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

Annurev Pharmtox 010611 134540

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

PA53CH04-Nestler ARI 5 December 2012 12:48

ANNUAL
REVIEWS Further
Epigenetic Mechanisms
Click here for quick links to
Annual Reviews content online, of Depression and
including:
• Other articles in this volume
• Top cited articles
Antidepressant Action
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

• Top downloaded articles


by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

• Our comprehensive search


Vincent Vialou, Jian Feng, Alfred J. Robison,
and Eric J. Nestler
Fishberg Department of Neuroscience and Friedman Brain Institute, Mount Sinai
School of Medicine, New York, New York 10029; email: eric.nestler@mssm.edu

Annu. Rev. Pharmacol. Toxicol. 2013. 53:59–87 Keywords


First published online as a Review in Advance on DNA methylation, histone modification, noncoding RNA, limbic brain,
September 27, 2012
early-life stress, transgenerational
The Annual Review of Pharmacology and Toxicology
is online at pharmtox.annualreviews.org Abstract
This article’s doi: Epigenetic mechanisms, which control chromatin structure and function,
10.1146/annurev-pharmtox-010611-134540
mediate changes in gene expression that occur in response to diverse stim-
Copyright  c 2013 by Annual Reviews. uli. Recent research has established that environmental events and behavioral
All rights reserved
experience induce epigenetic changes at particular gene loci and that these
changes help shape neuronal plasticity and function and hence behavior.
Some of these changes can be stable and can even persist for a lifetime.
Increasing evidence supports the hypothesis that aberrations in chromatin
remodeling and subsequent effects on gene expression within limbic brain
regions contribute to the pathogenesis of depression and other stress-related
disorders such as post-traumatic stress disorder and other anxiety syndromes.
Likewise, the gradually developing but persistent therapeutic effects of an-
tidepressant medications may be achieved in part via epigenetic mechanisms.
This review discusses recent advances in our understanding of the epigenetic
regulation of stress-related disorders and focuses on three distinct aspects of
stress-induced epigenetic pathology: the effects of stress and antidepressant
treatment during adulthood, the lifelong effects of early-life stress on sub-
sequent stress vulnerability, and the possible transgenerational transmission
of stress-induced abnormalities.

59
PA53CH04-Nestler ARI 5 December 2012 12:48

INTRODUCTION
The importance of environmental influences in shaping brain structure and function has high-
lighted the profoundly plastic nature of the brain. In fact, all organisms continuously face stressful
events and changes in their environment that can affect their homeostasis. Stress responses are
reactions to these external challenges; they include changes in the central nervous system and
in various peripheral organs that aim to reinstate the initial homeostasis (1). The perception of a
stressful situation activates a large number of neuronal circuits, such as the hypothalamic-pituitary-
adrenal (HPA) axis, the locus coeruleus, and the autonomic noradrenergic centers in the brain
stem. These and many other initial stress responses target numerous limbic brain regions, such
as the prefrontal cortex, hippocampus, amygdala, and ventral striatum (also termed nucleus ac-
cumbens or NAc) (Figure 1) (2, 3). These changes in the central nervous system directly affect
learning and memory, alertness, arousal, and perhaps basal anxiety, and they promote adaptive
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

behavioral responses to subsequent stresses (4).


by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

The inability to regulate and terminate these stress responses can result in many forms of dys-
regulation, as illustrated by sustained elevations of glucocorticoid levels, which profoundly affect
neuroendocrine systems including immune responses, metabolism, and reproduction (5). Indeed,
stressful experiences increase the prevalence of a wide array of health impairments, including
coronary artery disease, chronic pulmonary disease, and certain cancers (6, 7). Stress can also have
negative consequences on behavioral adaptations and predispose some individuals to depression-
or anxiety-related disorders (8). However, susceptibility to stress varies strongly among individuals,
such that most are resilient: They can maintain normal physiological and psychological function
despite being subjected to horrendous stress (9, 10). Under the right conditions, prior exposure
to moderate levels of stress might promote future resilience, a phenomenon referred to as stress
inoculation (11). Although stress-related disorders such as depression, post-traumatic stress dis-
order (PTSD), and other anxiety syndromes are partly hereditary, a majority of the risk appears
to be nongenetic. The central hypothesis in the field, therefore, is that exposure to a range of
stressful stimuli in combination with an individual’s genetic constitution determines his or her
initial coping strategy, maladaptive versus resilient behavioral responses, and ultimate response to
treatment (12, 13).
Stress-induced changes in gene expression within the brain’s limbic circuitry have long been
posited to mediate this interplay between genes and environment (14). Altered expression of
several candidate genes has received a great deal of attention. Induction of brain-derived neu-
rotrophic factor (BDNF) in hippocampus is important in the efficacy of antidepressant treatment,
and its repression may contribute to the pathophysiology of depression in animal models (15).
In contrast, sustained induction of BDNF in the NAc, which persists for at least 1 month after
chronic social-defeat stress, mediates depression-like behavioral abnormalities, whereas reversal
of this effect induces an antidepressant-like response (9, 16). Other examples include suppres-
sion of the WNT/DVL/GSK3β/β-catenin signaling cascade in several limbic brain regions in
response to chronic stress, with opposite changes seen upon antidepressant treatment (17–20),
and the long-lasting downregulation of glucocorticoid receptors (GRs) in hippocampus resulting
from a deficiency in maternal care (21). More recently, genome-wide measures of gene expression
have been used to take a more open-ended view of alterations that occur in limbic regions after
chronic stress or antidepressant treatment (9, 16, 22–29). This work has also extended to the study
of resilience in chronic stress models to identify specific genes whose altered expression actively
opposes the development of stress-induced abnormalities (9, 30, 31).
In these various studies, the fact that chronic stress or antidepressant treatment can induce
altered expression of certain genes that persists for weeks after the last exposure suggests the

60 Vialou et al.
PA53CH04-Nestler ARI 5 December 2012 12:48

PFC Hipp

DR
NAc
VTA LC
PVN

AMY
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

Gene Region MeDNA Associated mechanisms Behavioral paradigm

BDNF Hipp Predator stress105

BDNF PFC Early-life stress146


CRF PVN Chronic mild stress (females)103
Social defeat
(reversed by imipramine)31

P11 PFC FSL depression model110


FSL with escitalopram110

GDNF NAc Social defeat – sus and res108


MeCP2/HDAC2 Social defeat – sus only108
MeCP2/CREB Social defeat – res only108
AVP PVN MeCP2 Maternal separation144

GR Hipp High LG (maternal attention)151

Figure 1
Effect of stress on DNA methylation and gene expression. Image of sagittal section of rodent brain depicts
several interconnected regions that are implicated in stress responses (top). Table (bottom) lists the small
number of specific genes whose methylation is altered by stress, the brain regions in which this regulation
occurs, and the types of stress involved. Transcriptional mechanisms associated with such altered DNA
methylation are listed in the “Associated mechanisms” column. Arrows indicate increase (red ) or decrease
( green) in gene expression (“Gene” column) or methylation (“MeDNA” column). References are in
superscript. Abbreviations: AMY, amygdala; AVP, arginine vasopressin; BDNF, brain-derived neurotrophic
factor; CRF, corticotropin-releasing factor; DR, dorsal raphe; FSL, Flinders Sensitive Line; GR,
glucocorticoid receptor; Hipp, hippocampus; LC, locus coeruleus; LG, licking and grooming; NAc, nucleus
accumbens; PFC, prefrontal cortex; PVN, paraventricular nucleus of hypothalamus; res, resilient; sus,
susceptible; VTA, ventral tegmental area.

involvement of epigenetic mechanisms, which are implicated in mediating highly stable changes
in gene expression during development and in adult tissues. Epigenetics, in its most general
sense, is the study of the regulation of a gene’s transcriptional potential in the absence of
changes to the DNA sequence. The burgeoning field of epigenetics is concerned with heritable
changes in transcriptional potential as well as the role of equivalent mechanisms in controlling

www.annualreviews.org • Epigenetic Mechanisms of Depression 61


PA53CH04-Nestler ARI 5 December 2012 12:48

transcriptional regulation during the lifetime of a single organism. Numerous mechanisms that
underlie epigenetic regulation have been described, including (a) direct modification of DNA’s
transcriptional potential by methylation of cytosine bases and (b) indirect regulation of DNA’s
accessibility through several types of chemical modification of the proteins involved in chromatin
structure, including histones and many nonhistone proteins (32). These mechanisms determine
the accessibility of a gene to the transcriptional machinery and ultimately the ability to produce
mRNA transcripts (Figure 2). An additional mode of epigenetic regulation is mediated by several
types of noncoding RNAs (ncRNAs), including small noncoding RNAs (sncRNAs) and long
noncoding RNAs (lncRNAs), which influence gene expression at multiple levels, for example,
by the modification of chromatin structure at specific genes, RNA splicing, and mRNA stability
(33).
Much of what we know about epigenetic mechanisms comes from developmental biology, in
which chromatin changes govern the acquisition and stability of cellular phenotypes throughout
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

differentiation (34, 35). Such cellular differentiation is accomplished by complex epigenetic reg-
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

ulation of individual genes via cues from surrounding cells and influences from the organism’s
environment. In contrast, many cell types in the adult organism no longer divide, but their pheno-
type, including their transcriptional potential, changes dramatically in response to environmental
challenges. In the brain, there are many thousands of cell types that have distinct phenotypes
that are subjected to experience-dependent phenotypic modification throughout the organism’s
lifetime. Growing evidence suggests that epigenetic mechanisms play a key role in this plasticity,
thereby mediating stable functional changes in the brain in response to environmental exposures
(36–41). Because they stably change diverse aspects of neuronal function, epigenetic modifica-
tions are implicated in learning and memory (42–44), social and maternal behaviors (45, 46), and
pathological behaviors, such as drug addiction (47, 48), depression (26), and PTSD (49). Such
epigenetic modifications could underlie the long-lasting behavioral abnormalities seen in these
disorders as well as the extraordinary interindividual variability in vulnerability to adversity. In
fact, epigenetics might partly explain the difficulty in identifying the specific genetic variations
that contribute to these syndromes, the significant discordance between monozygotic twins, and
the differential gender susceptibility to certain psychiatric diseases (50, 51). The study of epige-
netic regulation thus opens new doors to understanding normal behavioral traits as well as disease
etiology in humans. Although best demonstrated in cancer and stem cell biology, pathological
epigenetic marks have been observed in psychiatric disorders, for example, in postmortem tissue
of depressed patients (26, 52, 53) and of individuals who committed suicide (54). However, careful
interpretation is required because these epigenetic marks could represent the cause of the disability
or a mark of the disease state, or both.
In this review, we discuss recent progress toward understanding epigenetic regulation by stress
during early life and adulthood in the brain’s limbic circuitry, and how such mechanisms might
contribute to susceptibility versus resilience to stress-related disorders and to their reversal by
antidepressant treatments. We also address recent insight into possible epigenetic transmission of
behavioral adaptations to stress across generations.

