Location via proxy:   [ UP ]  
[Report a bug]   [Manage cookies]                

Ramakrishna Et Al

Download as pdf or txt
Download as pdf or txt
You are on page 1of 32

Annual Review & Research in Biology

1(4): 79-110, 2011


SCIENCEDOMAIN international
www.sciencedomain.org

Stem Cells and Regenerative Medicine –


A Review

V. Ramakrishna1*, P. B. Janardhan1 and L. Sudarsanareddy2

1
Department of Biotechnology and Bioinformatics, Yogi Vemana University,
Kadapa – 516003, India.
2
Department of Genomics & Genetics, Nanyang Technological University,
Singapore-637551.

Received 1st June 2011


Review Article Accepted 24th June 2011
Online Ready 15th July 2011

ABSTRACT

Regenerative medicine is a multidisciplinary field concerned with the replacement, repair or


restoration of injured tissues. This field emerged from the need for reconstruction in children
and adults in whom tissue has been damaged by diseases, trauma and congenital
anomalies. Stem cell research is a promising field with an alluring potential for therapeutic
intervention, and thus begs a critical understanding of the long-term consequences of stem
cell replacement. Stem cells have unrestricted potential to divide and this strength is used for
the regeneration and repair of cells within the body during tissue damage. Research on stem
cells is advancing knowledge about how an organism develops from a single cell and how
healthy cells replace damaged cells in adult organisms. This promising area of science is
also leading scientists to investigate the possibility of cell-based therapies to treat disease. In
our present review we tried to provide the information about stem cells and their significant
role in regenerative medicine for treatment of various diseases.

Keywords: Stem cells; embryonic stem cells; adult stem cells; stem cells treatment;
regenerative medicine; stem cell therapy.

____________________________________________________________________________________________

*Corresponding author: Email: vrkrishna70@gmail.com;


Annual Review & Research in Biology, 1(4): 79-110, 2011

CONTENTS

1. INTRODUCTION

1.1 Types of stem cells


1.1.1 Embryonic stem cells
1.1.1.1 Human embryonic germ cells
1.1.1.2 Amniotic epithelial cells
1.1.2 Umbilical cord blood stem cells
1.1.3 Human adult stem cells:
1.1.3.1 Hematopoietic stem cells
1.1.3.2 Mesenchymal stem cells
1.1.3.3 Neural stem cells
1.1.3.4 Pancreatic stem cells
1.1.3.5 Skin stem cells

2. STEM CELLS VERSUS REGENERATIVE MEDICINE

2.1 Embryonic stem cells in regenerative medicine


2.2 Adult stem cells in regenerative medicine

3. STEM CELL TREATMENT FOR MAJOR DISEASES

3.1. Stem cells therapy in neurological disorders


3.1.1 Parkinson’s disease (PD)
3.1.2 Alzheimer’s disease (AD)
3.2 Stem cells in the regeneration of skeletal muscle cells
3.3 Stem cells in cardiac repair
3.3.1 Embryonic stem cell
3.3.2 Human adult bone marrow–derived stem cells
3.3.3 Cardiac stem cells
3.3.4 Mesenchymal stem cells
3.3.5 Umbilical cord blood stem cells
3.4 Stem cells in orthopedic research
3.5 Stem cell in cancer therapies
3.6 Stem cells in the treatment of diabetes

4. FUTURE PROSPECTIVE

5. REFERENCES

80
Annual Review & Research in Biology, 1(4): 79-110, 2011

1. INTRODUCTION

Regenerative medicine is an emerging and rapidly evolving field of research and


therapeutics to restore, maintain and improve body functions (Polak et al., 2008). Daar and
Greenwood (2007) stated that regenerative medicine aims at ‘repair, replacement or
regeneration of cells, tissue or organs to restore impaired function’. It aids the body to form
new functional tissue to replace lost or defective tissue. Ultimately, this will help to provide
therapeutic treatment for conditions where current therapies are inadequate. Cell therapy
and tissue engineering are part of the broader field of regenerative medicine, whose aim is
the delivery of safe, effective and consistent therapies. The human body has an endogenous
system of regeneration and repair through stem cells, where stem cells can be found almost
in every type of tissue. This process is highly evolved through evolution, and so it is logical
that restoration of function is best accomplished by these cells. Therefore, stem cells hold
great promise for the future of translational medicine (NRC, 2002). Regenerative medicine is
also a primer for pediatricians (Bajada et al., 2008; Julia and Polak, 2009; Vacanti, 2010;
Longakar, 2010).

In the early 1900’s European researchers realized that the various type of blood cells - white
blood cells, red blood cells and platelets all came from a particular ‘stem cell’. Stem cells
were first studied by Becker et al. (1963), who injected bone marrow cells into irradiated
mice and noticed that nodules developed in the spleens of the mice in proportion to the
number of bone marrow cells injected. They concluded that each nodule arose from a single
marrow cell. Later on, they found by evidence that these cells were capable of infinite self-
renewal, a central characteristic of stem cells. Thus, stem cells by definition have two
essential properties, i.e. the capacity of self renewal, and the capacity to differentiate into
different cell lineages. Under the right conditions, or given the right signals, stem cells can
give rise (differentiate) to the many different cell types that make up the organism (figure-1).
Stem cell lineage determination is explained by several ideas, one among is focused on the
stem cells microenvironment or ‘niche’. A niche consists of signaling molecules, intercellular
communication and the interaction between stem cells and their neighboring extracellular
matrix. This three-dimensional microenvironment is thought to influence/control genes and
properties that define ‘stemness’ of the stem cells, i.e. self-renewal or development to
committed cells. An interesting theory put forward is that stem cells might be terminal
differentiation cells with the potential to display diverse cell types, depending on the host
niche. Adult stem cells that are implanted into a totally different niche (different germ layer)
can potentially differentiate into cell types similar to those found in the new environment. The
potential of stem cells and its plasticity are having invaluable properties for regenerative
medicine (Singh and Williams, 2008). Beneficiaries of regenerative medicine include the
increasingly ageing population, people with sports injuries and war casualties. The
tremendous technological progress achieved during the last decade in gene transfer
methods and imaging techniques, as well as recent increases in our knowledge of cell
biology, have opened new horizons in the field of regenerative medicine. Genetically
engineered cells are a tool for tissue engineering and regenerative medicine, albeit a tool
whose development is fraught with difficulties (Sheyn et al., 2010; Voog and Jones, 2010).
This review summarizes current knowledge of stem cells in regenerative medicine
particularly in the treatment of various diseases.

1.1 Types of Stem Cells

There are two main types of stem cells, embryonic and non-embryonic. Embryonic stem
cells (ESCs) are totipotent and, accordingly, they can differentiate into all three embryonic

81
Annual Review & Research in Biology, 1(4): 79-110, 2011

germ layers. On the other hand, non-embryonic stem cells (non-ESCs), also known as adult
stem cells, are just multipotent; their potential to differentiate into different cell types seems
to be more limited (Lee and Hui, 2006). Embryonic stem cells are derived from the inner cell
mass of a blastocyst (a very early embryo) and the adult stem cells are derived from mature
tissue. A large variety of cell types have been used for regenerative medicine, including
adult cells, resident tissue specific stem cells, bone marrow stem cells, embryonic stem cells
(Guillott et a. 2007) and the recent breakthrough discovery of induced pluripotent stem cells
from mature/adult cells (iPS) (Lensch. 2009).

1.1.1 Embryonic stem cells

Human embryonic stem cells (ES cells) are primitive (undifferentiated) cells that can self-
renew or differentiate into all cell types found in adult human body (Edwards, 2004; Gardner,
2007; Bajada et al., 2008; Pelligrini and Luca, 2010). The derivation of mouse ES cells was
first reported in 1981 (Evans and Kaufman, 1981; Martin, 1981) but it was not until 1998 that
the derivations of human ES cell lines were first reported (Thomson et al., 1998). A new era
in stem cell biology began in 1998 with the derivation of cells from human blastocysts and
fetal tissue with the unique ability of differentiating into cells of all tissues in the body.
Embryonic stem cells are derived from embryos at a developmental stage before the time
that implantation would normally occur in the uterus. Each of the cells (blastomeres) of these
cleavage-stage embryos is undifferentiated. The first differentiation event in humans occurs
at approximately five days of development, when an outer layer of cells committed to
becoming part of the placenta (trophectoderm) separates from the inner cell mass (ICM).
The ICM cells have the potential to generate any cell type of the body, but after implantation,
they are quickly depleted as they differentiate to other cell types with more limited
developmental potential. The ICM derived cells can continue to proliferate and replicate
them indefinitely and still maintain the developmental potential to form any cell type of the
body (Figure 1).

Bongso et al. (1994) first described isolation and culture of cells of the inner cell mass of
human blastocysts, and techniques for deriving and culturing stable hES cell lines were first
reported in 1998 (Thomson et al., 1998). The trophectoderm was removed from 5th day
blastocysts consisting ICM of 30-34 cells, was placed into tissue culture. The possible
sources of stem cells are embryos created via In vitro Fertilization (IVF) (Lanzendorf et al.,
2001), embryos or fetuses obtained through elective abortion and embryos created via
somatic cell nuclear transfer (SCNT) or cloning. They can be isolated by immunosurgery
from the inner cell mass of the embryo during the blastocyst stage, and are usually grown on
feeder layers consisting of mouse embryonic fibroblasts or human feeder cells (Richards et
al., 2002). More recent reports have shown that these cells can be grown without the use of
a feeder layer (Amit et al., 2003), and thus avoid the exposure of these human cells to
mouse viruses and proteins. These cells have demonstrated longevity in culture by
maintaining their undifferentiated state for at least 80 passages when grown using published
protocols (Reubinoff et al., 2000; Bajada et al., 2008). The source of ESCs opens a
Pandora’s Box of ethical dilemmas, including the moral status of the embryo, the sanctity of
life (Towns and Jones, 2004) and the possible use of saviour siblings as a source of ESCs.
These add to the long-standing accusation to scientists of tampering with the natural
process of life. The ethical debate relates to whether it is right to use human tissue in an
abnormal manner. Life-saving situations where the strongest ethical arguments can be
made to support the use of cells that are from an embryo that will not become an
independent human life.

82
Annual Review & Research in Biology, 1(4): 79-110, 2011

Fig. 1. General hierarchy for the stem cell niche


Zygote and early cell division stages (blastomeres) to the morula stage are defined as Totipotent. At the blastocyst stage, only the
cells of the inner cell mass (ICM) retain the capacity (Pluripotent) to build up all three primary germ layers, the endoderm, mesoderm,
and ectoderm as well as the primordial germ cells (PGC), the founder cells of male and female gametes. In adult tissues, multipotent
stem and progenitor cells exist in tissues and organs to replace lost or injured cells. The dashed lines indicate the extent of
possibilities of the adult stem cells may also be developing into cells of other lineage. Embryonic stem (ES) cells, derived from the
ICM, have the developmental capacity to differentiate in vitro into cells of all somatic cell lineages as well as into male and female
germ cells.

83
Annual Review & Research in Biology, 1(4): 79-110, 2011

As non-ESCs use becomes more widespread, then acceptance of ESCs treatments may
increase. When ethical obstacles are overcome, ESCs might be introduced for treating
several conditions, including diabetes (Soria et al., 2000), spinal cord injuries (Hendricks et
al., 2006) and liver (Duan et al., 2007) and heart transplantation (Kofidis et al., 2005).
Recently Guenou et al. (2009) demonstrated that human embryonic stem cells (hESCs) can
differentiate into mature keratinocytes able to generate a pluristratified epithelium on
immunodeficient mice (Pelligrini and Luca, 2010). Jukes et al (2010) reviewed on
chondrogenic and osteogenic differentiation of mouse and human embryonic stem cells
(ESCs) and their potential in cartilage and bone tissue engineering.

Embryonic stem cells have been shown to differentiate into cells from all three embryonic
germ layers in vitro (figure-1). Skin and neurons formed from ectodermal differentiation
(Reubinoff et al., 2001; Schuldiner et al., 2001; Zhang et al., 2001; Adewumi et al., 2007),
blood, cardiac cells, cartilage, endothelial cells, and muscle formed from mesodermal
differentiation (Kaufman et al., 2001; Kehat et al., 2001; Levenberg et al., 2002) and
pancreatic cells from endodermal differentiation (Assady et al., 2001). In addition, as further
evidence of their pluripotency, embryonic stem cells can form embryoid bodies, the cell
aggregations that contain all three embryonic germ layers, while in culture, and can form
teratomas in vivo (Itskovitz-Eldor et al., 2000; Knoepler, 2009).
.
1.1.1.1 Human embryonic germ cells

Embryonic germ cells are derived from primordial germ line cells in early fetal tissue. Unlike
embryonic stem cells, animal experiments on embryonic germ cells have been limited. In
1998 the isolation, culture, and partial characterization of germ cells derived from the
gonadal ridge of human tissue obtained from abort uses were reported (Shamblott et al.,
1998). There are fewer data from animal embryonic germ cell experiments than from ES cell
experiments, but it is generally assumed that the range of potential fates will be relatively
limited compared to ES cells, because the embryonic germ cells are much further along in
development (5-9 weeks). Fetal tissue may provide committed progenitors, but the feasibility
of large scale sourcing and manufacturing of products utilizing such cells is questionable.
Furthermore, the behavior of these cells in vivo is not well understood; significant research
will be required to avoid unwanted outcomes, including ectopic tissue formation i.e.,
additional, unwanted tissue, tumor induction, or other abnormal development (Adewumi et
al., 2007).

