedded” into emerging biological systems,
altering health trajectories. The Life Course
Health Development (LCHD) model posits that
health is an emergent capacity of human beings
that dynamically develops over time in response
to multiple-nested, ever-changing genetic, biological, behavioral, social, and economic contexts. Multiple risk and protective factors
influence development of key biological systems,
including the anatomic and biochemical determinants of hearing ability, during critical and sensitive periods of development (see Table 2). Health,
at individual and population levels, is also influenced by the timing and sequence of biological,
S.A. Russ et al.
352
Table 2 Risk and protective factors for hearing loss across the life span
Life stage
Prenatal
Perinatal
Early childhood
Middle childhood,
adolescence, and adulthood
Risk factor
1. Syndrome association with HL
2. Family history of permanent
childhood SNHL
3. Craniofacial anomalies
4. In utero TORCH
1. NICU >48 h
2. Jaundice-exchange Tx
3. Ototoxic medications
4. Meningitis
Protective factor
1. Maternal rubella immunization
Pre- and Perinatal risk factors, plus:
1. Parent/caregiver concern
2. Persistent pulmonary hypertension
with ventilation.
3. Conditions requiring ECMO
4. Syndromes associated with
progressive hearing loss (e.g.,
neurofibromatosis, osteopetrosis,
Usher’s)
5. Neurodegenerative disorder (e.g.,
Hunter’s, sensorimotor neuropathies,
Friedreich’s ataxia,
Charcot-Marie-Tooth)
6. Head trauma
7. Recurrent/persistent OME for ≥ 3
months
Early childhood risk factors plus:
Noise
Drug/chemical exposure
Head trauma
Otosclerosis
cultural, and historic events and experiences.
Application of the LCHD model to hearing health
challenges predominantly biomedical models
and suggests that there are multiple potential avenues for improving hearing health. As hearing
losses in childhood and in adult life have been
considered as separate issues, investigations into
adult hearing loss largely ignore early-life exposures. However, the LCHD model highlights the
importance of studying these links.
In this paper we consider the implications of
the LCHD model for understanding the mechanisms, pathways, and determinants of hearing
ability. We consider the implications of early hearing loss for health development over the life course
and the factors through the life course that contribute to hearing ability in adult life. We consider the
concept not just of hearing loss but of “hearing
1. Prompt treatment for neonatal
jaundice.
2. Avoid/monitor ototoxic medications.
3. Prompt antibiotic treatment for
meningitis
1. Immunization
2. Avoid/monitor ototoxic medications.
3. Prompt antibiotic treatment for
meningitis
4.Head injury prevention
5.Noise avoidance/protection
1. Noise avoidance/protection
2. Avoid/monitor ototoxic drugs
3. Head injury prevention
4. Immunizations
5. Higher family income
6. Education
health” and how to achieve it, the research priorities that are suggested by this review, and the
implications for policy and practice.
2
The Life Course Health
Development Model
and the Mechanisms,
Pathways, and Determinants
of Hearing Ability
According to Halfon and Forrest (2017), the
LCHD model is grounded in the following seven
principles:
1. Health development: Health development
integrates the concepts of health and developmental processes into a unified whole.
A Life Course Approach to Hearing Health
2. Unfolding: Health development unfolds continuously over the life span, from conception
to death, and is shaped by prior experiences
and environmental interactions.
3. Complexity: Health development results from
adaptive, multilevel, and reciprocal interactions between individuals and their physical,
natural, and social environments.
4. Timing: Health development is sensitive to the
timing and social structuring of environmental
exposures and experiences.
5. Plasticity: Health development phenotypes
are malleable and enabled and constrained by
evolution to enhance adaptability to diverse
environments.
6. Thriving: Optimal health development promotes survival, enhances well-being, and protects against disease.
7. Harmony: Health development results from
the balanced interactions of molecular, physiological, behavioral, cultural, and evolutionary processes.
Taken together, these seven principles suggest
that in order to understand the mechanisms
underlying hearing loss and hearing health from
a life course standpoint, it is essential to explore
the role of multiple risk and protective factors
operating at multiple levels to influence hearingrelated outcomes. In addition, scientists must
study the emergence and development of hearing
health trajectories over extended time frames,
including the pivotal role of social relationships
in the development of functional hearing capacity. Researchers must consider the critical importance of timing in relation to sensitive periods
and turning points in the development of hearing
abilities. All of these principles point to the
importance of adopting a developmental perspective on hearing health. The following section takes each of these issues in turn, providing
a review of the evidence pertaining to each.
353
single or principal cause for clinic