OVERVIEW OF EPIGENETIC MECHANISMS

DNA Methylation
Cytosine bases can be covalently modified by methylation at the 5 position, resulting in a methyl
group projecting into the major groove of DNA (55). In mammals, such 5-methylcytosine (5mC)
predominantly occurs in the palindromic sequence 5 -CpG-3 (and occasionally 5 -CpNpG-3 )

62 Vialou et al.
PA53CH04-Nestler ARI 5 December 2012 12:48

Repressed
M Active A
A
M P

HDAC HAT

Transcription
factor

HMT
pol II
M
M M
A
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

M P
P
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

Heterochromatin Euchromatin

Residue Mark/effect Region Experimental manipulation


General Acet NAc Social stress in stress-sensitive mice, reversed by imipramine108
acetyl-H3 Hipp Social stress, reversed by imipramine116, 119, 120
M Electroconvulsive seizures124
M AMY Social stress116
M 27 A
mPFC Social stress117
H3 2Me NAc Broad changes with social stress, reversed by imipramine127
lysine 27 3Me Hipp Restraint stress126 and social stress120
NAc Social stress in stress-resistant mice108
H3 Acet NAc Temporally modulated by stress26
lysine 14 Hipp Regulation in LR/HR rats118
H3 1Me Hipp Restraint stress126
A 14 lysine 9 2Me NAc Social stress125
Broad changes with social stress, reversed by imipramine127
Hipp Restraint stress and fluoxetine126
M 3Me Hipp Restraint stress, reversed by fluoxetine126
9
M A
M H3 Acet Hipp Antidepressants and electroconvulsive seizures120,124
lysine 4 2Me Hipp Social stress and imipramine120
M
M M
4 A 3Me NAc, Hipp Restraint and social stress108,126
General Acet Hipp Regulation in LR/HR rats118
acetyl-
H2B
General Acet Hipp Regulation in LR/HR rats118
acetyl-
H4
H3 Acet Forebrain Maternal separation148
lysine 12

Figure 2
Chromatin modifications regulated by stress or antidepressant treatment. Illustration (top) indicates histone octamers ( pink) in
heterochromatin (left) and euchromatin (right), along with associated proteins and histone tail/DNA modifications. Table (bottom) lists
histone tail modifications of specific residues—depicted on the expanded histone tail illustration (left)—that are regulated by various
stress paradigms or antidepressant treatments within the indicated brain regions. Arrows indicate an increase (red ) or decrease ( green)
in specific modifications. References are in superscript. Abbreviations: A, acetylation; P, phosphorylation; M (in a square), histone
methylation; M (in a circle), DNA methylation; AMY, amygdala; HAT, histone acetyltransferase; HDAC, histone deacetylase; Hipp,
hippocampus; HMT, histone methyltransferase; HR and LR, high responding and low responding, respectively (with respect to
baseline locomotor activity); mPFC, medial prefrontal cortex; NAc, nucleus accumbens; pol II, RNA polymerase II.

www.annualreviews.org • Epigenetic Mechanisms of Depression 63


PA53CH04-Nestler ARI 5 December 2012 12:48

and does not interfere with normal hydrogen bonding with complementary guanine bases.
Approximately 3% of all cytosines in the human genome are methylated (56), and proper 5mC
is required for cell differentiation, genetic imprinting, suppression of repetitive elements, and
X-chromosomal inactivation (57). The distribution of CpG bases in mammalian DNA is uneven,
occurring at high concentrations in select regions termed CpG islands. CpG islands overlap with
the promoters of 50–60% of human genes and are typically methylated to a much lesser extent
than are CpG dinucleotides found outside islands (58).
Although methylated cytosines can both prevent and promote the binding of various pro-
teins to DNA (59), CpG methylation in promoter regions is generally considered to repress gene
transcription (see below). DNA methylation is catalyzed by DNA methyltransferases (DNMTs),
a family of enzymes that include DNMT1, DNMT2, DNMT3a, and DNMT3b (60). These
enzymes play distinct roles: DNMT1 maintains methylation patterns during DNA replication,
whereas DNMT3a and DNMT3b appear to catalyze de novo methylation of previously unmethy-
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

lated double-stranded DNA (55, 61). Several DNA demethylation pathways have been proposed,
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

such as those mediated by methyl CpG binding domain protein 2 (MBD2) (62), growth arrest and
DNA-damage-inducible protein 45 alpha (GADD45a) (63), and GADD45b (64). Recent studies
indicate DNA oxidation/repair pathways may drive active DNA demethylation in nondividing neu-
rons. Also, ten-eleven-translocation (TET) enzymes oxidize 5mC into 5-hydroxymethylcytosine
(5hmC), which can be further oxidized into 5-formylcytosine and 5-carboxylcytosine (65–68).
TET family hydroxylase-induced oxidation-deamination thus represents a new candidate for the
long-sought DNA demethylase in brain (69). Interestingly, all three forms of 5mC derivatives
seem to be enriched in brain, and 5hmC displays a developmentally programmed acquisition in
neuronal cells (70).
DNMTs can directly interact with transcription factors, presumably allowing the methylation
of promoter regions at specific locations. In a recent study, nearly 80 transcription factors were
found to interact with DNMTs (71). This directed methylation enables gene-specific binding
of MBDs. MBDs repress gene transcription through the recruitment of corepressor complexes
that can interfere with the transcriptional machinery or directly regulate chromatin structure (see
below). MBDs are essential for normal development and growth; loss-of-function mutations of
the MBD methyl CpG binding protein 2 (MeCP2) are associated with Rett syndrome (72, 73),
and the MBD Np95/ICBP90-like ring finger protein regulates the cell cycle and is implicated in
tumorigenesis (74). Although originally thought to be a stable modification, DNA methylation
appears to be subject to dynamic regulation in the adult brain (64, 75, 76), and regulation of DNA
methylation at specific genes has been demonstrated in multiple cases (77–79).

Histone Modifications
In eukaryotic cells, DNA is densely packed into chromatin through interactions with large pro-
tein complexes named nucleosomes. An individual nucleosome core is composed of an octamer
containing four histone dimers—one dimer each of histones H2A, H2B, H3, and H4—around
which are wrapped 147 bp of DNA in approximately two superhelical turns (36). Variant histone
proteins or posttranslational modifications of histones cause changes in chromatin compaction
that are correlated with more open (euchromatin: transcriptionally permissive) and closed (hete-
rochromatin: transcriptionally repressive) states (80). The N-terminal tails of histones are exposed
at the surface of the nucleosome and exhibit multiple, reversible covalent modifications that alter
the accessibility of DNA to the transcriptional machinery in a regulated fashion (Figure 2). In
addition, histones can be substituted with variants, such as the CENP-A variant of H3, which
is found specifically in centromeric DNA and whose tail contains a significantly different set of

64 Vialou et al.
PA53CH04-Nestler ARI 5 December 2012 12:48

modifiable residues. These traits suggest that substitution of this histone may be essential for
mitosis (81).
Histone acetylation, which negates the positive charge of lysine residues in the histone tail,
is associated with transcriptional activation. Gene activation is likely due to a weakening of the
interaction with the DNA strand that depends on the negative charge of lysines (36, 82). In contrast,
lack of histone acetylation correlates with gene repression. Histones are acetylated by histone
acetyltransferases (HATs), which use acetyl coenzyme A as a cosubstrate, and are deacetylated by
histone deacetylases (HDACs). HATs comprise a large family of proteins and acetylate multiple
lysine residues in the tails of both H3 and H4. HDACs are composed of multiple families of proteins
divided into three classes. In the brain, Classes I and II appear to regulate histone deacetylation
at most genes, whereas Class III enzymes (comprising the sirtuin proteins) deacetylate numerous
nuclear and cytoplasmic substrates in addition to histones and are implicated in numerous cellular
functions (83–86).
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

Unlike the clear correlation of histone acetylation with transcriptional activation, histone
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

methylation can be associated with both transcriptional activation and repression depending on
the particular residue modified and the extent of methylation. Both lysine and arginine residues can
be methylated by various histone methyltransferases (HMTs), which use S-adenosylmethionine
as a cosubstrate. Methylated arginine residues are converted to citrulline by deaminases, whereas
lysines are demethylated by lysine demethylases (87). Unlike acetylation, methylation does not
substantially alter the charge of the target residues; however, it can dramatically change the
steric profile and potential molecular interactions through multivalent addition of mono-, di-, or
trimethyl groups. Examples of the bidirectional effects of histone methylation include trimethy-
lation of lysine 4 of H3 (H3K4me3), which is associated with transcriptional initiation, and di-
and trimethylation of lysines 9 and 27 of H3 (H3K9me2/3 and H3K27me2/3), which are associ-
ated with transcriptional repression (47). In contrast, H3K36me3 is associated with transcription
elongation.
Multiple additional modifications of histone tails are known, including phosphorylation, ubiq-
uitination, sumoylation, and ADP ribosylation (32, 36). For instance, phosphorylation of serine-10
of H3 is associated with increased gene transcription through a complex mechanism that involves
HAT recruitment and prevention of HMT activity at lysine 9 (47). Modification of one histone
can also affect subsequent modification of other histones within the nucleosome, as ubiquitina-
tion of H2B appears to be a prerequisite for H3K4 methylation and the resulting initiation of
transcription (88). The level of complexity in the nearly infinite number of possible combina-
tions of various modifications at multiple residues of histone tails, as well as the introduction of
histone variant subunits, has led to the proposal of a histone “code” whose “reading” by various
histone-interacting proteins is essential for the proper regulation of gene expression (81, 89).

Noncoding RNAs
Many types of ncRNAs are well known, such as ribosomal RNA (rRNA) and transfer RNA (tRNA).
More recently, many additional ncRNAs have been described, divided between the lncRNAs
and various short varieties, including microRNAs (miRNAs), small nucleolar RNAs (snoRNAs),
promoter-associated short RNAs (PASRs), Piwi-interacting RNAs (piRNAs), and transcription
initiation RNAs (tiRNAs) (33). lncRNAs are generally longer than 200 nucleotides each; they can
be spliced like mRNAs to form functional secondary structures and can act as precursors for various
sncRNAs (90). Although only a small number of functional lncRNAs have been characterized,
they have been shown to control gene expression programs at every level (91). lncRNAs appear to
function via multiple epigenetic mechanisms and can modulate the status of protein-coding genes

www.annualreviews.org • Epigenetic Mechanisms of Depression 65


PA53CH04-Nestler ARI 5 December 2012 12:48

through the recruitment of chromatin remodeling complexes to specific regions of the genome,
thereby regulating the chromatin structure of a single promoter region, a cluster of genes, or an
entire chromosome (90). lncRNAs can also serve as enhancers to regulate gene transcription (92).
miRNAs are the best studied sncRNAs in brain. They are posttranscriptional regulators that can
bind to complementary sequences on target mRNAs and lead to translational repression or mRNA
degradation (93). miRNAs appear to have the ability to bridge with other epigenetic mechanisms
to regulate neural plasticity (94). In addition, sncRNAs can influence epigenetic regulation of
gene expression. For instance, sncRNAs bound to Argonaute proteins (and other factors required
for inhibitory RNAs or RNAi, such as Dicer) generate H3K9 methylation and are implicated in
the process of heterochromatin formation through the recruitment of lysine methyltransferases
(95). In Caenorhabditis elegans, short interfering RNAs (siRNAs) and RNAi trigger long-term gene
silencing in a dominantly heritable fashion, with multiple chromatin remodeling factors required
for maintenance of the silent state (96).
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

REGULATION OF THE EPIGENOME BY STRESS AND


ANTIDEPRESSANT TREATMENT IN ADULTHOOD
Considerable evidence suggests that chromatin remodeling is a dynamic process that occurs
throughout life in many organs including the brain (34, 97, 98). Patterns of DNA methylation
and histone acetylation gradually change over time in monozygotic twins (99). Such drift might
not occur randomly and could represent the effect of the environment and the accumulation of
the individual’s experience. Chromatin remodeling processes are necessary for learning and mem-
ory, and aberrant epigenetic modification can lead to cognitive deficits (100, 101). Within this
context, an increasing number of studies have shown that exposure to stress promotes alterations
in various epigenetic marks—in particular, histone acetylation and methylation as well as DNA
methylation—in various limbic brain regions (Figures 1 and 2). Additionally, experimental ma-
nipulation of the epigenetic machinery within these regions potently controls mood states and
responses to stress and antidepressant treatments.