1.1.1.2 Amniotic epithelial cells

The latest addition to our repertoire of stem cells is amniotic fluid stem cells. The embryo is
known to shed a variety of cells into the surrounding amniotic fluid during development.
Amniotic epithelial cells (AECs) derived from the amniotic membrane in human placenta,
also express the markers that are present on pluripotent ESCs and EGCs, such as Oct-4,
Nanog, and alkaline phosphatase. They can also differentiate as ESCs and EGCs in the cell
lineages from three germ layers, including pancreatic endocrine cells and hepatocytes
(endoderm), cardiomyocytes (mesoderm), and neural cells (ectoderm) in vitro (Tamagawa
et al., 2004; Miki et al., 2006; Nicol et al., 2007; De Coppi et al., 2007). Because of the
observation that they proliferate at a high rate without apparent loss of pluripotency or
teratogenic potential when transplanted in immunodeficient animals, amniotic fluid stem cells
were credited with being a safer alternative to hESCs. Such claims, however, may be
premature and these results must be independently verified, with further work required to

84
Annual Review & Research in Biology, 1(4): 79-110, 2011

ascertain whether they really do have the same degree of pluripotency as hESCs (Abdelkrim
et al., 2009).

1.1.2 Umbilical cord blood stem cells

In the late 1980s, umbilical cord blood (ucb) was recognized as an important clinical source
of HSCS (Barker et al., 2003; Koh et al., 2004). Blood from the placenta and umbilical cord
is a rich source of hematopoietic stem cells, and these cells are typically discarded with the
afterbirth. Several approaches have been tested to overcome the cell dose issue, including,
with some success, pooling of cord blood samples (Koh et al., 2004; Wagner et al., 2007;
Harries et al., 2007; Abdelkrim et al., 2009). Cord blood stem cells have been in routine
clinical practice for the past 20 years. The development of new therapeutic protocols in
regenerative medicine requires the use of stem cells and umbilical cord blood is an
important and readily available source of cells for these applications. The latest concepts in
routine transplantation of cord blood are reviewed followed by the critical role of cord blood
stem cells in regenerative medicine research and novel approaches using cord blood as a
source of whole blood for transfusion (Seres and Hollands, 2010). cord blood stem cell
technology has many advantages over embryonic and other adult stem cells for several
reasons, including the following: (i) cord blood represents a potentially unlimited source of
stem cells that can in theory be collected at every birth; (ii) cord blood is relatively simple to
process and store using tried and tested technology and, once frozen in liquid nitrogen, is
biologically stable; (iii) the collection of cord blood is a non-invasive procedure with no
danger to either mother or baby. If cord blood is not collected, it is discarded as biological
waste; and (iv) cord blood carries low risk of infection (Hollands, 2009).

1.1.3 Human adult stem cells

Stem cells that are found in developed tissue, regardless of the age of the organism at the
time are referred to as adult stem cells. Adult stem cells encompass a variety of populations
of undifferentiated cells and are found in most adult tissues, where they act as reservoirs
during the normal turnover and regeneration of an organ or tissue. Both their potency and
proliferative potential are typically narrower than those of their embryonic counterparts. Adult
stem cells are hidden deep within organs, surrounded by millions of ordinary cells, and may
help replenish some of the body’s cells when needed. An adult stem cell is undifferentiated
cells found among differentiated cells in many mammalian tissues contain stem cell
populations that might self renew and generate somatic cells normally as well as mobilize,
proliferate and differentiate. Until recently, it had been thought that a blood-forming cell in the
bone marrow - which is called a hematopoietic stem cell, could not give rise to the cells of a
very different tissue, such as nerve cells in the brain. Therefore, exploring the possibility of
using adult stem cells for cell-based therapies has become a very active area of
investigation by researchers. Rather, adult stem cells are defined by their functional
properties: high proliferative potential, substantial self-renewal capacity and ability to
differentiate into at least one type of mature functional progeny (Morrison et al., 1997;
Weissman, 2002; Eckfeldt et al., 2005; Abdelkrim et al., 2009; Jones et al., 2010).

1.1.3.1 Hematopoietic stem cells

These cells discovery marked the beginning of modern day stem-cell research.
Hematopoietic stem cells have characteristic morphologic appearances and cell-surface
markers (Lagasse et al., 2000) that allow them to be labeled and tracked in the bloodstream
and target tissues or to be isolated and cultured in vitro (Adewumi et al., 2007).

85
Annual Review & Research in Biology, 1(4): 79-110, 2011

Haematopoietic stem cells represent less than 0.05% of the total bone marrow, but they
have the potential to reconstitute all blood forming lineages. Because of the enormous
clinical implications of such ability, the haematopoietic stem cell compartment has historically
been the best characterized stem cell niche (Abdelkrim et al., 2009).

1.1.3.2 Mesenchymal stem cells

Mesenchymal stem cells (MSCs) are capable of self-renewal and differentiation into multiple
cell lineages. MSCs are multipotent and are easily derived from a variety of tissues,
including fat, skin and bone marrow. Presently, bone marrow is considered as a prime
source of MSCs and also gingival (Tomar et al., 2010). These cells are fibroblastic in
appearance and can be expanded for many passages. Most importantly, populations of
mesenchymal stem cells (MSCs) are strongly adherent, therefore can be isolated by
culturing marrow on an appropriate substrate and washing other cells off. Mesenchymal
stem cells can give rise to many kinds of connective tissue cells including those responsible
for remodeling of cartilage, bone, fat, and vascular tissue (Pittenger and Martin, 2004; Chen
et al., 2008). MSCs are likely to participate in maintenance of the essential
microenvironment necessary to support the hematopoietic stem cells in the bone marrow
(Dennis and Caplan, 2004). MSCs can be isolated from circulating blood (Zvaifler et al.,
2000), as well as from diverse nonhematopoietic tissues such as synovium (De Bari et al.,
2001), adipose tissue (Zuk et al., 2001), trabecular bone (Noth et al., 2002), dermis (Young
et al., 2001), dental pulp (Pierdomenico et al., 2005), and the lung (Sabatini et al., 2005).
Although MSCs have been detected in the lung (Sabatini et al., 2005), their origin(s) remain
unknown, and an evolving paradigm predicts that mesenchymal cells participating in lung
repair derive from the bone marrow (Epperly et al., 2003; Fine, 2004; Hashimoto et al., 2004)
and wound healing (Wu et al., 2007). Human adult mesenchymal stem cells (MSCs) are
non-hematopoietic, adherent fibroblast-like cells with intrinsic ability of self-renewal and
potential for multilineage differentiation. In vitro derived MSCs express a panel of
characteristic surface markers such as Thy-1 (CD90), SH-2/endoglin (CD105), SH-3/4
(CD73), b-1-integrin (CD29), and CD44; and are negative for hematopoietic markers such as
CD34, CD14, and CD45. MSCs differentiate in vitro primarily into the cells of mesenchyme
lineage such as bone, cartilage, and adipose tissue (Chamberlain et al., 2007; Tomar et al.,
2010).

1.1.3.3 Neural stem cells

Neural stem cells can be defined operationally as cells that can continuously self-renew and
have the potential to generate intermediate and mature cells of both glial and neuronal
lineages (Gage, 2000; Baizabal et al., 2003). Generally neuronal stem cells have been
isolated from the brains of embryos and adults. Adult neural stem cells have the potential to
differentiate into multiple cell types of the brain, mainly oligodendrocytes, astrocytes, and
neurones, giving them a major therapeutic advantage over committed progenitor cells such
as those used in transplants for Parkinson's disease. Moreover, these neural stem cells may
be multipotent: when injected into blastocysts of mice they contributed to multiple types of
tissues in the embryos (Clarke et al., 2000). One study reported the generation of blood from
neuronal stem cells when transplanted into lethally irradiated recipients, but this has not yet
been reproduced (Bjornson et al., 1999). These neuronal stem cells have also been
observed to generate skeletal muscle when cultured with a cell line capable of differentiating
into muscle or when injected into regenerating muscle (Galli et al., 2000). In brain-injury
models neural stem cells (NSCs) proliferate in those neurogenic regions and are even able
to migrate toward the site of damage. NSCs are multipotent and capable of self-renewing. In

86
Annual Review & Research in Biology, 1(4): 79-110, 2011

vitro they cluster in ‘‘neurospheres,’’ which are able to differentiate into the 3 major
neuroectodermal lineages (neurons, astrocytes, and oligodendrocytes) (Brignier and
Gewirtz, 2010).

1.1.3.4 Pancreatic stem cells

The mammalian adult pancreas has three tissue types: the ductal tree, the exocrine acini,
which produces digestive enzymes, and the endocrine islets of Langerhans, composed of
insulin-producing β-cells, glucagon-producing α-cells, somatostatin-producing δ-cells, and
pancreatic polypeptide-producing γ -cells. Moreover, the multipotent cell progenitors have
also been identified within the ducts and islets in adult rodent and human pancreas (Seaberg
et al., 2004, Bouwens et al., 2005). It has been reported that the multipotent PSCs isolated
from the human fetal pancreas and expressing stem cell markers, such as nestin, ATP-
binding cassette transporter (ABCG2), and KIT, as well as epidermal growth factor receptor
(EGFR), hepatocyte growth factor receptor (c-Met), and glucagon-like peptide receptor were
able to form the islet like cell clusters (ICCs) when cultured ex vivo (Suen et al., 2005).
These ICCs can give rise to diverse pancreatic cell lineages, including insulin-secreting cells.
Type 1 diabetes results from the autoimmune destruction of cells in pancreatic islets, and
can be reversed by islet cell transplantation (Ma et al., 2008).

1.1.3.5 Skin stem cells

Numerous studies have revealed that the upper region of hair follicles, the bulge area,
constitutes the principal niche of multipotent stem cells, which are responsible for the long-
term growth of the hair follicles and epidermis regeneration after injury (Blanpain et al., 2004;
Morris et al., 2004; Li and Xie, 2005; Rendl et al., 2005; Millar et al., 2005; Levy et al., 2005).
More specifically, multipotent epithelial stem cells (bESCs) within the bulge area, which
express CD34, K5, and α 6-integrin, are able to proliferate and give rise to the follicular
epithelium, as well as to new cells constituting IFE and sebaceous glands after severe injury
(Flores et al., 2005; Sarin et al., 2005). The bulge area in adult mammalian hair follicle also
contains a pluripotent epidermal neural crest stem cell (eNCSC) population that shows
several properties similar to embryonic neural crest stem cells (Sieber-Blum et al., 2004).
The pluripotent eNCSCs in the bulge area are also able to self-renew and give rise to
multiple cell lineages in vivo, including melanocytes, neurons, Schwann cells, smooth
muscle cells, and chondrocytes (Sieber-Blum et al., 2004). It has been observed that each
individual multipotent cell from melanoma spheres was able to differentiate, like eNCSCs
under well-defined conditions, into multiple cell types, including melanocytes, adipocytes,
osteocytes, and chondrocytes (Shihuan Kuang et al., 2007; Fang et al., 2005). The small
clusters of multipotent stem cells, which express the specific markers, including K15, appear
to reside near the basement membrane of the epidermis (Watt, 2002).

2. STEM CELLS VERSUS REGENERATIVE MEDICINE

Stem cells have the potential to create miracles. for the first time in history, it became
possible for physicians to regenerate a damaged tissue with a new supply of healthy cells by
drawing on the unique ability of stem cells to create many of the body’s specialized cell
types. the prospect of exploiting stem cells more widely in regenerative medicine was
encouraged by the development of human assisted conception and growing evidence that
various adult cells retained greater versatility than had been suspected hitherto.

87
Annual Review & Research in Biology, 1(4): 79-110, 2011

2.1 Embryonic Stem Cells in Regenerative Medicine

Although the term stem cells are often used in reference to repair cells within an adult
organism, a more fundamental variety of stem cells is found in the early stage embryo.
Similarly, there is now emerging evidence of benefit following transplantation of human
embryonic stem cell derived neural progenitors and cardiomyocytes into animal models of
parkinson’s disease and myocardial injury respectively (Ben-hur et al., 2004: Kofidis et al.,
2006; Singh and Williams, 2008; Liu et al., 2008). In the mouse, there is now proof of
concept for the use of ES cell-derived tissues to treat models of diabetes (Serra et al.,
2002), parkinson’s disease (Frenck et al., 1998; Singh and Williams, 2008), myocardial
infarction (Rufer et al., 1999), spinal injury (Reyes et al., 2001), and a severe genetic
immune disorder (Abdelkrim et al., 2009). In as much as this type of experimentation with
mouse ES cells has gotten under way in only the past 10 years, the progress is
encouraging. For most cell types of interest, this is not yet really feasible, though in some
areas (neural progenitors) pure populations of precursor cells may routinely be obtained
from ES cultures, expanded in numbers, and differentiated into mature cells. For
neurodegenerative diseases, it is better to transplant neural progenitor cells or fully mature
neurons. conversely, recent evidence suggests that even in their undifferentiated state,
human ESCS express discrete levels of HLA class I antigens that increase as the cells
mature (Martin et al., 2005; Bajada et al., 2008). An interesting viewpoint has recently been
proposed that irreversible arrest of cell division rather than the death of each and every cell
correspond to the organism death of the embryo (Landry et al., 2004; Gardner, 2007).
Embryonic stem (ES) cells are pluripotent cells that can give rise to derivatives of all three
embryonic germ layers. Due to its characteristics, the patient-specific ES cells are of great
potential for transplantation therapies. Considering future clinical use, the differentiation from
ES to neurons, cardiomyocytes and many other types of cells provide basic cognition and
experience to regenerative medicine (Ma et al., 2008).