Regulation of DNA Methylation in Adult Responses to Stress


Given that DNA methylation is a relatively stable epigenetic mark in postmitotic cells, it likely
participates in the long-lasting change in gene expression that underlies the development of stress-
induced abnormalities. Recent reports show changes in CpG methylation levels at gene promoters
that are implicated in HPA axis reactivity and antidepressant treatment. Depression-like symp-
toms induced by chronic social-defeat stress are accompanied by the sustained upregulation of
corticotropin-releasing factor (CRF), which is not observed in mice that are resilient to the stress
(31). CRF, which is expressed by neurons of the paraventricular nucleus of hypothalamus (PVN),
controls HPA axis activity as well as several other stress responses in the brain. Stress induc-
tion of the Crf gene is accompanied by a decrease in DNA methylation at its promoter. Chronic
imipramine treatment was sufficient to reduce Crf mRNA levels and increase DNA methylation at
the Crf promoter only in socially defeated mice. A related observation is that conditional knock-
out of MeCP2 in the PVN, which reduces the repression of certain methylated genes, induces an
abnormal physiological stress response (102). This observation shows that epigenetic regulation
of genes is critical for appropriate regulation of the HPA axis. Interestingly, female rats exposed to
chronic mild stress show increased DNA methylation at the Crf promoter in the PVN, suggesting
sex-specific alterations in HPA axis activity (103).

66 Vialou et al.
PA53CH04-Nestler ARI 5 December 2012 12:48

Activation of the HPA axis and the subsequent release of glucocorticoids have a profound im-
pact on hippocampal function because hippocampal neurons express high levels of GR. Therefore,
the hippocampus has received a lot of attention for its role in stress-induced behavioral and cog-
nitive deficits and antidepressant responses. One mechanism by which stress affects hippocampal
function is through repression of BDNF, as noted above (104). Chronic exposure to predator
stress increases methylation of the Bdnf promoter in hippocampus (105), possibly contributing
to the morphological changes and neuronal impairments reported after chronic stress in animal
models and in depressed humans (106, 107).
Genetic background influences the outcome of stress exposure in animals and humans, and
such background differences could be mediated in part by DNA sequence–directed differences in
epigenetic modifications in response to stress, a possibility that is just beginning to be explored.
Uchida et al. (108) report that chronic mild stress increases anxiety- and depression-like behaviors
in BALB/C mice, effects not seen under the same conditions in C57BL/6 mice. Exposure to stress
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

differentially regulates glial cell–derived neurotrophic factor (GDNF) expression in the NAc in the
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

two strains of mice (108); BALB/C mice display decreased GDNF expression, whereas C57BL/6
mice show increased expression. The authors report that DNA methylation and MeCP2 binding
were increased at the Gdnf promoter in the NAc of both strains after chronic stress and that these
changes were associated with increased expression of DNMT1 and DNMT3a. However, the two
strains are distinguished by the protein complexes that associate with MeCP2 at the Gdnf pro-
moter. In BALB/C mice, MeCP2 reportedly interacts with HDAC2 to decrease H3 acetylation
and concomitantly represses Gdnf transcription in the NAc, whereas the association of MeCP2
with transcriptional activator CREB (cAMP response element–binding protein) is purported to
enhance GDNF expression in the NAc of C57BL/6 mice. The authors propose that these differ-
ences are driven by different methylation patterns at the Gdnf promoter, although much further
work is needed to validate these findings as well as understand their molecular basis. Neverthe-
less, these observations are consistent with reports that MeCP2 can act as a repressor or activator
of gene transcription (109). In the Flinders Sensitive Line genetic rodent model of depression,
elevations in DNA methylation at the P11 promoter were reduced by chronic administration of
escitalopram, leading to an increase in P11 and a decrease in DNMTs in the prefrontal cortex
(110). P11 interacts with serotonin and perhaps other receptors and regulates their function, and
decreased levels of P11 have been observed in postmortem depressed patients and associated
with depression-like behaviors in mice (111, 112). Finally, decreased DNA methylation at the
promoter of the serum interleukin-6 (IL-6) gene in peripheral blood mononuclear cells was asso-
ciated with higher levels of IL-6 in adults who had a history of depression (113). IL-6 is part of
the inflammatory response system that is dysregulated in some depressed individuals (114).
As noted, induction of DNMTs occurs in the NAc after chronic social-defeat stress (115),
and recent evidence demonstrates directly that this adaptation contributes to the development
of depression-like behaviors. Overexpression of DNMT3a in the NAc facilitates vulnerability to
the deleterious effects of the stress. This mechanism appears critical for the establishment of mal-
adaptive behaviors because intra-NAc infusion of two DNMT inhibitors, RG108 and zebularine,
reversed the behavioral deficits induced by chronic stress (108, 115). Because DNMTs generally
function to repress gene transcription, these data suggest that the development of stress-induced
behavioral abnormalities is accompanied in the NAc by the downregulation of genes implicated
in reward and motivation. In contrast, DNA methylation in other brain regions, such as those
important for the HPA axis (discussed above), constitutively represses genes that participate in
stress responses. Therefore, drugs that globally affect DNA methylation are unlikely to be vi-
able therapeutic approaches, although targeting site-specific DNA methylation may be a valuable
pathway for the development of future therapies.

www.annualreviews.org • Epigenetic Mechanisms of Depression 67


PA53CH04-Nestler ARI 5 December 2012 12:48

Regulation of Histone Acetylation in Adult Responses to Stress


Although considered more labile, some histone posttranslational modifications can contribute to
the persisting abnormalities of stress-related psychopathology. After chronic social defeat in mice,
the NAc displays a transient decrease in total cellular levels of H3 acetylation followed by a more
persistent increase that lasts for at least 10 days after the last stress (26). This lasting increase may
be mediated via the sustained repression of HDAC2 expression in this brain region. Interestingly,
increased H3 acetylation and decreased HDAC2 levels are seen in the NAc of depressed humans.
Nevertheless, these changes seem to be adaptive because local infusion of HDAC inhibitors into
the NAc, or local inhibition of HDAC2 via overexpression of a dominant negative mutant of the
enzyme, exerts potent antidepressant-like actions in several behavioral assays (26, 108). Moreover,
local administration of an HDAC inhibitor produced a pattern of gene expression changes in the
NAc that was somewhat similar to that seen with systemic fluoxetine treatment (26). Chronic
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

social-defeat stress induces more transient increases in global H3 acetylation levels in other brain
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

regions, such as hippocampus, amygdala, and medial prefrontal cortex, and these effects may also
be adaptive because HDAC inhibitors behave like antidepressants when administered into these
regions as well (116, 117). However, other groups have observed decreased H3 acetylation levels
in hippocampus after chronic stress and the reversal of these effects by imipramine treatment (116,
118, 119).
Nevertheless, several groups have demonstrated antidepressant-like effects of HDAC inhibitors
upon systemic administration (120–122); such inhibitors have no effect on depression-like behav-
iors in nonstressed mice (123). These studies thus suggest that HDAC inhibitors show some
potential as novel antidepressant agents. However, the lack of specificity of the inhibitors used,
and their limited ability to penetrate the brain, are major caveats that must be considered. Given
the lack of brain-specific HDAC isoforms, it seems unlikely that HDAC inhibitors of acceptable
safety and specificity can be developed for treatment of depression. Still, these data can buttress
drug discovery efforts by helping identify the genes important in depression models, after which
these genes can be targeted for drug development. For example, comparison of genes within a
given limbic region regulated by fluoxetine versus HDAC inhibitors might allow identification
of novel mechanisms that are unique to HDAC inhibition and perhaps capable of better, faster
antidepressant responses (26).
Currently, little is known about the specific genes at which stress-induced changes in his-
tone acetylation mediate such regulation of depression-like behavior. Chronic social-defeat stress
downregulates Bdnf transcription in hippocampus, and this downregulation is reversed by an-
tidepressant treatment (120). Although the repression may be mediated by enhanced DNA and
histone methylation (see below), antidepressant reversal of this effect is associated with increased
H3 acetylation at the Bdnf promoter (120, 124). This hyperacetylation resulting from antide-
pressant treatment is mediated by the downregulation of HDAC5 in hippocampus, which is
necessary for the behavioral effects of chronic antidepressant administration (120). Further work
is needed to identify, in a genome-wide manner, the many other genes involved in mediating
the antidepressant-like effects of enhanced histone acetylation at the levels of several limbic brain
regions.

Regulation of Histone Methylation in Adult Responses to Stress


Chronic social defeat, coincident with the sustained increase in H3 acetylation in the NAc noted
above, causes a global decrease in levels of H3K9me2, which is a repressive mark mediated
via downregulation of the associated HMTs G9a and G9a-like protein (125). These latter

68 Vialou et al.
PA53CH04-Nestler ARI 5 December 2012 12:48

adaptations represent stress-induced pathological changes because conditional knockout of


G9a from the NAc, or its pharmacological inhibition, promoted susceptibility to social stress
(125). In contrast, G9a overexpression in the NAc, which increases H3K9me2 levels, promotes
stress resilience and induces antidepressant-like responses (26, 125). One gene product that
contributes to these effects is Ras, a small GTPase upstream of the ERK signaling cascade.
Stress-induced decreases in H3K9me2 at the Ras gene in the NAc lead to an increase in Ras
expression and to subsequent activation of the ERK signaling cascade, including activation of
CREB, which then induces depression-like behavioral abnormalities (Figure 3). Interestingly,
two adaptations associated with gene activation (reduced H3K9me2 and increased H3 acety-
lation) exert opposite behavioral effects. One explanation is that different genes are affected
by these modifications, a possibility that now requires direct investigation with genome-wide
methods.
Chronic stress also alters levels of histone methylation in hippocampus. Differential effects
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

on global levels of H3K4me3, H3K9me3, and H3K27me3 within distinct hippocampal subfields
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

occur in response to acute versus chronic restraint stress (126). Interestingly, our group found that
chronic social-defeat stress increases H3K27me3 levels at the Bdnf gene promoter in hippocampus
in concert with its repression (120). Histone repressive marks are often associated with increased
DNA methylation, and this finding is consistent with increased methylation observed at this gene
in response to other forms of stress (105).
The discussion above highlights the need for genome-wide assessments of chromatin changes,
which will allow identification of previously unknown mechanisms involved in stress-related
disorders and antidepressant action. Such studies are just now getting under way. ChIP-chip
analysis (chromatin immunoprecipitation followed by genome-wide promoter arrays) for
H3K9me2 identified numerous genes that display increases or decreases in this chromatin
mark 1 month after chronic social-defeat stress (127) (Figure 4). Interestingly, a similar global
pattern of H3K9me2 binding was observed after prolonged adult social isolation. Although
these two forms of chronic stress are ethologically distinct, they induce some similar behavioral
abnormalities (9, 128). Furthermore, imipramine treatment reverses the large majority of
these histone methylation changes in the NAc induced by chronic social-defeat stress, and
the pattern of chromatin changes induced by imipramine resembles that seen in resilient
mice (127). These novel findings suggest that one mechanism by which antidepressants work
is to induce some of the same molecular adaptations that occur naturally in more resilient
individuals, and that a new path forward in antidepressant drug discovery is to mimic such natural
resilience mechanisms. For example, these data implicated the WNT/DVL/GSK3β/β-catenin
pathway in mediating susceptibility versus resilience in the NAc, as well as in mediating
antidepressant action; these findings were later validated by direct manipulation of this cascade
(20).
Another ChIP-chip study mapped the genome-wide binding of SETDB1, an H3K9 HMT,
in mouse prefrontal cortex and identified the NR2B NMDA glutamate receptor subunit as one
key target (129). Interestingly, the authors of this study also demonstrated that overexpression
of SETDB1 in forebrain exerts antidepressant-like actions. This observation is consistent with
those noted above: that G9a, another H3K9 HMT, also produces antidepressant-like effects
when overexpressed in the NAc (125). The identification of NR2B as one relevant substrate
emphasizes the need in the field to further characterize the influence of chronic stress and an-
tidepressant treatments on synaptic plasticity in several key limbic regions (130). Loss of KAP1,
a protein implicated in transcriptional repression, also increases stress vulnerability (131), high-
lighting the large number of chromatin mechanisms that likely control adaptations to chronic
stress.