2.2 Adult Stem Cells in Regenerative Medicine

Adult stem cell populations have been most thoroughly characterized in mouse and human
bone marrow, where they continuously replenish the differentiated cells of the peripheral
blood lost through attrition. From studies of the haematopoietic system it has been possible
to define a stem cell as a cell with the capacity to self renew and to generate cells of multiple
diverse lineage within the tissue in which the stem cell resides. The ability of the
haematopoietic stem cells within bone marrow to give rise to all blood elements has been
extensively exploited in the clinic for transplantation of bone marrow and stem cells. Recent
reports suggest that some of these stem cells can differentiate outside of their tissue of
origin. Both muscle and neural tissue appear to be a source of hematopoietic stem cells
(Jackson et al., 1999; Galli et al., 2000; Ma et al., 2008), whereas bone marrow may house
muscle precursor cells (Ferrari et al., 1998). Moreover, bone marrow stroma, which contains
mesenchymal stem cells (Liechty et al., 2000; Abdelkrim et al., 2009), may also give rise to
neurons and glia (Mezey et al., 2000; Woodbury et al., 2000; Ma et al., 2008). Indeed, the
breadth of lineage capabilities for both the mesenchymal stem cells and hematopoietic stem
cells of bone marrow are subjects of active study and lively debate (Liechty et al., 2000;
Weissman, 2000; Gardner, 2007; Gorden, 2008; Bajada et al., 2008).

88
Annual Review & Research in Biology, 1(4): 79-110, 2011

3. STEM CELL TREATMENT FOR MAJOR DISEASES

3.1 Stem Cells Therapy in Neurological Disorders

The mature central nervous system (CNS) has a limited capacity for self-repair; therefore
many different cell-engineering strategies are used to regenerate damaged neurons. Stem
cells will provide an inexhaustible source of neurons and glia for therapies aimed at cell
replacement or neuroprotection in disorders affecting the brain and spinal cord. The most
obvious and familiar application of stem cell research for nervous system disorders is
through cell replacement therapy. The possibility of using stem cells as a source of neurons
that can be implanted to replace cells and circuits lost in Parkinson’s disease, amyotrophic
lateral sclerosis (ALS), Huntington’s disease, or Alzheimer’s disease is an exciting prospect
(Baizabal et al., 2003; Singh and Williams, 2008; Ma et al., 2008; Sheyn et al., 2010). Vawda
et al. (2007) review potential sources of cellular replacements, including embryonic stem
cells, fetal and neonatal neural stem cells and a variety of mesenchymal stem cells. They
also reviewed published studies to illustrate where stem cell therapies have been evaluated
for therapeutic gain and discuss the hurdles that will need to be overcome to achieve
therapeutic benefit. Overall, they concluded that children with paediatric brain injuries or
inherited genetic disorders that affect the brain are worthy candidates for stem cell
therapeutics. Neurodegenerative diseases are characterized by progressive and gradual
irreversible loss of physiologically active neurons over a period of several years, ultimately
leading to significant morbidity and mortality (Steiner et al., 2006). The two most common
age-related neurodegenerative disorders are Parkinson’s and Alzheimer’s diseases.

3.1.1 Parkinson’s disease (PD)

Parkinson's disease (PD) is a very common neurodegenerative disorder that affects more
than 2% of the population over 65 years of age. PD is pathologically hallmarked by the
presence of intraneuronal Levy bodies and a progressive neurodegeneration of
dopaminergic neurons in the substantia nigra, resulting in depletion of striatal dopamine.
This is clinically manifested in motor dysfunctions and rigidity, sometimes combined with rest
tremor and postural changes (Fahn et al., 2003). Factors that support this notion include the
knowledge of the specific cell type (DA neurons) needed to relieve the symptoms of the
disease. It is thought that PD may be the first disease to be amenable to treatment using
stem cell transplantation. The hope is that research on human ES cells may reveal methods
for producing an infinite supply of dopamine neurons for transplant into patients and the
isolation of human embryonic stem (hES) cells (Thomson, et al., 1998) has stimulated
research aimed at the selective generation of specific cell types for regenerative medicine.

At NIH, Lee et al., (2006) have used a progressive expansion, selection, and differentiation
strategy to convert mouse ES cells to a mixed population of mature neurons in tissue culture
with up to 30% having the characteristics of dopamine cells. Using different approach,
Kawasaki et al., (2000) have generated dopamine neurons from mouse ES cells without
embroid body formation. Clinical trials of the transplantation of human fetal dopaminergic
neurons have shown that cell replacement can produce major, long-lasting improvement in
some patients (Lindvall et al., 2004). So it is promising that cells with properties of
dopaminergic neurons have been generated in vitro from stem cells of various sources, such
as ES cells and stem cells isolated from bone marrow and fetal brain (Takagi et al., 2005).
To make stem-cell therapy for PD, dopaminergic neurons with the characteristics of
substantia nigra neurons must be produced in large numbers. Some patients will need
implants in several areas of the brain (Piccini et al., 2005); optimum recovery will require a

89
Annual Review & Research in Biology, 1(4): 79-110, 2011

tailor-made grafting procedure based on preoperative imaging. It will also be necessary to


develop strategies that hinder disease progression. One possible approach to prevent the
death of existing neurons could be to transplant human stem cells engineered to express
neuroprotective molecules such as glial-cell-line-derived neurotrophic factor (GDNF)
(Behrstock et al., 2006).

Regarding human stem cell therapy, scientists are developing a number of strategies for
producing dopamine neurons from human stem cells in the laboratory for transplantation into
humans with Parkinson's disease (Anderson and Lenz, 2006, Singh and Williams, 2008). To
overcome the shortcomings of fetal/embryonic tissues as sources for neural grafts and
invasive surgical procedures, embryonic stem cells (Ho and Li, 2006), neural stem cells
derived from fetal or adult brain (Sanberg, 2007) and other tissue stem cells derived either
from bone marrow or umbilical cord blood have been experimentally applied to generate
dopaminergic neurons (Singh and Williams, 2008). Such cells will help to provide a clinically
competent and effective therapeutic regime without the need for further interventions. Stem
cells graft strategies are: in vitro pre-differentiation to dopaminergic neurons prior to
transplantation; or in vivo differentiation of stem cells after implantation into the striatum or
substantia nigra. The most important clinical issue is the ability to generate functional
dopamine neurons and establish the role of other cell types, such as glial cells, present in
the mesencephalic fetal grafts in the differentiation and function of these neurons. Site-
specific integration in to the brain parenchyma is essential to replace dopamine in a
physiologically natural fashion. This requires transplanting a cell population with a high
percentage of live cells secreting a consistent and standard amount of dopamine that is
capable of interlinking with the host cells to replace damaged neuronal circuitry without
immune rejection. Importantly, the loss of a single phenotype of cells, together with the
uniform pathology that characterizes Parkinson’s disease, suggests treatment regimes
based on the substitution of this single neuronal cell type. The successful generation of an
unlimited supply of dopamine neurons could make neurotransplantation widely available for
Parkinson's patients at some point in the future.

3.1.2 Alzheimer’s disease (AD)

Alzheimer’s disease (AD) is characterized by neuronal and synaptic loss throughout the
brain, involving the basal forebrain cholinergic system, amygdala, hippocampus and several
cortical areas. Patient’s memory and cognitive performance is progressively impaired; they
develop dementia; and are likely to die prematurely. Current therapies, such as treatment
with acetyl cholinesterase inhibitors to enhance cholinergic function, give only partial and
temporary alleviation of symptoms. The pathological changes seen in AD offer an extremely
problematic situation for cell replacement. Given the widespread and progressive damage in
the brains of patients with AD, it is unlikely that the mechanisms for instructing transplanted
NS cells to differentiate into new neurons will be intact. In theory, transplanting cholinergic
neurons generated from NS cells in vitro could prevent cognitive decline caused by the
degeneration of basal forebrain cholinergic neurons. But to provide long-lasting symptomatic
benefit, this approach would require the existence of intact target cells within the patient’s
brain, and these are highly likely to be damaged. However, because stem cells can be
genetically modified and have migratory capacity after transplantation, they could be used
for the delivery of factors that can modify the course of the disease. In support of this
approach, basal forebrain grafts of fibroblasts that produce nerve growth factor (NGF)- which
counteracts cholinergic neuronal death, stimulates cell function and improves memory in
animal models have been of some benefit in patients with AD (Tuszynski et al., 2005). Cell-
based treatment should therefore have the potency to differentiate into the three requisite

90
Annual Review & Research in Biology, 1(4): 79-110, 2011

cell types, be capable of engraftment and survival as transplants, and cause axonal
regeneration and directional synaptogenesis, thereby inducing simultaneous morphological
and functional modulation of cells and the disease environment. The engrafted cells
contribute to functional restoration by providing neurons for processing and transmitting
signals, oligodendrocytes for remyelination and neurotrophic factors to defend the existing
host cells (Singh and Williams, 2008).

3.2 Stem Cells in the Regeneration of Skeletal Muscle Cells

Muscle injuries and trauma are common phenomena and may result in diminished muscle
function and structural deformation. Although muscle tissue has a regenerative ability, it is
limited and decreases with the age of the patient; once muscle loses its ability to regenerate,
the defect fills up with scar and adipose tissue. Muscle dystrophies are severe diseases and
often lethal, as in the case of Duchenne muscular dystrophy (DMD). DMD is caused by the
absence of a key protein, dystrophin, which causes chronic degeneration of the myofibers.
Although extensive studies of this disease have been made, there is still no therapy that will
prevent or halt the muscle deterioration. Muscle regeneration thus seems an attractive
treatment (Jarvinen et al., 2007; Jukes et al., 2010; Sheyn et al., 2010). The capacity for this
regenerative response is primarily due to a mononuclear cell population termed satellite
cells. In 1961, Alexander Mauro utilized ultra structural techniques and identified the satellite
cells as a rare cell population that is resident in adult skeletal muscle of the frog. The satellite
cells either fuse to form multinucleated myotubes or re-establish a residual pool of quiescent
satellite cells that have the capability of supporting additional rounds of regeneration
(Shihuan Kuang et al., 2007; Fabien et al., 2007). Pulse-chase experiments have suggested
that the satellite cell pool was heterogenous and promoted the notion that a pool of satellite
cells maintained a residual pool consistent with a self-renewal mechanism (Schultz et al.,
1996). More recent studies using cultured myofibers demonstrate that activated satellite cells
principally differentiate and express MyoD family members. In contrast, activated satellite
cells may exit the cell cycle, re-establish a quiescent state, and stimulated to reenter the cell
cycle, suggesting that satellite cells are capable of self-renewal or replenishment of a
residual pool of cells (Olguin and Olwin 2004; Zammit et al., 2004; Shihuan Kuang et al.,
2007).

Collins et al. (2005) observed that the transplantation of one myofiber containing as few as
seven satellite cells gave rise to >100 new myofibers containing >25,000 differentiated donor
myonuclei. Importantly, these transplanted satellite cells gave rise to anatomically distinct
satellite cells that participated in the repopulation of myofibers following a newly induced
injury with a myotoxin. Further studies will unravel the mechanisms that direct the self-
renewal process, define the proliferative capacity of the satellite cell population in vivo, and a
hierarchical satellite cell cascade (Weismann et al., 2000). The satellite cell hierarchy will
serve as a platform for cell-based therapies (Stephane et al., 2007). The development of
tissue-specific scaffolds that can successfully imitate and support proper muscle tissue
organization and allow muscle vascularization and innervation will clearly promote
regeneration field (Rawlands et al, 2007; Fujita et al., 2009; Jukes et al., 2010). Finally,
muscle dystrophies, such as DMD, affect the various muscles of the body in a multifocal
manner, making it harder to treat damaged muscles with a local implantation method
(Benchaouir et al., 2007). To address this challenge, intra-arterial and systemic cell-delivery
techniques must be improved and understanding of the homing mechanism of stem cells
must be increased.

91
Annual Review & Research in Biology, 1(4): 79-110, 2011

3.3 Stem Cells in Cardiac Repair

Cardiovascular disease is a leading cause of death throughout the world. Globally each
year, approximately 16.7 million people die of cardiovascular disease, accounting for 29% of
all deaths in that time period (WHO, 2006; Sheyn et al., 2010). Restoration of cardiovascular
function is the ultimate goal of stem-cell-based therapy. In principle, cardiovascular stem
cells can improve cardiac function via de novo cardiomyogenesis, enhanced myocardial
neovascularization and prevention of postinfarct remodeling. Stem cell transplantation to
improve cardiac function has received mixed results in human clinical trials (Liu et al., 2008).
Strategies to regenerate damaged cardiac tissue by cardiomyocyte transplantation may limit
post infarction cardiac failure (Kofidis et al., 2005; Liu et al., 2008). Recent research is
providing early evidence that adult and embryonic stem cells may be able to replace
damaged heart muscle cells and establishes new blood vessels. Cardiomyocyte, the heart
muscle cell that contract to eject the blood out of the heart's main pumping chamber (the
ventricle). Two other cell types are important to a properly functioning heart are the vascular
endothelial cell, which forms the inner lining of new blood vessels, and the smooth muscle
cell, which forms the wall of blood vessels. The potential capability of both embryonic and
adult stem cells to develop into these cell types in the damaged heart is now being explored
as part of a strategy to restore heart function to people who have had heart attacks or have
congestive heart failure (Figure 2). Various types of stem cells are using in cardiac therapy,
which has been described below.