www.annualreviews.org • Epigenetic Mechanisms of Depression 69


PA53CH04-Nestler ARI 5 December 2012 12:48

Repeated cocaine
Normal State of enhanced stress vulnerability

Cortical or VTA projection Cortical or VTA projection

DAR, GluR, DAR, GluR,


BDNF BDNF
nAChR, etc. nAChR, etc.
TrkB TrkB

Ras-GRF1 Ras-GRF1 Ras GDP


GDP
Ras
Raf P
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

CYTOPLASM Raf
MEK1/2 P
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

MEK1/2

ERK1/2 ERK1/2 P

CREB G9a GLP


NUCLEUS
CREB P
GLP me2
G9a
AC
DNA
Ras Ras pCREB

AC

H-Ras1 H-Ras1
pCREB pCREB pCREB

Figure 3
Enhanced vulnerability to stress via cocaine-induced priming of BDNF signaling in the NAc. Repeated cocaine administration
increases vulnerability to the depression-like effects of social-defeat stress via priming of BDNF signaling through Ras induction in the
NAc. Under control conditions (left), BDNF activation of TrkB signaling is limited. However, after repeated cocaine administration
(right), BDNF-TrkB signaling is elevated in the NAc, causing enhanced phosphorylation and activity of downstream-signaling
mediators such as CREB. This cocaine-initiated maladaptive response occurs not only through increased BDNF signaling in the
NAc but also through increased Ras expression as a result of decreased G9a binding, and decreased H3K9me2, at the H-Ras1 gene
promoter. Chronic stress is associated with similar adaptations in this brain region. Ras also appears to be a target for CREB, creating a
positive feed-forward loop, promoting CREB activation and Ras expression as well as depression-like behavior. Abbreviations: BDNF,
brain-derived neurotrophic factor; CREB, cAMP response element–binding protein; DAR, dopamine receptor; GLP, G9a-like protein;
GluR, glutamate receptor; NAc, nucleus accumbens; nAChR, nicotinic acetylcholine receptor; pCREB, phospho-CREB; VTA, ventral
tegmental area. Modified from Reference 125.

Regulation of miRNAs in Adult Responses to Stress


Additional forms of epigenetic regulation are likely involved in stress and antidepressant action.
Chronic administration of fluoxetine reportedly increases levels of miRNA-16 (miR-16), which
targets the serotonin transporter (SERT) transcript in serotonergic neurons and decreases SERT
expression (132). More surprisingly, fluoxetine downregulates miR-16 levels in noradrenergic

70 Vialou et al.
PA53CH04-Nestler ARI 5 December 2012 12:48

a
b H3K9me2
Susceptible
Susceptible
+ vehicle
H3K9me2 Altered in Altered in
Susceptible Resilient susceptibility 422 275 604 susceptibility
+ imipramine

Susceptible pCREB
+ vehicle Susceptible
pCREB
Susceptible Resilient Normal in 350 211 267
+ imipramine resilience imipramine

Figure 4
Regulation of H3K9me2 and phospho-CREB (pCREB) binding in the nucleus accumbens after chronic social-defeat stress. (a) The
heat maps show that, for both marks, the large majority of changes (red, up; green, down) seen in susceptible mice are not seen after
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

chronic imipramine; furthermore, most changes observed in susceptible mice are not seen in resilient mice, and vice versa. The lower
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

bars in each heat map are normalized to the upper bars, meaning that green would depict reversal of a red change, not a change in the
opposite direction. (b) The Venn diagrams show the appreciable overlap between apparent mechanisms of imipramine and of resilience.
Modified from Reference 127.

neurons of the locus coeruleus and thereby enables certain serotonergic phenotypes in those cells.
Further work is needed to validate this adaptation and explore its contribution to the antidepressant
effects of fluoxetine. In addition, constitutive and ubiquitous manipulations of SIRT1, a Class III
HDAC discussed above, affect basal levels of anxiety, and increased SIRT1 activity is associated
with increased anxiety-like behaviors (133). SIRT1 also decreases the expression of miR-134 in
hippocampus, thereby enhancing CREB and BDNF levels (134); these increased levels could
contribute to the effects of SIRT1 on anxiety and stress responses. These early reports highlight
the need for additional studies of miRNAs in stress and antidepressant action.

EPIGENETIC REGULATION BY EARLY-LIFE STRESS


During development, the brain is sensitive to environmental changes, such as parental care and
stressful events. Cognitive stimulation can lead to broad effects on brain structure and function,
hence influencing cognitive and affective function in children, adolescents, and young adults (6,
7). Maltreatment, neglect, and trauma during childhood increase the risk of depressive, anxiety,
and substance abuse disorders as well as the risk of becoming an abusive parent (135–139). In
contrast, more mild forms of stress may promote resilience (10, 11). Such bidirectional regulation
during early life of susceptibility to stress-related illness in adulthood is likely mediated by stable
alterations in chromatin structure of specific genes in brain. Longitudinal studies revealed changes
in DNA methylation in buccal epithelial cells that were associated with early-life adversity (140),
although how such changes relate to brain function remains unknown. Nevertheless, animal mod-
els have clearly identified epigenetic reprogramming of gene expression as a stable mechanism by
which early-life experience, in particular stress and maternal care, affects the individual’s responses
to stress later in life (141).
One of the most commonly used procedures for inducing early-life stress in rodents is periodic
maternal separation during early postnatal life. It is characterized by long-term physiological
and behavioral alterations, including elevated glucocorticoid levels and hyperactivity of the
HPA axis in response to subsequent stresses (1, 142, 143). Murgatroyd et al. (144) reported
that arginine vasopressin (AVP) undergoes increased expression in the PVN with maternal
separation. In addition to playing a role in water homeostasis, AVP is a key regulator of the HPA
axis. AVP is secreted by PVN neurons and, in conjunction with CRF, regulates the release of

www.annualreviews.org • Epigenetic Mechanisms of Depression 71


PA53CH04-Nestler ARI 5 December 2012 12:48

adrenocorticotropin hormone (145). Early-life stress activates calcium/calmodulin-dependent


protein kinase II (CaMKII) and downstream phosphorylation of MeCP2 at serine-438 (S438) in
the PVN, which leads to the release of MeCP2 from the Avp gene enhancer (144). This process
is associated with activation of the Avp gene and increased HPA axis stress reactivity, both of
which are sustained into adulthood. However, lifelong Avp gene induction is not accompanied by
persistent CaMKII activation and MeCP2-S438 phosphorylation. Rather, kinase activation and
MeCP2 phosphorylation are followed by Avp gene enhancer demethylation. Presumably, both
CaMKII activation and MeCP2 phosphorylation exert broad transcriptional effects beyond the
Avp gene, which remain a focus for future investigations. Similarly, newborn rats that are exposed
to stressed caretakers for the first postnatal week display significant increases in DNA methylation
at regulatory regions (exons IV and IX) of the Bdnf gene in prefrontal cortex and hippocampus,
along with persistent BDNF downregulation in prefrontal cortex but not hippocampus (146). Such
early-life regulation of BDNF expression might influence stress vulnerability later in life (147).
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

Although the causal role of DNA methylation in transcriptional regulation during postnatal
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

brain development has been challenged (148), these studies of early-life stress demonstrate that
DNA methylation is an ideal mechanism, at least from a heuristic point of view, for lifelong
changes in transcriptional potential. Early-life stress can also modify histone acetylation levels
through changes in HDACs. Maternal separation in BALB/C mice decreases HDACs levels in
the adult forebrain, leading to a concomitant increase in acetylated H4 lysine 12 (149). Activation
of HDACs and the resulting decrease in acetylated H4 worsen the effect of stress, suggesting that
the observed reduction in HDAC expression is a positive adaptive process.
In rodents, the style of maternal care has long-term effects on the behavioral and endocrine re-
sponses to stress in offspring (142). Maternal care for the neonatal rat is measured by the frequency
of pup licking and grooming (LG) behavior, which is the major source of tactile stimulation that
regulates endocrine, cardiovascular, and behavioral responses (150). Variation in maternal behav-
ior is a naturally occurring phenomenon, which can have long-term effects on neural systems that
regulate learning and memory, neuroplasticity, and emotional and stress responses (150). In fact,
differential levels of maternal care profoundly influence hippocampal GR levels, hence affecting the
regulation of the HPA axis by stress. As adults, the offspring of high-LG mothers show increased
hippocampal GR expression and therefore enhanced negative feedback regulation by glucocorti-
coids in comparison with adult animals reared by low-LG mothers (21). This range of GR expres-
sion levels has been traced inversely to levels of methylation at the GR gene promoter (Figure 5).
Lower levels of methylation enhance the accessibility of the promoter to transcription factors and
chromatin regulatory proteins such as CREB-binding protein (CBP) (151, 152). Increased binding
of CBP, a HAT that acetylates specific sites of H3 and H4, leads to increased histone acetylation at
the GR promoter in high-LG offspring (153, 154). All effects of high-LG rearing can be reversed
in the adult offspring by intracerebroventricular infusion of L-methionine, which acts as a methyl
group donor (155). Similarly, infusion of the HDAC inhibitor trichostatin A reverses the changes
induced by low-LG rearing (151). These findings once again demonstrate the interplay between
DNA methylation and histone modifications and show that changes in the epigenome established
by the environment during early development can be reversed by pharmacological approaches in
adults—emphasizing the plasticity of chromatin regulation in the adult brain. Although low-LG
rats show a heightened response to stress, they show greater contextual learning under stressful
conditions, suggesting that the long-term behavioral adaptations induced by maternal care have
some adaptive value (156, 157), i.e., that increased stress responsiveness might prepare the indi-
vidual to survive in more aversive environments. McGowan et al. (158) demonstrated the clinical
relevance of these observations in rats: They found increased methylation of the GR promoter and
reduced GR expression in the hippocampus of suicide victims with a life history of childhood abuse.

72 Vialou et al.
PA53CH04-Nestler ARI 5 December 2012 12:48

Inattentive mothering causes


methyl marks to be added

Attentive mothering causes


methyl marks to be removed
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

Figure 5
Long-term epigenetic effect of maternal care. Studies in rats have shown that epigenetics can influence maternal behavior and that this
effect can be passed from one generation to the next by acting on the pup’s brain alone, without altering germ cells. When pups are
born, genes involved in regulating the animals’ responses to stress are differentially methylated, enhancing sensitivity to stress. If the
pups are raised by a mother that displays high levels of grooming behavior, many of these methyl marks are removed, leaving the
animals less responsive to stress. When these pups mature, they, too, will be more attentive mothers. If the pups, however, are raised by
a mother that displays low levels of grooming, their genes will gain methyl marks. They grow up to be more stress responsive and
display lower grooming levels toward their own pups.

Other behaviors also depend on mother-infant interactions. Maternal LG behavior affects


female offspring in their own expression of maternal care. Indeed, maternal care is a matrilineally
transmitted behavior that is mediated in part by long-term alteration of oxytocin expression in the
hypothalamic medial preoptic area (MPOA) and its regulation by estrogen (157). Oxytocin plays
a central role in social bonding and maternal care. Female offspring of low-LG mothers exhibit
decreased expression of estrogen receptor-alpha (ERα) through increased methylation at the ERα
gene promoter. Such downregulation of ERα reduces the ability of estrogen to activate oxytocin
receptor expression in the MPOA (45), hence reducing the ability of oxytocin itself to promote
maternal care. This epigenetic regulation of ERα expression within the MPOA emerges during
the first postnatal week and is maintained into adulthood. Cross-fostering studies reveal a direct
relationship between the maternal behavior and the behavioral and physiological development of
the pup. In contrast, female offspring of high-LG animals exhibit reduced methylation at the ERα
promoter and increased ERα expression. Consequently, these females have increased estrogen
sensitivity, along with greater oxytocin function, which drives LG behavior. Later changes in the
social environment can reverse these behavioral adaptations (159), suggesting that these epigenetic
marks are subjected to modification throughout life.
A major need in the field is to extend these studies of AVP, GR, and ERα to identify the pre-
sumably many additional genes whose methylation might further contribute to these behavioral
phenotypes. Through the use of a high-density tiling array, the adult rat chromosome 18, which
contains the GR gene, was examined for DNA methylation, H3K9 acetylation, and transcriptional
changes under differential LG mothering (160). The study demonstrated global epigenomic dif-
ferences in adult brain, which span gene promoters, coding regions, and gene ends as well as
intergenic regions. The data suggest that early-life maternal care has a broad effect on transcrip-
tion, with hundreds of genes implicated (45, 160, 161). Among these genes are those encoding
several protocadherin isoforms as well as rRNA, and hypermethylation of the rRNA promoter
was observed in the hippocampus of humans with a history of early childhood neglect/abuse who

www.annualreviews.org • Epigenetic Mechanisms of Depression 73


PA53CH04-Nestler ARI 5 December 2012 12:48

died by suicide (162). Further work is needed to characterize the functional effects of these and
the many other observed promoter modifications on lifelong stress vulnerability.
Early-life stress also encompasses environmental exposures during pregnancy. An extensive
literature explores the potential effects of prenatal stress on the development of depression and
other psychiatric syndromes (163–165), which would be expected to have an epigenetic basis. In
humans, depressed mood during the third trimester of pregnancy induced an increased DNA
methylation at the GR promoter in blood leukocytes of the offspring (166). This effect might
relate to the finding that offspring of Holocaust survivors with PTSD showed lower 24-h urinary
cortisol excretion than did offspring of survivors without PTSD (167, 168). These early, intriguing
observations define more mechanistic experiments that are now being carried out in rodent models,
as discussed further below (see Possible Heritability of Behavioral Adaptations to Stress).
This discussion of epigenetic consequences of early-life stress focuses on DNA methylation,
which, because of its stability, is an attractive mechanism for mediating lifelong behavioral adap-
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

tation. However, the notion that changes in DNA methylation are subject to dynamic and bidi-
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

rectional regulation in fully differentiated neurons goes against a prevailing view in the field that
it represents a permanent modification during cell differentiation (169). Several studies noted
above report that de novo DNA methylation occurs in postnatal neurons (151) and that DNA
demethylation occurs throughout life (75, 76, 151). The idea that a wave of de novo DNA methy-
lation occurs in neurons early in life is supported by the finding that DNMT expression peaks in
neurons during early postnatal life (170). These observations raise the interesting possibility that
DNA methylation serves a crucial function in mediating the critical time windows for activity-
dependent synaptic formation and refinement, along with associated behavioral adaptations, long
known to exist in early life (171).
Usually, tens to hundreds of CpG dinucleotides exist in a given gene’s regulatory and coding
regions, and the combination of “on” (CpG demethylation) and “off” (CpG methylation) signals
at each CpG site produces a complex methylome for each gene. Because methylation changes
at distinct regions of a gene might exert different functional effects, measures of total DNA
methylation do not provide an accurate assessment of that gene’s transcriptional potential. We
are still learning about the functional consequences of different patterns of DNA methylation
at neural genes, and much further work is needed to translate the methylome—which acts in
concert with the newly defined 5hmC, as well as large numbers of other forms of chromatin
modifications—into an understanding of the gene-specific epigenetic effects of early-life stress.

POSSIBLE HERITABILITY OF BEHAVIORAL


ADAPTATIONS TO STRESS
Our discussion up to this point has focused on a role for epigenetic mechanisms in mediating
stable, in some cases lifelong, neural and behavioral adaptations in response to environmental
challenges. Far more provocative is the idea of the transmission of parental adaptations to envi-
ronmental challenges to offspring when the young do not experience the challenges themselves
(Figure 6). Inheritance of acquired traits might offer an advantage to the population by increasing
the fitness of the species and by enabling adaptation to variations in the environment at a faster
rate than natural selection would allow. Such nongenetic inheritance of acquired behavioral
traits is reminiscent of the ideas put forth by Jean-Baptiste Lamarck in his 1809 book, Philosophie
Zoologique; these ideas, however, have been widely discredited. Rather, the transgenerational
transmission of behavioral traits and psychiatric disorders in humans is believed to be mediated
via genetic mechanisms working in concert with parenting and culture. Nevertheless, recent
advances in epigenetics have renewed interest in the possibility of nongenetic, nonbehavioral

74 Vialou et al.
PA53CH04-Nestler ARI 5 December 2012 12:48

Adult
Exposure to stressful events alters
chromatin composition in neurons,
thus affecting neuronal function
Possible influence on germ cells,
although the mediator of such
effects remains unknown

Early life Gametes and zygote


Prenatal stress, differential Possible inheritance of
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

levels of maternal care, and epigenetic modifications


through ncRNAs,
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

early-life stress alter brain


development and have DNA methylation, and
long-term effects on stress histone modification
reactivity and cognition

Figure 6
Contribution of epigenetic modifications throughout life cycle. Environmental challenges, such as stress,
have been associated with several short- and long-term chromatin modifications in the brain that affect neural
development and function. Gametes and primordial germ cells are also sensitive to stress and might carry
epigenetic transformation through subsequent generations. The development of germ cells is characterized
by the genome-wide demethylation of DNA in gametes and the establishment of parental chromatin marks
required for genomic imprinting. Following fertilization, another genome-wide DNA demethylation phase
occurs. However, some epigenetic marks (e.g., imprinted genes, DNA methylation at a specific location,
some histone posttranslational modifications) escape such reprogramming. Moreover, thousands of
noncoding RNAs (ncRNAs) that are present in the gametes could transmit their information and potentially
affect the cellular phenotype in a nongenomic manner. Modified with permission from Reference 41.

transmission of behavioral experience via epigenetic mechanisms through the germline. Such
use of the term epigenetics, to be distinguished from its use to describe epigenetic mechanisms
occurring within a single organism, continues to cause confusion in the field (172).
Several groups have begun to explore the transmission of stress-related pathologies to subse-
quent generations via epigenetic mechanisms. Franklin et al. (173) showed that most of the behav-
ioral alterations induced by early-life maternal separation are expressed by the F1 offspring of males
subjected to maternal separation. These male F1 offspring, when bred with normal females, trans-
mitted their behavioral abnormalities almost exclusively to female offspring for two generations
(F2 and F3). Maternal separation was reported to alter DNA methylation levels in the promoter
regions of the genes encoding MeCP2, the CRF2 receptor, and the cannabinoid receptor-1 in the
germline of the separated males. Similar DNA methylation patterns were observed in the cere-
bral cortex of female offspring. No differences in maternal care were observed, suggesting that
stress-related behavior was transmitted in a paternal epigenetic but sexually dimorphic manner. In
a model of early-life stress with impaired maternal care, abnormal DNA methylation and expres-
sion patterns of BDNF in the prefrontal cortex are induced for up to two generations (146). Such
effects on methylation patterns are not totally reversed by cross-fostering, suggesting that some of
these effects do not result from postnatal experiences. However, females with a history of early-life
maltreatment show increased anxiety while pregnant, which could influence fetus development.
Although prenatal and postnatal effects cannot be ruled out, transgenerational perpetuation of

www.annualreviews.org • Epigenetic Mechanisms of Depression 75


PA53CH04-Nestler ARI 5 December 2012 12:48

DNA methylation changes could be attributable to epigenetic gametic inheritance as well. Prena-
tal stress during the early gestation stage leads to dysmasculinized male offspring with altered stress
responsiveness (174, 175). This effect was transmitted to the F2 male generation and was associ-
ated with a reduction in miRNA levels in the brains of male neonates (176). These miRNAs were
sensitive to testosterone levels and may play a role in organizing the brain’s sexual dimorphism.
Although these studies support the robust transmission of behavioral experience through the
germline, different effects were obtained in a model of adult exposure to stress. Dietz et al. (177)
first showed that dramatic depression- and anxiety-like behavioral abnormalities induced in adult
male mice by chronic social-defeat stress were transmitted to both their male and female off-
spring. Abnormalities in peripheral biomarkers of chronic stress (plasma levels of BDNF, vascular
endothelial growth factor, and corticosterone) were also transmitted to the offspring. However,
the vast majority of these abnormalities were not observed in offspring of socially defeated males
generated by in vitro fertilization; only subtle behavioral effects were apparent. These data suggest
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

that the primary mode of transmission of the effects of chronic stress may be behavioral rather
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

than epigenetic, perhaps because female mice adjust their reproductive investment depending on
interactions with an impaired male; such adjustment is a known phenomenon (178). In contrast,
the process of in vitro fertilization may have corrupted any epigenetic marks in the sperm of
defeated males. The discrepancies between this study of adult mice and those involving early-life
stress raise the possibility that epigenetic mechanisms may be more important when they occur
during development.
There are several reports of the transgenerational epigenetic transmission of other behavioral
experiences, such as diet, drugs of abuse, and chemical toxins. Exposure to cocaine in adult male
mice mated with naive females resulted in offspring that displayed deficits in sustained attention and
spatial working memory performance (179). Paternal cocaine exposure is associated with a decrease
in DNMT1 and an increase in DNMT3 levels in spermatozoa, which might cause alterations in
the DNA methylation of certain genes. Maternal high-fat diet resulted in increased body size
and reduced insulin sensitivity in offspring, and these traits persisted across multiple generations
(180). At the third generation, this phenotype was transmitted only to female offspring through
the paternal lineage, suggesting a stable germline-based transgenerational mode of inheritance
(181). This effect might be the consequence of the regulation of paternally imprinted genes that
influence developmental changes in growth and body size and that result in paternal transmission
specifically to daughters (181). Similarly, exposure to endocrine disruptors (vinclozolin) affects
DNA methylation at imprinted genes in the male germline, leading to behavioral and physiological
abnormalities that are transgenerationally persistent (182–184).
What are the most plausible mechanisms for such epigenetic heritability? Epigenetic marks
such as DNA methylation and histone modifications are extensively erased in primordial germ cells
during gametogenesis in mice (E10.5–E13.5) and immediately after fertilization in early embryo-
genesis to ensure the totipotency of cells in the early embryo. This epigenetic reprogramming
limits the potential in mammals for epigenetic transgenerational inheritance. However, mam-
mals have a few known epigenetic marks that are resistant to both phases of reprogramming:
transgenes, retrotransposons, and heterochromatin at centromeres (172, 178, 185–187). During
spermatogenesis, most of the histones are replaced by protamines, but some histones are retained
in a locus-specific manner and could play a role in early embryo development (188). If retained af-
ter fertilization, these epigenetic modifications could participate in the transgenerational transfer
of information. Another possible mechanism of transgenerational phenotypic persistence occurs
through cytoplasmic RNAs, including mRNA, siRNA, piRNA, and miRNA (189). Indeed, RNAs
from gametes allow the newly fertilized oocyte to initiate transcription and have been observed in
large quantity in human sperm (190).