3.3.1 Embryonic stem cell

Human ES cell derived cardiomyocytes display structural and functional properties of early-
stage cardiomyocytes that couple electrically with host cardiomyocytes when transplanted
into normal myocardium (Kehat et al., 2004). In theory, infinite numbers of cardiomyocytes
could be obtained from human ES cell clones. Human ES cells differentiate into
spontaneously beating cells with a cardiomyocyte phenotype. When transplanted into
infarcted myocardium, ES cell derived cardiomyocytes engraft and improve cardiac function
in several rodent models (Yang et al., 2002; Min et al., 2002) (figure-2). In view of the risks
associated with the broad differentiation potential of ESCs in vivo, only sporadic reports
employed these cells in their uncommitted state to repair the damaged heart (Hodgson et al.,
2004; Kofidis et al., 2004; Kofidis et al., 2005). The differentiation of ESCs into
cardiomyocytes has been attributed to the paracrine effects of host signals, including
members of the TGF-β superfamily of proteins (Behfar et al., 2002; Kofidis et al., 2005a).

Some degree of cardiomyogenic commitment was claimed to enhance engraftment of the


cells into the myocardium, attenuate the probability of ESCs to acquire undesired cell
lineages, and thereby reduce the risk of teratoma formation (Tzukerman et al., 2003).
Although immature myocytes have been obtained from ESCs and their morphological,
phenotypical, and functional properties characterized (Hess et al., 2003; Kehat et al., 2004),
the residual growth potential of the committed cells was not established. There are no
rigorous methodologies for ESC differentiation into cardiomyocytes in vitro. It is emblematic
that eight distinct growth factors have forced ESCs to acquire only in part a specific cell
phenotype while other undesired cells were consistently present in the preparations
(Benvenisty, 2000).

92
Annual Review & Research in Biology, 1(4): 79-110, 2011

Fig. 2. Cardiac repair by different stem cells


(A): Embryonic stem cell generates the cardiomyocytes and improves cardiac function in infarcted heart. (B): Human adult bone
marrow stem cells generate the myocytes, endothelial stem cells and smooth muscle cells in ischemic myocardium. (C): Cardiac
stem cell forms the cardiocyetes and coronary vessels in the infarcted heart. (D): Mesenchymal stem cells (MSC) differentiate into
cardiomyocytes and endothelial cells in vivo when transplanted to the injured heart and cured the infracted heart. (E): The umbilical
cord blood stem cell homed to the infarcted hearts; it reduced infarct size, and enhanced neovascularization with capillary endothelial
cells of human.

93
Annual Review & Research in Biology, 1(4): 79-110, 2011

3.3.2 Human adult bone marrow–derived stem cells

Bone marrow elements contribute to cardiac repair in the infarcted heart served as the
rationale for adult bone marrow cell therapy after myocardial infarction (MI). The main
attraction of bone marrow-derived stem cell transplantation for scientists remains their innate
plasticity. Proof of principal has been shown by trans-differentiating marrow stromal cells into
cardiomyocytes in vitro. This has mainly been accomplished by treating cells with chemicals
promoting DNA demethylation, such as 5-aza-deoxycytidine (Kuethe et al., 2004). Orlic and
coworkers (2001) have shown that lineage-negative, c-kit–positive bone marrow–derived
cells differentiate into new cardiomyocytes after MI. Murry and colleagues (2004) reported
that lineage-negative, c-kit++ cells did not differentiate into cardiomyocytes. Despite the
conflicting evidence regarding the ability of bone marrow derived cells to transdifferentiate,
their efficacy in preventing remodeling has been demonstrated in many laboratories;
therefore, clinical efficacy trials have progressed in parallel with ongoing mechanistic
laboratory trials to determine the precise molecular mechanism by which these cells exert
their beneficial effects.

In the past 5 years, bone marrow mononuclear cells (BMMNC) transplantation has become
the most widely studied cell-based therapy for human applications. These experiments
demonstrated the capacity of adult BMSCs to give rise to new myocytes, endothelial cells,
and smooth muscle cells in ischemic myocardium. Moreover, in murine models of bone
marrow stem cell (BMSC) therapy it is extremely difficult to assess the specific effects of
myocyte versus nonmyocyte (e.g., endothelial cell) differentiation on the functional
improvement in myocardial performance after injury. Therefore, we must be cautious in
attributing benefit to a specific cell when the bone marrow contains cells of multiple lineage
and phenotype, not to mention the host of potential regulatory factors released by these
cells. Indeed, the whole concept of bone marrow hematopoietic stem cell (HSC) plasticity, at
least recently, has been questioned by experiments that demonstrate that single labeled
HSCs injected into lethally irradiated mice reconstitute peripheral blood leukocytes, but do
not contribute appreciably to nonhematopoietic tissues (Wagers et al., 2002). Further
information is also necessary on the homing and organ-specific differentiation signals
required for various stem cells, and this includes characterization of integrin and other
adhesion molecule structure/function relationships on these cells. This creates a conceptual
hurdle and technical challenge when trying to assess the therapeutic potential of bone
marrow stem cell therapy for human myocardial regeneration. For instance, without knowing
the contribution, if any, of human bone marrow to cardiomyocyte formation in vivo, clinical
trials of these cells seem premature. Currently, we do not have sufficiently specific bone
marrow stem cell markers to allow study of stem cell activity in human hearts of
nontransplanted subjects, an area that requires further investigation. However, it is likely that
future intense efforts to characterize unique stem cell surface markers will make such
analysis possible.

3.3.3 Cardiac stem cells

Several laboratories have recognized undifferentiated cells that are clonogenic, express
stem and endothelial progenitor cell (EPC) antigens/markers, and appear to have the
properties of adult cardiac stem cells in the adult heart of small and large mammals,
including humans. These cells most likely mediate endogenous mechanisms for minor repair
and for replacement of ongoing cell turnover within the adult heart. Cardiac progenitor cells
are multipotent in vitro and give rise to cardiomyocytes and coronary vessels in vivo (figure-
2), thus possessing the fundamental properties of stem cells (Beltrami et al., 2003; Oh et al.,

94
Annual Review & Research in Biology, 1(4): 79-110, 2011

2003; Messina et al., 2004; Linke et al., 2005; Pfister et al., 2005; Tomita et al., 2005; Dawn
et al., 2005). This discovery has provided the missing link between the documentation of
small dividing myocytes (Anversa et al., 2006; Levy et al., 2005) and the uncertainty
concerning the origin of these repopulating cells, and it has laid the groundwork for the
possibility of introducing the use of these cells in the treatment of the failing human heart
(Levy et al., 2005; Dawn et al., 2005). Messina and coworkers (2004) reported on the
identification of cardiospheres, clusters of self-adherent cells that grew from cultured adult
cardiac tissue derived from both human and murine hearts. These cells were shown to be
clonogenic and capable of transdifferentiation in vitro, and they induced both myocardial and
vascular regeneration after MI.

Cardiac stem cells can be harvested from patients and expanded ex vivo to generate large
numbers of cells. A recent report by Urbanek et al. (2005) demonstrated that cardiac stem
cells increase in number immediately after MI, but in the chronic phase, the numbers fall,
and the remaining cardiac stem cells have less regenerative potential. To date, there are no
clinical trials of human cardiac stem cells. However, Smith et al. (2005) demonstrated that
cardiospheres could be grown from human endomyocardial biopsy specimens. These
cardiospheres represent an easily accessible option for autologous stem cell therapy,
making the possibility of clinical testing of this approach feasible. The Specialized Centers
for Cell-Based Therapy initiative of the NHLBI has funded clinical trials of cardiac stem cells
that should begin in the near future. Skeletal myoblasts or satellite cells are the reservoir of
regenerative cells for skeletal muscle tissue; they have the ability for self-renewal and
differentiation if muscle injury occurs (Kuethe et al., 2005). They have many desirable
features as donor cells, including the ability to be amplified in an undifferentiated state in
vitro and high resistance to tissue ischemia. A growing body of experimental data and initial
clinical studies has shown not only engraftment of donor cells but also improvement in global
cardiac pump function.

3.3.4 Mesenchymal stem cells

MSCs differentiate into cardiomyocytes and endothelial cells in vivo when transplanted to the
heart in both noninsured and MI models (Mangi et al., 2003; Davani et al., 2003; Chen et al.,
2008) (Figure-2). These results suggest that MSCs act by regenerating functionally effective,
integrated cardiomyocytes and possibly new blood vessels. MSCs also have been injected
into infarcted myocardium via catheter-based approach in pigs, resulting in regeneration of
myocardium, reduced infarct size, and improved regional and global cardiac contractile
function (Amado et al., 2005). Importantly, the latter study used allogeneic MSCs, which did
not produce evidence of rejection, whereas autologous cell-based therapy poses no risk of
rejection, an “off the shelf” allogeneic cell product would be much more cost effective and
much easier to administer and could potentially allow delivery of greater numbers of cells
than autologous cell therapy. As such, MSCs may allow allogeneic cell therapy while
avoiding rejection (Sheyn et al., 2010).

3.3.5 Umbilical cord blood stem cells

Several populations of cells derived from UCB are the possible sources of stem cells for
cardiac repair. Kogler and colleagues (2004) have described a population of cells from
human UCB called unrestricted somatic stem cells. Human unrestricted somatic stem cells
(Kim et al., 2005) when delivered by direct injection at thoracotomy in immunosuppressed
pigs after MI, improved perfusion and wall motion, reduced infarct scar size, and enhanced
global cardiac function. Ma et al. (2005) injected human mononuclear UCB cells, a small

95
Annual Review & Research in Biology, 1(4): 79-110, 2011

fraction (1%) of which were CD34+, intravenously 1 day after MI in NOD/scid mice. The cells
homed to the infarcted hearts, reduced infarct size, and enhanced neovascularization with
capillary endothelial cells of both human and mouse origin (Figure 2). Intramyocardial
injections of MSCs are used to treat a variety of cardiovascular diseases as well as non-
cardiovascular conditions (Seres and Hollands, 2010). The clinical success of cell therapy
can be advanced if two seemingly unrelated tasks are undertaken: unification of clinical
protocols and tweaking of gene modification to meet the specific disease at hand.

3.4 Stem Cells in Orthopedic Research

Despite the natural and physiological regenerative capacity of bone, large bone defects,
such as those observed after bone tumor resection and severe fracture, lack a template for
orchestrated regeneration and require bone grafting. Approximately 5–10% of all fractures
are associated with impaired healing, resulting in significant patient morbidity, psychological
stress, and economic cost to society. After bone injuries, several molecular mechanisms
establish bone repair from stem/progenitor cells. Inflammation factors attract regenerative
cells which expand and differentiate in order to build up a bone highly similar to that before
injury. Bone marrow (BM) mesenchymal stem cells (MSCs) as skeletal stem cells and
endothelial progenitors (EPCs) are at the origin of such reparation mechanisms
(Deschaseaux et al., 2010). Novel developments in total joint arthroplasty and recent
progress in understanding the plasticity of stem cells and in applying knowledge to the
design of constructs to repair tissue are also described. The multidisciplinary approach to
understanding the mechanisms of tissue degradation and the development of therapeutics
for eliciting a reparative response is leading to the development of novel therapeutic
strategies. Furthermore, exciting advances in the manufacture and characterization of
scaffolds, combined with the emerging availability of multipotential stem cells, likely will lead
to important advances in efforts to engineer replacement musculoskeletal tissues (Sheyn et
al., 2010).

Several studies demonstrated the existence of cells (specifically, cells in tendon and
ligament) with multilineage differentiation potential. Traditionally, it has been assumed that
differentiated cells in tendon and ligament have a stable phenotype. Lee et al. (2006a)
reported that cells derived from synovial fluid in knees with anterior cruciate ligament injury
can differentiate into osteoblasts, adipocytes, and chondrocytes. Steiner et al. (2006)
reported that cells derived from culture specimens of the anterior cruciate ligament obtained
at the time of anterior cruciate ligament reconstructive surgery also have multilineage
differentiation potential. In that study, the investigators were careful to remove the synovial
covering over the torn anterior cruciate ligament in order to obtain anterior cruciate ligament
cells; nonetheless, the possibility remains that synovial-derived cells had infiltrated the torn
anterior cruciate ligament after injury. De Mos et al. (2006) reported that cells derived from
human tendon also have multilineage differentiation potential, which may explain the findings
of increased glycosaminoglycan deposition, calcifications, and lipid accumulation in
degenerative tendon. Additional support for the potential of stem cells to improve meniscal
healing was provided by a study involving the injection of synoviumderived stem cells
labeled with green fluorescent protein into the knees of wild-type rats that had a full-
thickness defect in the meniscus (Mizuno et al., 2006). The transplanted cells remained in
the meniscal defect and promoted healing in comparison with untreated meniscal defects.

Bone regeneration is a complex biological process involving a well-coordinated interplay


between different local or systemic soluble factors, extracellular matrix, MSCs and EPCs.
While there is compelling evidence that ex vivo expanded MSCs can effectively repair critical

96
Annual Review & Research in Biology, 1(4): 79-110, 2011

bone defects, this has not be proven as yet for native MSCs in either animal or human
models, due to the lack of a consensus regarding their phenotypic properties, and also
because convincing data about their perivascular anatomic location have only recently
started to emerge. Likewise, although several studies have reported the important role of
EPCs in bone healing, there is an ongoing controversy regarding the different populations
isolated in mice and human beings in terms of their characteristics and their potential
functional and/or phenotypic overlap (Deschaseaux et al., 2010). Therefore, although recent
advances on the field favour the potential of MSCs and EPCs in bone regeneration
considerable research needs still to be done to unravel the biology of these cells in bone
turnover.

3.5 Stem Cell in Cancer Therapies

Several in vitro and in vivo studies carried out with variety of cancer cell line types and on
different animal models to discover new therapeutic targets to block the growth and/or
survival of the cancer cells. Stem cell transplantation may also constitute an option as
adjuvant therapy for cancer, particularly in the patients receiving high doses of
chemotherapeutic agents and/or radiation that, along with killing cancer cells cause the
severe damage to normal tissues and/or destroy the hematopoietic cells. Thus, the stem cell
transplants might replace the endogenous stem cells destroyed by high-dose cancer
treatment, thereby producing healthy hematopoietic cell lineages and improving the immune
system defense (Phatak et al., 2007; Knoepfler, 2009).

The autologous or allogeneic transplantation of UCB, and BM stem cells and their
progenitors might be effectuated in combination with HDCT for numerous aggressive cancer
forms to replace BM and blood-forming cells that have been destroyed by chemotherapy.
AML and high-grade lymphoma are among the principal types of cancer that are usually
treated with hematopoietic cell support as adjuvant therapy (Wang et al., 2005; Aoudjhane et
al., 2005). The different subtypes of AML appear to result from distinct mutations at the level
of HSCs, the appearance of which may give rise to primitive leukemic stem cells (LSCs)
possessing a specific phenotype, such as CD90-, CD117-, and CD123+ (Cui et al., 2003).
These malignant LSCs are able to self-renew, might generate a heterogeneous AML cell
population, thereby maintaining leukemic blasts (Wang et al., 2005; Hope et al., 2004).
Interestingly, it is also proposed that the maintenance of LSCs in quiescent status might
contribute to their survival after chemotherapeutic treatment and leukemia relapse
(Senugupta et al 2007). Moreover, the ex vivo expansion of HSCs or the mobilization of
HSCs from BM into the peripheral blood by using mobilizing agents such as G-CSF and
AMD3100 might also lead to a great number of stem cells and their progenitors in
bloodstream, thereby decreasing the recovery time after HDCT (De Clercq et al., 2005).

The differentiated HSC-derived progenitors, such as dendritic cells, which are among the
most efficient cells of the immune system in presenting an antigen to helper/cytotoxic T
lymphocytes, might also be used as adjuvant treatment in cancer immunotherapy to
eliminate the neoplastic cells that express immunogenic antigens at their surface (Perillo et
al., 2004; den Brok et al., 2005). Furthermore, UCB also contains a substantial amount of
CD16-/CD56+ natural killer cells that might be expanded in the presence of IL-12 or IL-15
and that show a high rate of proliferation and cytotoxic effects against some cancers,
particularly leukemia (Cohen et al., 2004). In addition, the chemoprotection against
myelotoxicity induced by HDCT may also be counteracted by genetic manipulations in HSCs
conferring to their progenitor’s resistance to certain cytotoxic effects of drugs, such as the
expression of MDR1.

97
Annual Review & Research in Biology, 1(4): 79-110, 2011

3.6 Stem Cells in the Treatment of Diabetes

Type I diabetes represents a major disease entity that has tremendous appeal as a target for
cell replacement therapy. Embryonic stem cells (ESCs) have been studied based on their
multipotential capacity. It has been reported that insulin-producing cells could differentiate
from murine (Soria et al., 2000; Lumelsky et al., 2001) and human ESCs in vitro (Assady et
al., 2001; Segev et al., 2004). The regenerative property of ESCs has been further
evidenced by the results that transplantation of ESC-derived cells normalized or ameliorated
elevated blood glucose levels in diabetic mice (Soria et al., 2000; Hori et al., 2002; Blyszczuk
et al., 2003). The generation of ES-derived insulin-producing pancreatic endocrine cells may
be critical to the treatment of diabetes (Mfopou et al., 2007). This enabled lineage selection
and maturation of insulin-expressing cells, upon transplantation, resulted in the normalization
of glycemia in streptozotocin-induced diabetic mice (Soria et al., 2000). In contrast, the
spontaneous differentiation of mES cells in vitro generated only a small fraction of cells
(0.1%) with characteristics of insulin-producing β-like cells (Shiroi et al., 2002). The insulin-
positive clusters, however, failed to normalize high blood glucose levels in transplantation
experiments (Lumelsky et al., 2001). Rather than producing insulin themselves, most of the
cells took up this hormone from the culture medium (Rajagopal et al., 2003; Singh and
Williams, 2008).

By modifying the differentiation protocols and using genetically modified mES cells, two
groups successfully generated insulin-producing cells (Blyszczuk et al., 2003; Leon-Quinto
et al., 2004). Blyszczuk et al., (2003) showed that constitutive expression of the pancreatic
developmental control gene Pax4 and histotypic differentiation were essential for the
formation of insulin expressing cells, which were found to contain secretory granules typical
of both embryonal and adult β-cells. Importantly, these cells coexpressed C-peptide and
normalized blood glucose levels after transplantation into diabetic mice (Mfopou et al., 2007;
Blyszczuk et al., 2004). Similarly, lineage selection using mES cells transfected with a
plasmid containing the Nkx6.1 promoter upstream of a neomycin-resistance gene could be
used to generate insulin-producing cells that normalized glycemia after transplantation into
diabetic animals (Leon-Quinto et al., 2004). Also treatment of mES cells with a
phosphoinositide 3-kinase (PI 3-K) inhibitor during terminal stages of differentiation
generated ES cell progeny expressing various β-cell specific markers. Following engraftment
into diabetic mice, these cells also improved the glycemic status and enhanced animal
survival (Hori et al., 2002). Initial experiments with hES cells indicate that in vitro
differentiation generates 1% insulin-secreting cells that show at least some characteristics of
pancreatic endocrine cells (Assady et al., 2001). A modification of the differentiation protocol
(Lumelsky et al., 2001, Rajagopal et al., 2003) allowed the generation of insulin producing
clusters from hES cells (Segev et al., 2004), but further improvements are necessary for
generating functional islet like cells. These reports are provocative, but much additional
work remains to characterize the functional nature of the cells as glucose regulators, and to
document adequate, regulated production of insulin, which in one case was some 50 - fold
less than native β cells (Lumelsky et al., 2001). Indeed, some of these reports have been
called into question by subsequent studies showing that apoptotic cells can take up insulin
from the culture medium and give the illusion of producing insulin without actually doing so
(Rajagopal et al., 2003).

Some difficulties in generating beta cells from ES cells may stem from attempts to apply
factors characteristic of late pancreatic development to essentially very early-stage cells.
Real clinical impact awaits the clear directed differentiation of appropriate cell populations.
Since the end of the 20th century, bone marrow–derived cells have been reported to give

98
Annual Review & Research in Biology, 1(4): 79-110, 2011

rise to endodermal-origin cells (Petersen et al., 1999; Krause et al., 2001). In the pancreatic
tissue, several reports described the regeneration of bone marrow–derived pancreatic beta
cells based on mouse syngeneic or allogeneic transplantation assay. Ianus et al., (2003)
first suggested the contribution of bone marrow–derived cells to generate insulin-producing
cells. Hess et al. (2003) further demonstrated the improvement of blood glucose levels
following bone marrow transplantation using chemically induced diabetic mice. Although
donor bone marrow–derived insulin-producing cells were present in the recipient mice, the
authors suggested that the improved glucose levels in diabetic recipient mice were due to
the regeneration of host-derived beta cells rather than that of donor bone marrow–derived
insulin-producing cells as evidenced by increased numbers of BrdU-labeled green
fluorescent protein (GFP) - insulin+ cells, not GFP+ insulin+ cells at 4–7 days after
transplantation. Lanus et al. (2003) reported the donor (Ins2-Cre mice) bone marrow–
derived insulin-producing cells in recipient (Rosa-lox-GFP mice) pancreatic tissue, which
could likely be generated through a fusion-independent mechanism. On the other hand,
bone marrow–derived stem cells contributed to the regeneration of other endodermal tissue–
derived cells, such as hepatocytes, through cell fusion (Terada et al., 2002; Alvarez-Dolado
et al., 2003). Regeneration of pancreatic tissue is a complex mission due to the complicated
structure and function of the tissue. An enhanced understanding of the genes involved in
pancreatic differentiation, better control of insulin levels, and successful transplantation of
islet cells will dramatically improve the care of patients with diabetes.

4. FUTURE PROSPECTIVES

Studies of human ES cells have demonstrated an enormous potential for generating tissues
of therapeutic value. Adult stem cells (ASC) can be coaxed into differentiated cells not
normally associated with their “committed” state. The stimulation of endogenous stem cells
is based on the possibility that self-repair could be induced or augmented by stimulating the
patient’s own stem cells by administrating growth factors. Bone marrow cells, for example,
can be mobilized by stem cell factor and granulocyte- colony stimulating factor. In the case
of myocardial infarction, these mobilized cells seem to be home to an infarcted region to
promote myocardial repair. It is currently unclear whether the activation process or the
release of factors from activated stem cells is more important to this therapeutic approach. A
recent study showed that transplantation of adult bone marrow-derived cells reduces
hyperglycemia in diabetic mice by initiating endogenous pancreatic tissue regeneration.
Engraftment of bone marrow-derived cells to ductal and an islet structure was accompanied
by rapid proliferation of recipient pancreatic cells and neogenesis of insulin-producing cells
of recipient origin. This strategy may represent a previously unrecognized means by which
bone marrow derived cells can contribute to tissue restoration. Many potential endogenous
stem cell sources (liver, brain, skin, heart, bone marrow, and intestine) are now recognized
to be present in humans. Stimulation of endogenous sources of stem cells is currently only
achievable from bone marrow. With the rapid advance of stem cell research, it is likely,
however, that further advances will be made so that endogenous supplies can be mobilized
to more readily repair and replace damaged tissues following injury. We still have many
challenges ahead of us but researchers, clinicians, regulators and healthcare practitioners all
recognise the immense potential of this nascent field and wish to provide solutions rather
than hurdles. I hope one can look forward to many years of exciting research with immense
clinical potentials and to be able to provide, in years to come, many solutions to unmet
clinical needs.

99
Annual Review & Research in Biology, 1(4): 79-110, 2011

REFERENCES

Abdelkrim, H., Juan, D.B., Jane, W., Mohamed, A., Bernat, S. (2009). The immune
boundaries for stem cell based therapies: problems and prospective solutions. J. Cell.
Mol. Med., 13, 1464–1475
Adewumi, O., Aflatoonian, B., Ahrlund-Richter, L., et al. (2007). Characterization of human
embryonic stem cell lines by the International StemCell Initiative. Nature
Biotechnology, 25, 803–816.
Alvarez-Dolado, M., Pardal, R., Garcia-Verdugo, J.M., et al. (2003). Fusion of bone marrow-
derived cells with Purkinje neurons, cardiomyocytes and hepatocytes. Nature, 425,
968–973.
Amado, L.C., Saliaris, A.P., Schuleri, K.H., St. John, M., Xie, J.S., Cattaneo, S., Durand,
D.J., Fitton, T., et al. (2005). Cardiac repair with intramyocardial injection of allogeneic
mesenchymal stem cells after myocardial infarction. Proc. Natl. Acad. Sci. USA., 102,
11474 –11479.
Amit, M., Margulets, V., Segev, H., Shariki, K., Laevsky, I., Coleman, R., Itskovitz-Eldor, J.
(2003). Human feeder layers for human embryonic stem cells. Biol. Reprod., 68,
2150–2156.
Anderson, W.S., Lenz, F.A. (2006). Surgery insight: deep brain stimulation for movement
disorders. Nat. Clin. Pract. Neurol., 2(6), 310–320.
Anversa, P., Kajstura, J., Leri, A., et al. (2006). Life and death of cardiac stem cells. A
paradigm shift in cardiac biology. Circulation, 113, 1451–1463.
Aoudjhane, M., Labopin, M., Gorin, N.C., et al. (2005). Comparative outcome of reduced
intensity and myeloablative conditioning regimen in HLA identical sibling allogeneic
haematopoietic stem cell transplantation for patients older than 50 years of age with
acute myeloblastic leukaemia: A retrospective survey from the Acute Leukemia
Working Party (ALWP) of the European group for Blood and Marrow Transplantation
(EBMT). Leukemia, 19, 2304 –2312.
Assady, S., Maor, G., Amit, M., Itskovitz-Eldor, J., Skorecki, K.L., Tzukerman, M. (2005).
Insulin production by human embryonic stem cells. Diabetes, 50, 1691–1697.
Baizabal, J.M., Magaril, M.F., Jesu, S.O., Covarrubias, L. (2003). Neural Stem Cells in
Development and Regenerative Medicine. Archives of Medical Research, 34, 572–
588.
Bajada, S., Mazakova, I., Richardson, J.B., Ashammakhi, N. (2008). Updates on stem cells
and their applications in regenerative medicine. J. Tissue Eng. Regen. Med., 2, 169–
183.
Barker, J.N., Wagner, J.E. (2003). Umbilical cord blood transplantation: current practice and
future innovations. Crit. Rev. Oncol. Hematol., 48, 35 - 43.
Becker, A.J., McCulloch, E.A., Till, J.E. (1963). Cytological demonstration of the clonal
nature of spleen colonies derived from transplanted mouse marrow cells. Nature, 197,
452–454.
Behfar, A., Zingman, L.V., Hodgson, D.M., Rauzier, J.M., et al. (2002). Stem cell
differentiation requires a paracrine pathway in the heart. FASEB J., 16, 1558–1566.
Behrstock, S., Ebert, A., McHugh, J., Vosberg, S., Moore, J., et al. (2006). Human neural
progenitors deliver glial cell line-derived neurotrophic factor to parkinsonian rodents
and aged primates. Gene Ther., 13, 379–388
Beltrami, A.P., Barlucchi, L., Torella, D., et al. (2003).Adult cardiac stem cells are multipotent
and support myocardial regeneration. Cell, 114, 763–776.