76 Vialou et al.
PA53CH04-Nestler ARI 5 December 2012 12:48

Only maternally derived imprinted genes are protected from demethylation after fertilization
(191). Therefore, altered DNA methylation of these imprinted genes cannot persist transgen-
erationally unless new methylation sites are induced by environmental factors during critical
developmental periods. Genomic imprinting is a form of epigenetic regulation that distinguishes
the maternal and paternal genomes, leading to a bias in expression between maternally and pa-
ternally transmitted genes. Imprinted genes are silenced through DNA methylation and through
related chromatin modifications that have been established in the germline (192). Proper ex-
pression of imprinted genes is essential for embryonic development (193) and numerous post-
natal functions (46, 194–196). Not surprisingly, genetic abnormalities involving imprinted genes
cause developmental disorders such as Prader-Willi and Angelman syndromes. In addition to
gene imprinting and X-chromosomal inactivation, whereby DNA methylation coordinates the
random silencing of either X chromosome in females, growing evidence suggests that allele-
specific methylation occurs across the entire genome and is heterogeneous across tissues and
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

individuals (197, 198). Most of this allele-specific methylation is determined by DNA sequence,
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

hence showing Mendelian inheritance patterns. Furthermore, allele-specific methylation occurs


through non-cis events, which might contribute to the heritability of complex diseases (199). In-
deed, a recent genome-wide screen indicated that more than 1,300 loci show parental-origin allelic
expression bias with complex regulation throughout brain development in a cell-type and gender-
specific manner (200, 201). Moreover, DNA methylation at some nonimprinted genes escapes
the postfertilization DNA methylation reprogramming (202). Therefore, allele-specific methyla-
tion could contribute significantly to the heritability of complex etiological disorders, including
depression and other stress-related disorders, and could explain the significant discordance of
psychiatric disorders between monozygotic twins, the difference in gender susceptibility to cer-
tain psychiatric diseases, and the largely inconsistent genetic association studies of depression (50,
51).
This discussion highlights the difficulties in defining epigenetic inheritance of stressful expe-
riences. First, it is challenging to fully control for parental behavior. As noted, maternal behavior
may be affected by the behavior of the impregnating male. Second, although cross-fostering can
control for postnatal maternal care, it does not control for intrauterine factors, which might also
be crucial. Third, in vitro fertilization is not a perfect test for germline mechanisms because the
epigenetic state of sperm may vary as a function of how and when it is harvested with respect to the
stress exposure. Fourth, semen might contain other factors that influence the mother or offspring.
Ultimately, to establish epigenetic mechanisms for the transgenerational transmission of stressful
experiences, it will be essential to demonstrate epigenetic modifications at specific genes in sperm
or egg cells and to understand how they are transmitted into alterations in neural function in adult
offspring across several generations. No mechanisms by which such transmission can occur are
known; identifying them is a challenge for future research.

CONCLUSIONS
Epigenetic investigations promise to improve our understanding of the mechanisms by which
individuals exhibit widely varying responses to adverse life events, both during development and
in adulthood. Animal models of stress-related disorders are beginning to reveal specific chromatin
modifications that maintain stable patterns of gene expression and thereby mediate the aberrant
neuroplasticity associated with these disorders. Equivalent studies have focused on chromatin
modifications that contribute to antidepressant responses. Several different types of epigenetic
mechanisms have been implicated, including stress- or antidepressant-induced changes in DNA
methylation, histone acetylation and methylation, and miRNAs. However, the modifications

www.annualreviews.org • Epigenetic Mechanisms of Depression 77


PA53CH04-Nestler ARI 5 December 2012 12:48

examined to date represent the tip of the iceberg of the diverse and complex range of epigenetic
mechanisms that are likely involved in stress and antidepressant action.
Epigenetic mechanisms are regulated throughout life in neurons, and some stress-sensitive time
windows, such as early life, are particularly important. Although several specific genes have been
identified as being permanently regulated (e.g., GR in hippocampus, CRF and AVP in PVN),
many other genes probably play a role in stress vulnerability as well. Moreover, in addition to
sustained alterations in steady-state mRNA levels of certain genes, early-life stress probably also
modifies the transcriptional potential of many additional genes through their epigenetic priming
or desensitization. Identifying all of the genes that are epigenetically regulated by stress, and that
are important for controlling stress vulnerability across the life cycle, will therefore require the
superimposition of several levels of analysis. These analyses include genome-wide analyses of RNA
expression along with those of DNA methylation, histone modifications, chromatin remodeling
factors, and transcription factors. Such work will also require the optimization of bioinformatics
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

tools that enable the examination of the vast data sets involved.
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

This endeavor is further complicated by the large number of brain regions involved in stress-
related psychopathology. Most studies have focused by necessity (e.g., cost) on single brain regions,
despite the knowledge that depression and anxiety disorders involve abnormal functioning of nu-
merous brain circuits. Not only will each brain region have to be investigated independently, but
several distinct neuronal and non-neuronal cell types within each region will have to be charac-
terized separately. For example, epigenetic modifications that occur in glutamatergic pyramidal
neurons in prefrontal cortex are likely very different from those that occur in several different sub-
types of GABAergic interneurons, astrocytes, and oligodendrocytes. Fortunately, we are seeing
the emergence of tools that permit the cell type–specific investigation of chromatin modifications
in a heterogeneous tissue such as brain (203).
For a full understanding of epigenetic mechanisms of stress and antidepressant action, the field
must transition to genome-wide approaches, most particularly those that utilize massive parallel
sequencing following ChIP (ChIP-seq), RNA isolation (RNA-seq), or selective DNA isolation
(e.g., for methylcytosine). These recently developed methodologies provide whole-genome cov-
erage as well as improved sensitivity compared with that provided by older ChIP-chip and other
microarray approaches. As noted above, the combination of numerous chromatin modifications
(204) will allow the delineation of the stress and antidepressant epigenomes. As our knowledge
advances, we will identify other types of chromatin modifications and epigenetic regulation that
are sensitive to the environment, such as alterations in retrotransposons (205–207). Like cocaine
exposure (208), stress exposure might increase genomic insertion of retrotransposable elements
that regulate the transcriptome (209). A newly recognized form of DNA methylation, 5hmC, is
highly abundant in neurons and predominantly localized around the coding region in association
with other histone repressive marks, suggesting that it might participate in the epigenetic control
of neuronal function (70, 210). These are just some examples of the new lines of investigation that
could potentially reveal fundamentally novel mechanisms that control stress-related phenomena.
Finally, there has been interest, as noted in this review, in the possible therapeutic applications
of drugs aimed at chromatin-modifying enzymes (211). For example, HDAC inhibitors exert
potent antidepressant-like responses in diverse animal models. Whether such drugs offer realistic
possibilities for drug discovery remains uncertain, given that most chromatin-modifying proteins
are broadly expressed throughout the brain and peripheral tissues. Nevertheless, hundreds of
chromatin regulatory proteins are known; targeting those proteins that are enriched in brain is
a viable path forward. Moreover, there is no question that epigenetic characterization of stress
models will reveal a far more complete view of the range of proteins and ncRNAs that mediate
stress-induced pathology, the reversal of that pathology during antidepressant treatment, and the

78 Vialou et al.
PA53CH04-Nestler ARI 5 December 2012 12:48

resistance against developing such pathology in resilient individuals. This knowledge will provide
for the first time a comprehensive road map for future drug discovery efforts.

DISCLOSURE STATEMENT
E.J.N. receives consulting income from Merck Research Laboratories, PsychoGenics, Berg
Pharma, and Johnson & Johnson. The other authors report no affiliations, memberships, funding,
or financial holdings that might be perceived as affecting the objectivity of this review.

LITERATURE CITED
1. de Kloet ER, Joëls M, Holsboer F. 2005. Stress and the brain: from adaptation to disease. Nat. Rev.
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

Neurosci. 6:463–75
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

2. Chrousos GP. 2009. Stress and disorders of the stress system. Nat. Rev. Endocrinol. 5:374–81
3. Shin LM, Liberzon I. 2009. The neurocircuitry of fear, stress, and anxiety disorders. Neuropsychophar-
macology 35:169–91
4. McEwen BS. 2008. Central effects of stress hormones in health and disease: understanding the protective
and damaging effects of stress and stress mediators. Eur. J. Pharmacol. 583:174–85
5. von Holst D. 1998. The concept of stress and its relevance for animal behavior. Adv. Study Behav.
27:1–131
6. Hackman DA, Farah MJ, Meaney MJ. 2010. Socioeconomic status and the brain: mechanistic insights
from human and animal research. Nat. Rev. Neurosci. 11:651–59
7. Shonkoff JP, Boyce WT, McEwen BS. 2009. Neuroscience, molecular biology, and the childhood roots
of health disparities: building a new framework for health promotion and disease prevention. JAMA
301:2252–59
8. Yehuda R, McFarlane AC, Shalev AY. 1998. Predicting the development of posttraumatic stress disorder
from the acute response to a traumatic event. Biol. Psychiatry 44:1305–13
9. Krishnan V, Han MH, Graham DL, Berton O, Renthal W, et al. 2007. Molecular adaptations underlying
susceptibility and resistance to social defeat in brain reward regions. Cell 131:391–404
10. Feder A, Nestler EJ, Charney DS. 2009. Psychobiology and molecular genetics of resilience. Nat. Rev.
Neurosci. 10:446–57
11. Lyons DM, Parker KJ, Schatzberg AF. 2010. Animal models of early life stress: implications for under-
standing resilience. Dev. Psychobiol. 52:616–24
12. Uher R. 2011. Genes, environment, and individual differences in responding to treatment for depression.
Harv. Rev. Psychiatry 19:109–24
13. Kendler KS, Halberstadt LJ. 2012. The road not taken: life experiences in monozygotic twin pairs
discordant for major depression. Mol. Psychiatry. In press, doi: 10.1038/mp.2012.55
14. Hyman SE, Nestler EJ. 1996. Initiation and adaptation: a paradigm for understanding psychotropic drug
action. Am. J. Psychiatry 153:151–62
15. Duman RS, Monteggia LM. 2006. A neurotrophic model for stress-related mood disorders. Biol. Psychi-
atry 59:1116–27
16. Berton O, McClung CA, Dileone RJ, Krishnan V, Renthal W, et al. 2006. Essential role of BDNF in
the mesolimbic dopamine pathway in social defeat stress. Science 311:864–68
17. Gould TD, O’Donnell KC, Picchini AM, Dow ER, Chen G, Manji HK. 2008. Generation and behavioral
characterization of β-catenin forebrain-specific conditional knock-out mice. Behav. Brain Res. 189:117–
25
18. Gould TD, Einat H, O’Donnell KC, Picchini AM, Schloesser RJ, Manji HK. 2007. β-catenin overexpres-
sion in the mouse brain phenocopies lithium-sensitive behaviors. Neuropsychopharmacology 32:2173–83
19. Okamoto H, Voleti B, Banasr M, Sarhan M, Duric V, et al. 2010. Wnt2 expression and signaling is
increased by different classes of antidepressant treatments. Biol. Psychiatry 68:521–27

www.annualreviews.org • Epigenetic Mechanisms of Depression 79


PA53CH04-Nestler ARI 5 December 2012 12:48

20. Wilkinson MB, Dias C, Magida J, Mazei-Robison M, Lobo M, et al. 2011. A novel role of the WNT-
dishevelled-GSK3β signaling cascade in the mouse nucleus accumbens in a social defeat model of
depression. J. Neurosci. 31:9084–92
21. Liu D, Diorio J, Tannenbaum B, Caldji C, Francis D, et al. 1997. Maternal care, hippocampal gluco-
corticoid receptors, and hypothalamic-pituitary-adrenal responses to stress. Science 277:1659–62
22. Karssen AM, Her S, Li JZ, Patel PD, Meng F, et al. 2007. Stress-induced changes in primate prefrontal
profiles of gene expression. Mol. Psychiatry 12:1089–102
23. Surget A, Wang Y, Leman S, Ibarguen-Vargas Y, Edgar N, et al. 2009. Corticolimbic transcriptome
changes are state-dependent and region-specific in a rodent model of depression and of antidepressant
reversal. Neuropsychopharmacology 34:1363–80
24. Andrus BM, Blizinsky K, Vedell PT, Dennis K, Shukla PK, et al. 2012. Gene expression patterns in
the hippocampus and amygdala of endogenous depression and chronic stress models. Mol. Psychiatry
17:49–61
25. Bernard R, Kerman IA, Thompson RC, Jones EG, Bunney WE, et al. 2011. Altered expression of
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

glutamate signaling, growth factor, and glia genes in the locus coeruleus of patients with major depression.
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