100
Annual Review & Research in Biology, 1(4): 79-110, 2011

Benchaouir, R., Meregalli, M., Farini, A., D’Antona, G. (2007).Restoration of human


dystrophin following transplantation of exon-skipping-engineered DMD patient stem
cells into dystrophic mice. CellStem Cell, 1(6), 646–657.
Ben-Hur, T., Idelson, M., Khaner, H., Pera, M., et al. (2004). Transplantation of human
embryonic stem cell derived neural progenitors improves behavioural deficit in
Parkinsonian rats. Stem Cells, 22, 1246–1255
Benvenisty, N, (2000). Effects of eight growth factors on the differentiation of cells derived
from human embryonic stem cells. Proc. Natl. Acad. Sci. USA., 97, 11307–12.
Bjornson, C.R., Rietze, R.L., Reynolds, B.A., Magli, M.C., Vescovi, A.L. (1999). Turning
brain into blood: a hematopoietic fate adopted by adult neural stem cells in vivo.
Science, 283, 534-537.
Blanpain, C., Lowry, W.E., Geoghegan, A., Polak, L., Fuchs, E. (2004). Self-renewal,
multipotency, and the existence of two cell populations within an epithelial stem cell
niche. Cell, 118, 635–648.
Blyszczuk, P., Asbrand, C., Rozzo, A., Kania, G., et al. (2004). Embryonic stem cells
differentiate into insulin-producing cells without selection of nestin-expressing cells.
Int. J. Dev. Biol., 48, 1095–1104.
Bongso, A., Fong, C.Y., Ng, S.C., Ratnam, S. (1994). Isolation and culture of inner cell mass
cells from human blastocysts. Human Reproduction. 9, 2110-2117.
Bouwens, L., Rooman, I. (2005). Regulation of pancreatic beta-cell mass. Physiol. Rev. 85,
1255–1270.
Brignier, A.C., Gewirtz, A.M.. (2010). Embryonic and adult stem cell therapy. J. Allergy. Clin.
Immunol,, 25, S336-344.
Chamberlain, G., Fox, J., Ashton, B., Middleton, J. (2007). Concise review: mesenchymal
stem cells: their phenotype. Differentiation capacity, immunological features, and
potential for homing. Stem cells, 25, 2739–2749.
Chen, Y., Shao, J.Z., Xiang, L.X., Xong, X.J., Zhang, G.R. (2008). Mesenchymal stem cells:
A promising candidate in regenerative medicine. The International Journal of
Biochemistry & Cell Biology, 40, 815–820
Clarke, D.L., Johansson, C.B., Wilbertz, J., Veress, B., Nilsson, E., Karlstrom, H., et al.
(2000). Generalized potential of adult neural stem cells. Science, 288, 1660-1663.
Cohen, Y, Nagler, A. (2004). Umbilical cord blood transplantation—how, when and for
whom? Blood Rev., 18, 167–179.
Cui, J.W., Zhang, X.M., Wang, G.J. (2003). Progress in the studies of acute myelogenous
leukemia stem cell. Circulation, 104, 1046 –1052.
Daar, A.S., Greenwood, H.L. (2007). A proposed definition of regenerative medicine. J.
Tissue Eng. Regen. Med., 1, 179–184.
Davani, S., Marandin, A., Mersin, N., Royer, B., Kantelip, B., et al. (2003). Mesenchymal
progenitor cells differentiate into an endothelial phenotype, enhance vascular density,
and improve heart function in a rat cellular cardiomyoplasty model. Circulation, 108,
253–258.
Dawn, B., Stein, A.B., Urbanek, K., et al (2005). Cardiac stem cells delivered intravascularly
traverse the vessel barrier, regenerate infarcted myocardium, and improve cardiac
function. Proc. Natl. Acad. Sci. USA., 102, 3766 –3771.
De Bari,, Dell’Accio, F., Tylzanowski, P., Luyten, F.P. (2001). Multipotent mesenchymal stem
cells from adult human synovial membrane. Arthritis Rheum., 44, 1928–1942.
De Clercq, E. (2005). Potential clinical applications of the CXCR4 antagonist bicyclam
AMD3100. Mini Rev. Med. Chem., 5, 805– 824.
De Coppi, P., Bartsch, G.Jr., Siddiqui, M.M., et al. (2007). Isolation of amniotic stem cell
lines with potential for therapy. Nat. Biotechnol., 25, 100–6.

101
Annual Review & Research in Biology, 1(4): 79-110, 2011

De Mos, M., Jahr, H., Weinans, H., Verhaar, J., Van Osch, G. (2006). A possible role for
tendon cell differentiation in the development of tendinosis. In: Transactions of the
52nd Annual Meeting of the Orthopedic Research Society; Chicago, IL.
den Brok, M.H., Nierkens, S., Figdor, C.G., et al. (2005). Dendritic cells: Tools and targets
for antitumor vaccination. Expert Rev. Vaccines., 4, 699 –710.
Dennis, J.E., Caplan, A.I. (2004). Bone Marrow Mesenchymal Stem Cells. In: Sell S (ed)
Stem Cells Handbook. Humana Press, Totowa, pp 107–118.
Deschaseaux, F., Pontikoglou, C., Luc, S. (2010). Bone regeneration: the stem/progenitor
cells point of view. J. Cell. Mol. Med., 14, 103-115
Dominici, M., Le Blanc, K., Mueller, I., et al. (2006). Minimal criteria for defining multipotent
mesenchymal stromal cells. The International Society for Cellular Therapy position
statement. Cytotherapy, 8, 315–317.
Duan, Y., Catana, A., Meng, Y., et al. (2007). Differentiation and enrichment of hepatocyte-
like cells from human embryonic stem cells in vitro and in vivo. Stem Cells, 25(12),
3058–3068.
Eckfeldt, C.E., Mendenhall, E.M., Verfaillie, C.M. (2005). The molecular repertoire of the
‘almighty’ stem cell. Nat. Rev. Mol. Cell Biol., 6, 726–737.
Edwards, R.G. (2004). Stem cells today: A. Origin and potential of embryo stem cells.
Reproductive biomedicine, 8, 275-306.
Epperly, M.W., Guo, H., Gretton, J.E., Greenberger, J.S. (2003). Bone marrow origin of
myofibroblasts in irradiation pulmonary fibrosis. Am. J. Respir. Cell Mol. Biol., 29,
213–224.
Evans, M.J., Kaufman, M.H. (1981). Establishment in culture of pluripotential cells from
mouse embryos. Nature, 292, 154-156.
Fabien, L.G., Rudnicki, M.A. (2007). Skeletal muscle satellite cells and adult myogenesis.
Current Opinion in Cell Biology, 19, 628–633.
Fahn, S. (2003). Description of Parkinson’s disease as a clinical syndrome. Ann. N. Y. Acad.
Sci., 991, 1–14.
Fang, D., Nguyen, T.K., Leishear, K., et al. (2005). A tumorigenic subpopulation with stem
cell properties in melanomas. Cancer Res., 65, 9328 –9337.
Ferrari, G., Cusella-De, A.G., Coletta, M., Paolucci, E., Stornaiuolo, A., Cossu, G., Mavilio,
F. (1998). Muscle regeneration by bone marrow-derived myogenic progenitors.
Science, 279, 1528–1530.
Fine, A. (2004). Marrow cells progenitors of lung tissue. Blood Cells Mol. Dis., 32, 95–96.
Flores, I., Cayuela, M,L., Blasco, M.A. (2005). Effects of telomerase and telomere length on
epidermal stem cell behavior. Science, 309, 1253–1256.
Frenck, R.W.Jr., Blackburn, E.H., Shannon, K.M. (1998). The rate of telomere sequence loss
in human leukocytes varies with age. Proc. Natl. Acad. Sci. USA., 95, 5607-5610.
Fujita, H., Shimizu, K., Nagamori, E. (2009). Application of a cell sheet-polymer film complex
with temperature sensitivity for increased mechanical strength and cell alignment
capability. Biotechnol. Bioeng., 103(2), 370–377.
Gage, F.H. (2000). Mammalian neural stem cells. Science, 287, 1433–1438.
Galli, R., Borello, U., Gritti, A., Minasi, M.G., Bjornson, C., Coletta, M., et al. (2000). Skeletal
myogenic potential of human and mouse neural stem cells. Nat. Neurosci., 3, 986-91.
Gardner, R.L. (2007). Stem cells and regenerative medicine: principles, prospects and
problems. C. R. Biol., 330(6-7), 465-473.
Gordon, M.Y. (2008). Stem cells for regenerative medicine Biological attributes and clinical
application. Experimental Hematology, 36, 726–732
Guenou, H., Nissan, X., Larcher, F., Feteira, J., Lemaitre, G., et al. (2009). Human
embryonic stem-cell derivatives for full reconstruction of the pluristratified epidermis: a
preclinical study. Lancet, 374, 1745–1753.

102
Annual Review & Research in Biology, 1(4): 79-110, 2011

Guillott, P.V., Cui, W., Fisk, N.M., Polak, J.M. (2007). Stem cell differentiation and expansion
for clinical applications of tissue engineering. J. Cell Mol. Med., 11(5), 935–44.
Harris, D.T., Badowski, M., Ahmad, N., et al. (2007). The potential of cord blood stem cells
for use in regenerative medicine. Expert Opin. Biol. Ther., 7, 1311–22.
Hashimoto, N., Jin, H., Liu, T., Chensue, S.W., Phan, S.H. (2004). Bone marrow–derived
progenitor cells in pulmonary fibrosis. J. Clin. Invest., 113, 243–252.
Hendricks, W.A., Pak, E.S., Owensby, J.P, et al. (2006). Predifferentiated embryonic stem
cells prevent chronic pain behaviors and restore sensory function following spinal cord
injury in mice. Mol. Med., 12(1–3), 34–46.
Hess, D., Li, L., Martin, M., et al. (2003). Bone marrow-derived stem cells initiate pancreatic
regeneration. Nat. Biotechnol., 21, 763–770.
Ho, H.Y., Li, M. (2006). Potential application of embryonic stem cells in Parkinson’s disease:
drug screening and cell therapy. Regen. Med., 1(2), 175–182.
Hodgson, D.M., Behar, A., Zingman, L.V., Kane, G.C., et al. (2004). Stable benefit of
embryonic stem cell therapy in myocardial infarction. Am. J. Physiol. Heart Circ.
Physiol., 287, H471–479.
Hollands, P. (2009). Cord blood stem cells – the basic science. In:Bhattacharya, N.,
Stubblefield, P. (Eds.), Frontiers of Cord Blood Science. Springer-Verlag, London, pp.
19–25.
Hope, K.J., Jin, L., Dick, J.E. (2004). Acute myeloid leukemia originates from a hierarchy of
leukemic stem cell classes that differ in self-renewal capacity. Nat. Immun., 5, 738–
743.
Hori, Y., Rulifson, I.C., Tsai, B.C., et al. (2002). Growth inhibitors promote differentiation of
insulin-producing tissue from embryonic stem cells. Proc, Natl, Acad, Sci, USA.,
99,16105–16110.
Ianus, A., Holz, G.G., Theise, N.D., et al. (2003). In vivo derivation of glucose competent
pancreatic endocrine cells from bone marrow without evidence of cell fusion. J. Clin.
Invest. 111, 843–850.
Itskovitz-Eldor, J., Schuldiner, M., Karsenti, D., Eden, A., et al. (2000). Differentiation of
human embryonic stem cells into embryoid bodies compromising the three embryonic
germ layers. Mol. Med., 6, 88–95.
Jackson, K.A., Mi, T., Goodell, M.A. (1999). Hematopoietic potential of stem cells isolated
from murine skeletal muscle. Proc. Natl. Acad. Sci. USA., 96, 14482–14486.
Jarvinen, T.A., Järvinen, T.L., Kääriäinen, M., et al. (2007). Muscle injuries: optimising
recovery. Best Pract. Res. Clin. Rheumatol., 21(2), 317–331.
Jones, R., Lebkowski, J., McNiece, I. (2010). Stem cells. Biol. Blood Marrow Transplant., 16:
S115-S118.
Jukes, J.M., van Blitterswijk, C.A., Boer, J. (2010). Skeletal tissue engineering using
embryonic stem cells. J. Tissue. Eng. Regen. Med., 4, 165–180.
Julia, D., Polak, M. (2009). Regenerative medicine: A primer for pediatricians. Early Human
Development, 85, 685–689
Kaufman, D.S., Hanson, E.T., Lewis, R.L., Auerbach, R., Thomson, J.A. (2001).
Hematopoietic colony-forming cells derived from human embryonic stem cells. Proc.
Natl. Acad. Sci. USA., 98, 10716–10721.
Kawasaki, H., Mizuseki, K., Nishikawa, S., Kaneko, S., Kuwana, Y., Nakanishi, S.,
Nishikawa, S.I., Sasai, Y. (2000). Induction of midbrain dopaminergic neurons from ES
cells by stromal cell-derived inducing activity. Neuron, 28, 31–40.
Kehat, I., Kenyagin, K.D., Snir, M., Segev, H., Amit, M., Gepstein, A., Livne, E., Binah, O.,
Eldor, J.I., Gepstein, L. (2001). Human embryonic stem cells can differentiate into
myocytes with structural and functional properties of cardiomyocytes. J. Clin. Invest.,
108, 407–14.

103
Annual Review & Research in Biology, 1(4): 79-110, 2011

Kehat, I., Khimovich, L., Caspi, O., Gepstein, A., Shofti, R., et al. (2004). Electromechanical
integration of cardiomyocytes derived from human embryonic stem cells. Nat.
Biotech., 10, 1–8.
Kim B-O, Tian, H., Prasongsukarn, K., Wu, J., Angoulvant, D., et al. (2005). Cell
transplantation improves ventricular function after a myocardial infarction: a preclinical
study of human unrestricted somatic stem cells in a porcine model. Circulation, 112,
96-104.
Knoepfler, P.S. (2009). Deconstructing Stem Cell Tumorigenicity: A Roadmap to Safe
Regenerative Medicine. Stem Cells, 27, 1050–1056
Kofidis, T., de Bruin, J.L., Hoyt, G., et al. (2005a). Myocardial restoration with embryonic
stem cell bioartificial tissue transplantation. J. Heart Lung Transpl., 24(6), 737–744.
Kofidis, T., deBruin, J.L., Yamane, T., Balsam, L.B., et al. (2004). IGF-1 promotes
engraftment, differentiation and functional improvement after transfer of embryonic
stem cells for myocardial restoration. Stem Cells, 22, 1239–1245.
Kofidis, T., deBruin, J.L., Yamane, T., Tanaka, M., et al. (2005b). Stimulation of paracrine
pathways with growth factors enhances embryonic stem cell engraftement and host-
specific differentiation in the heart after ischemic myocardial injury. Circulation, 111,
2486–2493.
Kofidis, T., Lebl, D.R., Swignenburg, R.J., Greeve, J.M., et al. (2006). Allopurinol/uricase and
ibuprofen enhance engraftment of cardiomyocyte-enriched human embryonic stem
cells and improve cardiac function following myocardial injury. Eur. J. Cardio-thoracic
Surgery, 29, 50–55.
Kogler, G., Sensken, S., Airey, J.A., Trapp, T., Muschen, M., et al. (2004). A new human
somatic stem cell from placental cord blood with intrinsic pluripotent differentiation
potential. J. Exp. Med., 200, 123–135.
Koh, L.P., Chao, N.J. (2004). Umbilical cord blood transplantation in adults using
myeloablative and nonmyeloablative preparative regimens. Biol. Blood Marrow
Transplant., 10, 1-22.
Krause, D.S., Theise, N.D., Collector, M.I., et al. (2001). Multi-organ, multi-lineage
engraftment by a single bone marrow-derived stem cell. Cell, 105, 369–377.
Kuethe, F., Figulla, H.R., Herzau, M., Voth, M., et al. (2005). Treatment with granulocyte
colony-stimulating factor for mobilization of bone marrow cells in patients with acute
myocardial infarction. Am. Heart J., 150, 115.
Lagasse, E., Connors, H., Al-Dhalimy, M., et al. (2000). Purified hematopoietic stem cells
can differentiate into hepatocytes in vivo. Nat. Med., 6, 1229– 1234.
Landry, D.W., Zucker, H.A. (2004.). Embryonic death and the creation ofhuman embryonic
stem cells. J. Clin. Invest., 114, 1184–1186.
Lanzendorf, S.E., Boyd, C.A., Wright, D.L., Muasher, S., Oehninger, S., Hodgen, G.D.
(2001). Use of human gametes obtained from anonymous donors for the production of
human embryonic stem cell lines. Fertil. Steril., 76, 132-137.
Lee, E.H., Hui, J.H.P. (2006). The potential of stem cells in orthopaedic surgery. J. Bone Jt.
Surg., 88(7), 841–853.
Lee, S.R., Kim, H.Y., Rogowska, J., Zhao, B.Q., et al. (2006). Involvement of matrix
metalloproteinase in neuroblast cell migration from the subventricular zone after
stroke. J. Neurosci., 26, 3491–3495.
Lee, S.Y., Miwa, M., Sakai, Y., Kuroda, R., Matsumoto, T., Kurosaka, M. (2006).
Mesenchymal stem cells can be obtained from human ACL injury-induced
hemathrosis of the knee. In: Transactions of the 52nd Annual Meeting of the
Orthopaedic Research Society; Chicago, IL.

104
Annual Review & Research in Biology, 1(4): 79-110, 2011

Lensch, M.W. (2009). Cellular reprogramming and pluripotency induction cellular. Br. Med.
Bull., 90, 19–35.
Leon-Quinto, T., Jones, J., Skoudy, A., Burcin, M., Soria, B. (2004). In vitro directed
differentiation of mouse embryonic stem cells into insulin-producing cells.
Diabetologia, 47, 1442–1451.
Levenberg, S., Golub, J.S., Amit, M., Itskovitz-Eldor, J., Langer, R. (2002). Endothelial cells
derived from human embryonic stem cells. Proc. Natl. Acad. Sci. USA., 99, 4391–
4396.
Levy, V., Lindon, C., Harfe, B.D., et al. (2005). Distinct stem cell populations regenerate the
follicle and interfollicular epidermis. Dev. Cell., 9, 855–861.
Lewitzky, M., Yamanaka, S. (2007). Reprogramming somatic cells towards pluripotency by
defined factors. Curr. Opin. Biotechnol., 18(5), 467-73.
Li, L., Xie, T. (2005). Stem cell niche: Structure and function. Annu. Rev. Cell. Dev. Biol., 21,
605– 631.
Liechty, K.W., MacKenzie, T.C., Shaaban, A.F., Radu, A., et al. (2000). Human
mesenchymal stem cells engraft and demonstrate site-specific differentiation after in
utero transplantation in sheep. Nat. Med., 6, 1282–1286.
Lindvall, O., Kokaia, Z., Martinez-Serrano, A. (2004). Stem cell therapy for human
neurodegenerative disorders—how to make it work. Nature Med., 10, S42–S50.
Linke, A., Muller, P., Nurzynska, D., et al. (2005). Stem cells in the dog heart are self-
renewing, clonogenic, and multipotent and regenerate infracted myocardium,
improving cardiac function. Proc. Nat. Acad. Sci. USA., 102, 8966–8971.
Liu, Y.H., Ravi, K., Wu, S.M. (2008). Cardiovascular stem cells in regenerative medicine:
ready for prime time? Drug Discovery Today: Therapeutic Strategies, 5(4), 201.
Longaker, M.T. (2010). Regenerative medicine: a surgeon's perspective. J. Pediatric Surg.,
45, 11–18.
Lumelsky, N., Blondel, O., Laeng, P., et al (2001). Differentiation of embryonic stem cells to
insulin-secreting structures similar to pancreatic islets. Science, 292, 1389–1394.
Ma, M., Sha, C., Zhou, Z., Zhou, Q., Li, Q. (2008). Generation of patient-specific pluripotent
stem cells and directed differentiation of embryonic stem cells for regenerative
medicine. Journal of Nanjing Medical University, 22(3), 135-142
Ma, N., Stamm, C., Kaminsk,i A., Li, W., Kleine, H.D., Muller-Hilke, B., et al. (2005). Human
cord blood cells induce angiogenesis following myocardial infarction in NOD/scid-mice.
Cardiovasc. Res., 66, 45–54.
Mangi, A.A., Noiseux, N., Kong, D., He, H., et al. (2003). Mesenchymal stem cells modified
with Akt prevent remodeling and restore performance of infarcted hearts. Nat. Med.,
9,1195–1201.
Martin, G.R. (1981). Isolation of a pluripotent cell line from early mouse embryos cultured in
medium conditioned by teratocarcinoma stem cells. Proc. Natl. Acad. Sci. USA., 78,
7634-7638.
Martin, M.J., Muotri, A., Gage, F., Varki, A. (2005). Human embryonic stem cells express an
immunogenic nonhuman sialic acid. Nat. Med., 11, 228–232.
Messina, E., De Angelis, L., Frati, G. et al. (2004). Isolation and expansion of adult cardiac
stem cells from human and murine heart. Circ. Res., 95, 911–921.
Mezey, E., Chandross, K.J., Harta, G., Maki, R.A., McKercher, S.R. (2000). Turning blood
into brain: cells bearing neuronal antigens generated in vivo from bone marrow.
Science, 290, 1779–1782.
Mfopou, J.K., De Groote, V., Xu, X., Heimberg, H., Bouwens, L. (2007). Sonic hedgehog and
other soluble factors from differentiating embryoid bodies inhibit pancreas
development. Stem Cells, 25, 1156–1165.

105
Annual Review & Research in Biology, 1(4): 79-110, 2011

Miki, T., Lehmann, T., Cai, H., et al (2006). Stem cell characteristics of amniotic epithelial
cells. Stem cells, 23, 1549 –1559.
Millar, S.E. (2005). An ideal society? Neighbors of diverse origins interact to create and
maintain complex mini-organs in the skin. PLoS Biol., 3, 1873–1877.
Min, J.Y., Yang, Y., Sullivan, M.F., Ke, Q., Converso, K.L., et al. (2003). Long-term
improvement of cardiac function in rats after infarction by transplantation of embryonic
stem cells. J. Thorac. Cardiovasc. Surg., 125, 361–369.
Mizuno, H., Roy, A.K., Zaporojan, V., Vacanti, C.A., Ueda, M., Bonassar, L.J. (2006).
Biomechanical and biochemical characterization of composite tissue-engineered
intervertebral discs. Biomaterials, 27, 362-70.
Morris, R.J., Liu, Y., Marles, L., et al. (2004). Capturing and profiling adult hair follicle stem
cells. Nat. Biotechnol., 22, 411– 417.
Morrison, S.J., Shah, N.M., Anderson, D.J. (1997). Regulatory mechanisms in stem cell
biology. Cell. 88, 287–298.
Murry, C.E., Wiseman, R.W., Schwartz, S.M., Hauschka, S.D. (2004). Skeletalmyoblast
transplantation for repair of myocardial necrosis. J. Clin. Invest., 98, 2512–2523.
NRC (National Research Council) (2002). Stem Cells and the Future of Regenerative
Medicine Committee on the Biological and Biomedical Applications of Stem Cell
Research, Board on Life Sciences, National Research Council, Board on
Neuroscience and Behavioral Health, Institute of Medicine. ISBN: 0-309-50974-2, 112
pages, NATIONAL ACADEMY PRESS, Washington, D.C.
http://www.nap.edu/catalog/10195.html
Nicol, S., Margit Rosner, Michaela Hanneder, Alessandro Valli, Markus Hengstschläger
(2007). Stem Cells in Amniotic Fluid as New Tools to Study Human Genetic Diseases.
Stem Cell Rev., 3, 256–264.
Noth, U., Osyczka, A.M., Tuli, R., et al. (2002). Multilineage mesenchymal differentiation
potential of human trabecular bone-derived cells. J. Orthop. Res., 20, 1060–1069.
Oh, H., Bradfute, S.B., Gallardo, T.D., et al (2003). Cardiac progenitor cells from adult
myocardium: Homing, differentiation, and fusion after infarction. Proc. Natl. Acad. Sci.
USA., 100, 12313–12318.
Olguin, H.C., Olwin, B.B. (2004). Pax-7 up-regulation inhibits myogenesis and cell cycle
progression in satellite cells: A potential mechanism for self-renewal. Dev. Biol., 275,
375– 388.
Orlic, D., Kajstura, J., Chimenti, S., Limana, F., Jakoniuk, I., et al. (2001). Mobilized bone
marrow cells repair the infarcted heart, improving function and survival. Proc. Natl.
Acad. Sci. USA., 98, 10344 –10349.
Pereira, R.F., O'Hara., Laptev, A.V., et al. (1995). Cultured adherent cells from marrow can
serve as long-lasting precursor cells for bone, cartilage, and lung in irradiated mice.
Proc. Natl. Acad. Sci. USA., 92, 4857–4861.
Perillo, A., Bonanno, G., Pierelli, L., et al. (2004). Stem cells in gynecology and obstetrics.
Panminerva Med., 46, 49 –59.
Petersen, B.E., Bowen, W.C., Patrene, K.D., et al. (1999). Bone marrow as a potential
source of hepatic oval cells. Science, 284, 1168–1170.
Pfister, O., Mouquet, F., Jain, M., et al (2005). CD31- but not CD31+ cardiac sidepopulation
cells exhibit functional cardiomyogenic differentiation. Circ. Res., 97, 52– 61.
Phatak, P., Cookson, J.C., Dai, F., Smith, V., et al. (2007). Telomere uncapping by the
Gquadruplex ligand RHPS4 inhibits clonogenic tumour cell growth in vitro and in vivo
consistent with a cancer stem cell targeting mechanism. British Journal of Cancer, 96,
1223–1233.