Mol. Psychiatry 16:634–46


26. Covington HE III, Maze I, LaPlant Q, Vialou V, Ohnishi Y, et al. 2009. Antidepressant actions of histone
deacetylase inhibitors. J. Neurosci. 22:11451–60
27. Duric V, Banasr M, Licznerski P, Schmidt HD, Stockmeier CA, et al. 2010. A negative regulator of
MAP kinase causes depressive behavior. Nat. Med. 16:1328–32
28. Le-Niculescu H, Patel SD, Niculescu AB. 2010. Convergent integration of animal model and human
studies of bipolar disorder (manic-depressive illness). Curr. Opin. Pharmacol. 10:594–600
29. Shelton RC, Claiborne J, Sidoryk-Wegrzynowicz M, Reddy R, Aschner M, et al. 2011. Altered expression
of genes involved in inflammation and apoptosis in frontal cortex in major depression. Mol. Psychiatry
16:751–62
30. Vialou V, Robison AJ, LaPlant QC, Covington HE III, Dietz DM, et al. 2010. FosB in brain reward
circuits mediates resilience to stress and antidepressant responses. Nat. Neurosci. 13:745–52
31. Elliott E, Ezra-Nevo G, Regev L, Neufeld-Cohen A, Chen A. 2010. Resilience to social stress coincides
with functional DNA methylation of the Crf gene in adult mice. Nat. Neurosci. 13:1351–3
32. Gibney ER, Nolan CM. 2010. Epigenetics and gene expression. Heredity 105:4–13
33. Taft RJ, Pang KC, Mercer TR, Dinger M, Mattick JS. 2010. Non-coding RNAs: regulators of disease.
J. Pathol. 220:126–39
34. Bird A. 2007. Perceptions of epigenetics. Nature 447:396–98
35. Van Speybroeck L. 2002. From epigenesis to epigenetics: the case of C. H. Waddington. Ann. N. Y.
Acad. Sci. 981:61–81
36. Borrelli E, Nestler EJ, Allis CD, Sassone-Corsi P. 2008. Decoding the epigenetic language of neuronal
plasticity. Neuron 60:961–74
37. Costa-Mattioli M, Sossin WS, Klann E, Sonenberg N. 2009. Translational control of long-lasting synap-
tic plasticity and memory. Neuron 61:10–26
38. Jiang Y, Langley B, Lubin FD, Renthal W, Wood MA, et al. 2008. Epigenetics in the nervous system.
J. Neurosci. 28:11753–59
39. Neves G, Cooke SF, Bliss TV. 2008. Synaptic plasticity, memory and the hippocampus: a neural network
approach to causality. Nat. Rev. Neurosci. 9:65–75
40. Roth TL, Sweatt JD. 2009. Regulation of chromatin structure in memory formation. Curr. Opin. Neu-
robiol. 19:336–42
41. Dulac C. 2010. Brain function and chromatin plasticity. Nature 465:728–35
42. Korzus E, Rosenfeld MG, Mayford M. 2004. CBP histone acetyltransferase activity is a critical compo-
nent of memory consolidation. Neuron 42:961–72
43. Levenson JM, O’Riordan KJ, Brown KD, Trinh MA, Molfese DL, Sweatt JD. 2004. Regulation of
histone acetylation during memory formation in the hippocampus. J. Biol. Chem. 279:40545–59
44. Levenson JM, Sweatt JD. 2005. Epigenetic mechanisms in memory formation. Nat. Rev. Neurosci. 6:108–
18

80 Vialou et al.
PA53CH04-Nestler ARI 5 December 2012 12:48

45. Champagne FA, Weaver IC, Diorio J, Dymov S, Szyf M, Meaney MJ. 2006. Maternal care associated
with methylation of the estrogen receptor-α1b promoter and estrogen receptor-α expression in the
medial preoptic area of female offspring. Endocrinology 147:2909–15
46. Garfield AS, Cowley M, Smith FM, Moorwood K, Stewart-Cox JE, et al. 2011. Distinct physiological
and behavioural functions for parental alleles of imprinted Grb10. Nature 469:534–38
47. Maze I, Covington HE III, Dietz DM, LaPlant Q, Renthal W, et al. 2010. Essential role of the histone
methyltransferase G9a in cocaine-induced plasticity. Science 327:213–16
48. Robison AJ, Nestler EJ. 2011. Transcriptional and epigenetic mechanisms of addiction. Nat. Rev. Neurosci.
12:623–37
49. Chertkow-Deutsher Y, Cohen H, Klein E, Ben-Shachar D. 2010. DNA methylation in vulnerability to
post-traumatic stress in rats: evidence for the role of the post-synaptic density protein Dlgap2. Int. J.
Neuropsychopharmacol. 13:347–59
50. Feinberg AP. 2007. Phenotypic plasticity and the epigenetics of human disease. Nature 447:433–40
51. Mill J, Petronis A. 2007. Molecular studies of major depressive disorder: the epigenetic perspective.
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

Mol. Psychiatry 12:799–814


by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

52. Hobara T, Uchida S, Otsuki K, Matsubara T, Funato H, et al. 2009. Altered gene expression of histone
deacetylases in mood disorder patients. J. Psychiatr. Res. 44:263–70
53. Poulter MO, Du L, Weaver IC, Palkovits M, Faludi G, et al. 2008. GABAA receptor promoter hyper-
methylation in suicide brain: implications for the involvement of epigenetic processes. Biol. Psychiatry
64:645–52
54. Keller S, Sarchiapone M, Zarrilli F, Videtic A, Ferraro A, et al. 2010. Increased BDNF promoter methy-
lation in the Wernicke area of suicide subjects. Arch. Gen. Psychiatry 67:258–67
55. Newell-Price J, Clark AJ, King P. 2000. DNA methylation and silencing of gene expression. Trends
Endocrinol. Metab. 11:142–48
56. Nafee TM, Farrell WE, Carroll WD, Fryer AA, Ismail KM. 2008. Epigenetic control of fetal gene
expression. BJOG 115:158–68
57. Bird A. 2008. The methyl-CpG-binding protein MeCP2 and neurological disease. Biochem. Soc. Trans.
36:575–83
58. Wang Y, Leung FC. 2004. An evaluation of new criteria for CpG islands in the human genome as gene
markers. Bioinformatics 20:1170–77
59. Prokhortchouk E, Defossez PA. 2008. The cell biology of DNA methylation in mammals. Biochim.
Biophys. Acta 1783:2167–73
60. Weber M, Schubeler D. 2007. Genomic patterns of DNA methylation: targets and function of an
epigenetic mark. Curr. Opin. Cell Biol. 19:273–80
61. Kim JK, Samaranayake M, Pradhan S. 2009. Epigenetic mechanisms in mammals. Cell. Mol. Life Sci.
66:596–612
62. Bhattacharya SK, Ramchandani S, Cervoni N, Szyf M. 1999. A mammalian protein with specific
demethylase activity for mCpG DNA. Nature 397:579–83
63. Barreto G, Schafer A, Marhold J, Stach D, Swaminathan SK, et al. 2007. Gadd45a promotes epigenetic
gene activation by repair-mediated DNA demethylation. Nature 445:671–75
64. Ma DK, Jang MH, Guo JU, Kitabatake Y, Chang ML, et al. 2009. Neuronal activity–induced Gadd45b
promotes epigenetic DNA demethylation and adult neurogenesis. Science 323:1074–77
65. Tahiliani M, Koh KP, Shen Y, Pastor WA, Bandukwala H, et al. 2009. Conversion of 5-methylcytosine
to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 324:930–35
66. Kriaucionis S, Heintz N. 2009. The nuclear DNA base 5-hydroxymethylcytosine is present in Purkinje
neurons and the brain. Science 324:929–30
67. Ito S, Shen L, Dai Q, Wu SC, Collins LB, et al. 2011. Tet proteins can convert 5-methylcytosine to
5-formylcytosine and 5-carboxylcytosine. Science 333:1300–3
68. He YF, Li BZ, Li Z, Liu P, Wang Y, et al. 2011. Tet-mediated formation of 5-carboxylcytosine and its
excision by TDG in mammalian DNA. Science 333:1303–7
69. Guo JU, Su Y, Zhong C, Ming GL, Song H. 2011. Hydroxylation of 5-methylcytosine by TET1
promotes active DNA demethylation in the adult brain. Cell 145:423–34

www.annualreviews.org • Epigenetic Mechanisms of Depression 81


PA53CH04-Nestler ARI 5 December 2012 12:48

70. Szulwach KE, Li X, Li Y, Song CX, Wu H, et al. 2011. 5-hmC-mediated epigenetic dynamics during
postnatal neurodevelopment and aging. Nat. Neurosci. 14:1607–16
71. Hervouet E, Vallette FM, Cartron PF. 2009. Dnmt3/transcription factor interactions as crucial players
in targeted DNA methylation. Epigenetics 4:487–99
72. Amir RE, Van den Veyver IB, Wan M, Tran CQ, Francke U, Zoghbi HY. 1999. Rett syndrome is caused
by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2. Nat. Genet. 23:185–88
73. Nan X, Hou J, Maclean A, Nasir J, Lafuente MJ, et al. 2007. Interaction between chromatin proteins
MECP2 and ATRX is disrupted by mutations that cause inherited mental retardation. Proc. Natl. Acad.
Sci. USA 104:2709–14
74. Mori T, Li Y, Hata H, Ono K, Kochi H. 2002. NIRF, a novel RING finger protein, is involved in
cell-cycle regulation. Biochem. Biophys. Res. Commun. 296:530–36
75. Feng J, Zhou Y, Campbell SL, Le T, Li E, et al. 2010. Dnmt1 and Dnmt3a maintain DNA methylation
and regulate synaptic function in adult forebrain neurons. Nat. Neurosci. 13:423–30
76. Miller CA, Sweatt JD. 2007. Covalent modification of DNA regulates memory formation. Neuron
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

53:857–69
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

77. Sharma RP, Gavin DP, Grayson DR. 2010. CpG methylation in neurons: message, memory, or mask?
Neuropsychopharmacology 35:2009–20
78. Feng J, Fan G. 2009. The role of DNA methylation in the central nervous system and neuropsychiatric
disorders. Int. Rev. Neurobiol. 89:67–84
79. Day JJ, Sweatt JD. 2011. Epigenetic mechanisms in cognition. Neuron 70:813–29
80. Berger SL. 2007. The complex language of chromatin regulation during transcription. Nature 447:407–
12
81. Strahl BD, Allis CD. 2000. The language of covalent histone modifications. Nature 403:41–45
82. Cheung P, Allis CD, Sassone-Corsi P. 2000. Signaling to chromatin through histone modifications. Cell
103:263–71
83. Nakahata Y, Sahar S, Astarita G, Kaluzova M, Sassone-Corsi P. 2009. Circadian control of the NAD+
salvage pathway by CLOCK-SIRT1. Science 324:654–57
84. Kaeberlein M, McVey M, Guarente L. 1999. The SIR2/3/4 complex and SIR2 alone promote longevity
in Saccharomyces cerevisiae by two different mechanisms. Genes Dev. 13:2570–80
85. Sassone-Corsi P. 2012. Minireview: NAD+, a circadian metabolite with an epigenetic twist. Endocrinology
153:1–5
86. Bellet MM, Sassone-Corsi P. 2010. Mammalian circadian clock and metabolism—the epigenetic link.
J. Cell Sci. 123:3837–48
87. Rice JC, Allis CD. 2001. Histone methylation versus histone acetylation: new insights into epigenetic
regulation. Curr. Opin. Cell Biol. 13:263–73
88. Sun ZW, Allis CD. 2002. Ubiquitination of histone H2B regulates H3 methylation and gene silencing
in yeast. Nature 418:104–8
89. Jenuwein T, Allis CD. 2001. Translating the histone code. Science 293:1074–80
90. Qureshi IA, Mattick JS, Mehler MF. 2010. Long non-coding RNAs in nervous system function and
disease. Brain Res. 1338:20–35
91. Wang KC, Chang HY. 2011. Molecular mechanisms of long noncoding RNAs. Mol. Cell 43:904–14
92. Nagano T, Fraser P. 2011. No-nonsense functions for long noncoding RNAs. Cell 145:178–81
93. Bartel DP. 2009. MicroRNAs: target recognition and regulatory functions. Cell 136:215–33
94. Im HI, Hollander JA, Bali P, Kenny PJ. 2010. MeCP2 controls BDNF expression and cocaine intake
through homeostatic interactions with microRNA-212. Nat. Neurosci. 13:1120–27
95. Djupedal I, Ekwall K. 2009. Epigenetics: heterochromatin meets RNAi. Cell Res. 19:282–95
96. Vastenhouw NL, Brunschwig K, Okihara KL, Muller F, Tijsterman M, Plasterk RH. 2006. Gene
expression: long-term gene silencing by RNAi. Nature 442:882
97. Jirtle RL, Skinner MK. 2007. Environmental epigenomics and disease susceptibility. Nat. Rev. Genet.
8:253–62
98. Meaney MJ, Szyf M. 2005. Environmental programming of stress responses through DNA methylation:
life at the interface between a dynamic environment and a fixed genome. Dialogues Clin. Neurosci. 7:103–23