106
Annual Review & Research in Biology, 1(4): 79-110, 2011

Piccini, P., Pavese, N., Hagell, P., Reimer, J., Björklund, A., et al. (2005). Factors affecting
the clinical outcome after neural transplantation in Parkinson’s disease. Brain, 128,
2977–2986.
Pierdomenico, L., Bonsi, .L, Calvitti, M., Rondelli, D., et al. (2005). Multipotent mesenchymal
stem cells with immunosuppressive activity can be easily isolated from dental pulp.
Transplantation, 80, 836–842.
Pittenger, M.F., Martin, B.J. (2004). Mesenchymal stem cells and their potential as cardiac
therapeutics. Circ, Res., 95, 9 –20.
Polak, J., Mantalaris, S., Harding, S.E., (2008). Advances in Tissue Engineering. London:
Imperial College Press, pp. 1–903.
Rajagopal, J., Anderson, W.J., Kume, S., Martinez, O.I., Melton, D.A. (2003). Insulin staining
of ES cell progeny from insulin uptake. Science 299: 363.
Rendl, M., Lewis, L., Fuchs, E. (2005). Molecular Dissection of Mesenchymal Epithelial
Interactions in the Hair Follicle. PLoS Biol., 3, 1910 – 1924.
Reubinoff, B.E., Itsykson, P., Turetsky, T., Pera, M.F., Reinhartz, E., Itzik, A., Ben-Hur, T.
(2001). Neural progenitors from human embryonic stem cells. Nat. Biotechnol., 19,
1134–1140.
Reubinoff, B.E., Pera, M.F., Fong, C.Y., Trounson, A., Bongso, A. (2000). Embryonic stem
cell lines from human blastocysts: Somatic differentiation in vitro. Nat. Biotechnol., 18,
399–404.
Reyes, M., Lund, T., Lenvik, T., Aguiar, D., Koodie, L., Verfaillie, C.M. (2001). Purification
and ex vivo expansion of postnatal human marrow mesodermal progenitor cells.
Blood, 98, 2615-2625.
Richards, M., Fong, C.Y., Chan, W.K., Wong, P.C., Bongso, A. (2002). Human feeders
support prolonged undifferentiated growth of human inner cell masses and embryonic
stem cells. Nat. Biotechnol., 20, 933–936.
Rowlands, A.S., Hudson, J.E., Cooper-White, J.J. (2007). From scrawny to brawny: the
quest for neomusculogenesis; smart surfaces and scaffolds for muscle tissue
engineering. Expert Rev. Med. Devices., 4 (5), 709–728.
Rufer, N., Brummendorf, T.H., Kolvraa, S., et al. (1999). Telomere fluorescence
measurements in granulocytes and T lymphocyte subsets point to a high turnover of
hematopoietic stem cells and memory T cells in early childhood. J. Exp. Med., 190,
157-167.
Sabatini, F., Petecchia, L., Tavian, M., et al. (2005). Human bronchial fibroblasts exhibit a
mesenchymal stem cell phenotype and multilineage differentiating potentialities. Lab.
Invest., 85, 962–971.
Sanberg, P.R. (2007). Neural stem cells for Parkinson’s disease: to protect and repair. Proc.
Natl. Acad. Sci. USA., 104(29), 11869–11870.
Sarin, K.Y., Cheung, P., Gilison, D., et al. (2005). Conditional telomerase induction causes
proliferation of hair follicle stem cells. Nature, 436, 1048 –1052.
Schuldiner, M., Eiges, R., Eden, A., Yanuka, O., Itskovitz-Eldor, J., et al. (2001). Induced
neuronal differentiation of human embryonic stem cells. Brain Res., 913: 201–205.
Schultz, E. (1996). Satellite cell proliferative compartments in growing skeletal muscles. Dev.
Biol., 175, 84–94.
Seaberg, R.M., Smukler, S.R., Kieffer, T.J. et al. (2004). Clonal identification of multipotent
precursors from adult mouse pancreas that generate neural and pancreatic lineages.
Nat. Biotechnol., 22, 1115–1124.
Segev, H., Fishman, B., Ziskind, A. et al. (2004). Differentiation of human embryonic stem
cells into insulin-producing clusters. Stem Cells, 22, 265– 274.

107
Annual Review & Research in Biology, 1(4): 79-110, 2011

Sengupta, A., Banerjee, D., Chandra, S., Banerji, S.K., Ghosh, R., Roy, R., et al. (2007).
Deregulation and cross talk among Sonic hedgehog, Wnt, Hox and Notch signaling in
chronic myeloid leukemia progression. Leukemia, 21, 949–955.
Seres, K.B., Hollands, P. (2010). Cord blood: the future of regenerative medicine?
Reproductive BioMedicine Online, 20, 98– 102
Serra, V., von Zglinicki, T. (2002). Human fibroblasts in vitro senesce with a donor-specific
telomere length. FEBS Lett., 516, 71-74.
Shamblott, M.J., Axelman, J., Wang, S., Bugg, E.M., et al. (1998). Derivation of pluripotent
stem cells from cultured human primordial germ cells. PNAS., 95, 13726-13731.
Sheyn, D., Mizrahi, O., Benjamin, S., Gazit, Z., Palled, G., Gazit, D. (2010). Genetically
modified cells in regenerative medicine and tissue engineering. Adv. Drug Deliv. Rev.,
doi:10.1016/j.addr.2010.01.002.
Shihuan, K., Kazuki, K., Fabien Le, G., Michael, A.R. (2007). Asymmetric self-renewal and
commitment of satellite stem cells in muscle. Cell, 129, 999–1010.
Shiroi, A., Yoshikawa, M., Yokota, H., Fukui, H., Ishizaka, S., et al. (2002). Identification of
insulin-producing cells derived from embryonic stem cells by zinc-chelating dithizone.
Stem Cells, 20, 284–292.
Sieber-Blum, M., Grim, M. (2004). The adult hair follicle: Cradle for pluripotent neural crest
stem cells. Birth Defects Res. C Embryo Today, 72, 162–172.
Singh, P., Williams, D.J. (2008). Cell therapies: realizing the potential of this new dimension
to medical therapeutics. J. Tissue Eng. Regen. Med., 2, 307–319.
Smith, R.R., Barile, L., Cho, H.C., Abraham, M.R, Messina, E., Giacomello, A., Marban, E.
(2005). Unique phenotype of cardiospheres derived from human endomyocardial
biopsies. Circulation, 112, 334-338.
Soria, B, Roche, E., Berna, G., Leon-Quinto, T., Reig, J.A., Martin, F. (2000). Insulin-
secreting cells derived from embryonic stem cells normalize glycemia in
streptozotocin-induced diabetic mice. Diabetes, 49, 157–162.
Steiner, A.F., Karl, N., Pilapil, C., Noth, U., Evans, C.H., Murray, M.M. (2006). Multilineage
mesenchymal differentiation potential of cells migrating out of the anterior cruciate
ligament. In: Transactions of the 52nd Annual Meeting of the Orthopaedic Research
Society; Chicago, IL. Paper #1133.
Stephane C. Boutet, Marie-He´ le` ne Disatnik, Lauren S. Chan, Kevin Iori, and Thomas A.
Rando (2007). Regulation of Pax3 by proteasomal degradation of monoubiquitinated
protein in skeletal muscle progenitors. Cell, 130, 349–362.
Stock, U.A., Vacanti, J.P. (2001). Tissue engineering: current state and prospects. Ann. Rev.
Med. 52, 443–451.
Suen, P.M., Leung, P.S. (2005). Pancreatic stem cells: A glimmer of hope for diabetes?
JOP., 6, 422– 424.
Takagi, Y., Takahashi, J., Saiki, H., et al. (2005). Dopaminergic neurons generated from
monkey embryonic stem cells function in a Parkinson primate model. J. Clin. Invest.,
115, 102–109.
Tamagawa, T., Ishiwata, I., Saito, S. (2004). Establishment and characterization of a
pluripotent stem cell line derived from human amniotic membranes and initiation of
germ layers in vitro. Hum. Cell, 17, 125–130.
Terada, N., Hamazaki, T., Oka, M., et al. (2002). Bone marrow cells adopt the phenotype of
other cells by spontaneous cell fusion. Nature, 416, 542–545.
Thomson, J.A., Eldor, J.I., Shapiro, S.S., Waknitz, M.A., et al. (1998). Embryonic stem cell
lines derived from human blastocysts. Science, 282, 1145–1147.
Tomar, G.B., Srivastava, R.K., Gupta, N., Barhanpurkar, A.P., et al. (2010). Human gingiva-
derived mesenchymal stem cells are superior to bonemarrow-derived mesenchymal

108
Annual Review & Research in Biology, 1(4): 79-110, 2011

stem cells for cell therapy in regenerative Medicine. Biochemical and Biophysical
Research Communications, 393, 377–383
Tomita, Y., Matsumura, K., Wakamatsu, Y., et al. (2005). Cardiac neural crest cells
contribute to the dormant multipotent stem cell in mammalian heart. J. Cell Biol., 170,
1135–46.
Towns, C.R., Jones, D.G. (2004). Stem cells, embryos, and the environment: a context for
both science and ethics. J. Med. Ethics, 30(4), 410–413.
Tumbar, T., Guasch, G., Greco, V., et al. (2004). Defining the epithelial stem cell niche in
skin. Science, 303, 359–363.
Takagi, Y., Takahashi, J., Saiki, H., et al. (2005) A phase 1 clinical trial of nerve growth
factor gene therapy for Alzheimer disease. Nature Med., 11, 551–555.
Tzukerman, M., Rosenberg, T., Ravel, Y., Reiter, I., Coleman, R., Skorecki, K. (2003). An
experimental platform for studying growth and invasiveness of tumor cells within
teratomas derived from human embryonic stem cells. Proc. Natl. Acad. Sci. USA.,
100, 13507–13512.
Urbanek, K., Torella, D., Sheikh, F., Angelis, A.D., Nurzynska, D., et al. (2005). Myocardial
regeneration by activation of multipotent cardiac stem cells in ischemic heart failure.
Proc. Natl. Acad. Sci. USA., 102, 8692– 8697.
Vacanti, J. (2010). Tissue engineering and regenerative medicine: from first principles to
state of the art. Journal of Pediatric Surgery, 45, 291–294.
Vawda, R., Woodburry, J., Covey, M., Levison, S.W., Mehmet, H. (2007). Stem cell
therapies for perinatal brain injuries. Seminars in Fetal & Neonatal Medicine, 12, 259-
272.
Voog, J., Jones, D.L. (2010). Stem Cells and the Niche: A Dynamic Duo. Stem Cell, 6, 103-
115.
Wagers, A.J., Sherwood, R.I., Christensen, J.L., Weissman, I.L. (2002). Little evidence for
developmental plasticity of adult hematopoietic stem cells. Science, 297, 2256–2259.
Wagner, J.J, Barker, J. (2007). Umbilical cord blood transplantation: Novel approaches
toward improving engraftment. Biol. Blood Marrow Transplant., 10, 733.
Wang, J.C., Dick, J.E. (2005). Cancer stem cells: Lessons from leukemia. Tr. Cell Biol., 15,
494 –501.
Watt, F.M. (2002). Role of integrins in regulating epidermal adhesion, growth and
differentiation. EMBO J., 21, 3919 –3926.
Weissman, I.L. (2002) Stem cells - scientific, medical, and political issues. N. Engl. J. Med.,
346, 1576–1579.
Weissman, I.L. (2000). Stem cells: Units of development, units of regeneration, and units in
evolution. Cell, 100, 157–168.
Woodbury, D., Schwarz, E.J., Prockop, D.J., Black, I.B. (2000). Adult rat and human bone
marrow stromal cells differentiate into neurons. J. Neurosci. Res., 61, 364–370.
Wu, Y., Chen, L., Scott, P.G., et al. (2007). Mesenchymal stem cells enhance wound healing
through differentiation and angiogenesis. Stem Cells, 25, 2648–59.
Yang, Y., Min, J-Y., Rana, J.S., Ke, Q., Cai, J., et al. (2002). VEGF enhances functional
improvement of postinfarcted hearts by transplantation of ESC-differentiated cells. J.
Appl. Physiol., 93, 1140–1151.
Young, H.E., Steele, T.A., Bray, R.A., et al. (2001). Human reserve pluripotent mesenchymal
stem cells are present in the connective tissues of skeletal muscle and dermis derived
from fetal, adult, and geriatric donors. Anat. Rec., 264, 51–62.
Zammit, P.S., Golding, J.P., Nagata, Y., Hudon, V., Partridge, T.A., Beauchamp, J.R. (2004).
Muscle satellite cells adopt divergent fates: A mechanism for self-renewal? J. Cell
Biol., 166, 347–357.

109
Annual Review & Research in Biology, 1(4): 79-110, 2011

Zhang, S.C., Wernig, M., Duncan, I.D., Brustle, O., Thomson, J.A. (2001). In vitro
differentiation of transplantable neural precursors from human embryonic stem cells.
Nat. Biotechnol., 19, 1129–1133.
Zuk, P.A., Zhu, M., Ashjian, P., De Ugarte, D. A., Huang, J.I., Mizuno, H., et al. (2001).
Multilineage cells from human adipose tissue: implications for cell-based therapies.
Tissue Eng., 7, 211–228.
Zvaifler, N.J., Marinova-Mutafchieva, L., Adam, G., Edwards, C.J., et al. (2000).
Mesenchymal precursor cells in the blood of normal individuals. Arthritis Res., 2, 477–
488.
________________________________________________________________________
© 2011 Ramakrishna et al.; This is an Open Access article distributed under the terms of the Creative Commons
Attribution License (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use, distribution, and
reproduction in any medium, provided the original work is properly cited.

110

You might also like