82 Vialou et al.
PA53CH04-Nestler ARI 5 December 2012 12:48

99. Fraga MF, Ballestar E, Paz MF, Ropero S, Setien F, et al. 2005. Epigenetic differences arise during the
lifetime of monozygotic twins. Proc. Natl. Acad. Sci. USA 102:10604–9
100. Peleg S, Sananbenesi F, Zovoilis A, Burkhardt S, Bahari-Javan S, et al. 2010. Altered histone acetylation
is associated with age-dependent memory impairment in mice. Science 328:753–56
101. Michan S, Li Y, Chou MM, Parrella E, Ge H, et al. 2010. SIRT1 is essential for normal cognitive
function and synaptic plasticity. J. Neurosci. 30:9695–707
102. Fyffe SL, Neul JL, Samaco RC, Chao HT, Ben-Shachar S, et al. 2008. Deletion of Mecp2 in Sim1-
expressing neurons reveals a critical role for MeCP2 in feeding behavior, aggression, and the response
to stress. Neuron 59:947–58
103. Sterrenburg L, Gaszner B, Boerrigter J, Santbergen L, Bramini M, et al. 2011. Chronic stress induces
sex-specific alterations in methylation and expression of corticotropin-releasing factor gene in the rat.
PLoS ONE 6:e28128
104. Schmidt HD, Duman RS. 2007. The role of neurotrophic factors in adult hippocampal neurogenesis,
antidepressant treatments and animal models of depressive-like behavior. Behav. Pharmacol. 18:391–418
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

105. Roth TL, Zoladz PR, Sweatt JD, Diamond DM. 2011. Epigenetic modification of hippocampal Bdnf
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

DNA in adult rats in an animal model of post-traumatic stress disorder. J. Psychiatr. Res. 45:919–26
106. Sapolsky RM. 1996. Stress, glucocorticoids, and damage to the nervous system: the current state of
confusion. Stress 1:1–19
107. Gould E, McEwen BS, Tanapat P, Galea LA, Fuchs E. 1997. Neurogenesis in the dentate gyrus of
the adult tree shrew is regulated by psychosocial stress and NMDA receptor activation. J. Neurosci.
17:2492–98
108. Uchida S, Hara K, Kobayashi A, Otsuki K, Yamagata H, et al. 2011. Epigenetic status of Gdnf in the
ventral striatum determines susceptibility and adaptation to daily stressful events. Neuron 69:359–72
109. Chahrour M, Jung SY, Shaw C, Zhou X, Wong ST, et al. 2008. MeCP2, a key contributor to neurological
disease, activates and represses transcription. Science 320:1224–29
110. Melas PA, Rogdaki M, Lennartsson A, Bjork K, Qi H, et al. 2011. Antidepressant treatment is associated
with epigenetic alterations in the promoter of P11 in a genetic model of depression. Int. J. Neuropsy-
chopharmacol. 15:669–79
111. Svenningsson P, Chergui K, Rachleff I, Flajolet M, Zhang X, et al. 2006. Alterations in 5-HT1B receptor
function by p11 in depression-like states. Science 311:77–80
112. Anisman H, Du L, Palkovits M, Faludi G, Kovacs GG, et al. 2008. Serotonin receptor subtype and p11
mRNA expression in stress-relevant brain regions of suicide and control subjects. J. Psychiatry Neurosci.
33:131–41
113. Uddin M, Koenen KC, Aiello AE, Wildman DE, de los Santos R, Galea S. 2011. Epigenetic and
inflammatory marker profiles associated with depression in a community-based epidemiologic sample.
Psychol. Med. 41:997–1007
114. Schmidt HD, Shelton RC, Duman RS. 2011. Functional biomarkers of depression: diagnosis, treatment,
and pathophysiology. Neuropsychopharmacology 36:2375–94
115. LaPlant Q, Vialou V, Covington HE III, Dumitriu D, Feng J, et al. 2010. Dnmt3a regulates emotional
behavior and spine plasticity in the nucleus accumbens. Nat. Neurosci. 13:1137–43
116. Covington HE III, Vialou VF, LaPlant Q, Ohnishi YN, Nestler EJ. 2011. Hippocampal-dependent
antidepressant-like activity of histone deacetylase inhibition. Neurosci. Lett. 493:122–26
117. Hinwood M, Tynan RJ, Day TA, Walker FR. 2011. Repeated social defeat selectively increases FosB
expression and histone H3 acetylation in the infralimbic medial prefrontal cortex. Cereb. Cortex 21:262–71
118. Hollis F, Duclot F, Gunjan A, Kabbaj M. 2011. Individual differences in the effect of social defeat on
anhedonia and histone acetylation in the rat hippocampus. Horm. Behav. 59:331–37
119. Hollis F, Wang H, Dietz D, Gunjan A, Kabbaj M. 2010. The effects of repeated social defeat on
long-term depressive-like behavior and short-term histone modifications in the hippocampus in male
Sprague-Dawley rats. Psychopharmacology 211:69–77
120. Tsankova NM, Berton O, Renthal W, Kumar A, Neve RL, Nestler EJ. 2006. Sustained hippocampal
chromatin regulation in a mouse model of depression and antidepressant action. Nat. Neurosci. 9:519–25
121. Schroeder FA, Lin CL, Crusio WE, Akbarian S. 2007. Antidepressant-like effects of the histone deacety-
lase inhibitor, sodium butyrate, in the mouse. Biol. Psychiatry 62:55–64

www.annualreviews.org • Epigenetic Mechanisms of Depression 83


PA53CH04-Nestler ARI 5 December 2012 12:48

122. Tsankova N, Renthal W, Kumar A, Nestler EJ. 2007. Epigenetic regulation in psychiatric disorders.
Nat. Rev. Neurosci. 8:355–67
123. Gundersen BB, Blendy JA. 2009. Effects of the histone deacetylase inhibitor sodium butyrate in models
of depression and anxiety. Neuropharmacology 57:67–74
124. Tsankova NM, Kumar A, Nestler EJ. 2004. Histone modifications at gene promoter regions in rat
hippocampus after acute and chronic electroconvulsive seizures. J. Neurosci. 24:5603–10
125. Covington HE III, Maze I, Sun H, Bomze HM, DeMaio KD, et al. 2011. A role for repressive histone
methylation in cocaine-induced vulnerability to stress. Neuron 71:656–70
126. Hunter RG, McCarthy KJ, Milne TA, Pfaff DW, McEwen BS. 2009. Regulation of hippocampal H3
histone methylation by acute and chronic stress. Proc. Natl. Acad. Sci. USA 106:20912–17
127. Wilkinson MB, Xiao G, Kumar A, LaPlant Q, Renthal W, et al. 2009. Imipramine treatment and
resiliency exhibit similar chromatin regulation in the mouse nucleus accumbens in depression models.
J. Neurosci. 29:7820–32
128. Wallace DL, Han MH, Graham DL, Green TA, Vialou V, et al. 2009. CREB regulation of nucleus
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

accumbens excitability mediates social isolation–induced behavioral deficits. Nat. Neurosci. 12:200–9
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

129. Jiang Y, Jakovcevski M, Bharadwaj R, Connor C, Schroeder FA, et al. 2010. Setdb1 histone methyl-
transferase regulates mood-related behaviors and expression of the NMDA receptor subunit NR2B.
J. Neurosci. 30:7152–67
130. Belujon P, Grace AA. 2011. Hippocampus, amygdala, and stress: interacting systems that affect suscep-
tibility to addiction. Ann. N. Y. Acad. Sci. 1216:114–21
131. Jakobsson J, Cordero MI, Bisaz R, Groner AC, Busskamp V, et al. 2008. KAP1-mediated epigenetic
repression in the forebrain modulates behavioral vulnerability to stress. Neuron 60:818–31
132. Baudry A, Mouillet-Richard S, Schneider B, Launay JM, Kellermann O. 2010. miR-16 targets the
serotonin transporter: a new facet for adaptive responses to antidepressants. Science 329:1537–41
133. Libert S, Pointer K, Bell EL, Das A, Cohen DE, et al. 2011. SIRT1 activates MAO-A in the brain to
mediate anxiety and exploratory drive. Cell 147:1459–72
134. Gao J, Wang WY, Mao YW, Graff J, Guan JS, et al. 2010. A novel pathway regulates memory and
plasticity via SIRT1 and miR-134. Nature 466:1105–9
135. Kaufman J, Charney D. 2001. Effects of early stress on brain structure and function: implications for
understanding the relationship between child maltreatment and depression. Dev. Psychopathol. 13:451–71
136. Lupien SJ, McEwen BS, Gunnar MR, Heim C. 2009. Effects of stress throughout the lifespan on the
brain, behaviour and cognition. Nat. Rev. Neurosci. 10:434–45
137. Kaufman J, Zigler E. 1987. Do abused children become abusive parents? Am. J. Orthopsychiatry 57:186–92
138. Moffitt TE, Caspi A, Harrington H, Milne BJ, Melchior M, et al. 2007. Generalized anxiety disorder
and depression: childhood risk factors in a birth cohort followed to age 32. Psychol. Med. 37:441–52
139. Neigh GN, Gillespie CF, Nemeroff CB. 2009. The neurobiological toll of child abuse and neglect.
Trauma Violence Abuse 10:389–410
140. Essex MJ, Thomas Boyce W, Hertzman C, Lam LL, Armstrong JM, et al. 2011. Epigenetic vestiges of
early developmental adversity: childhood stress exposure and DNA methylation in adolescence. Child
Dev. In press, doi: 10.1111/j.1467-8624.2011.01641.x
141. Bale TL, Baram TZ, Brown AS, Goldstein JM, Insel TR, et al. 2010. Early life programming and
neurodevelopmental disorders. Biol. Psychiatry 68:314–19
142. Meaney MJ. 2001. Maternal care, gene expression, and the transmission of individual differences in stress
reactivity across generations. Annu. Rev. Neurosci. 24:1161–92
143. Millstein RA, Holmes A. 2007. Effects of repeated maternal separation on anxiety- and depression-related
phenotypes in different mouse strains. Neurosci. Biobehav. Rev. 31:3–17
144. Murgatroyd C, Patchev AV, Wu Y, Micale V, Bockmuhl Y, et al. 2009. Dynamic DNA methylation
programs persistent adverse effects of early-life stress. Nat. Neurosci. 12:1559–66
145. Lolait SJ, Stewart LQ, Jessop DS, Young WS III, O’Carroll AM. 2007. The hypothalamic-pituitary-
adrenal axis response to stress in mice lacking functional vasopressin V1b receptors. Endocrinology
148:849–56
146. Roth TL, Lubin FD, Funk AJ, Sweatt JD. 2009. Lasting epigenetic influence of early-life adversity on
the BDNF gene. Biol. Psychiatry 65:760–69

84 Vialou et al.
PA53CH04-Nestler ARI 5 December 2012 12:48

147. Roth TL, Sweatt JD. 2011. Epigenetic marking of the BDNF gene by early-life adverse experiences.
Horm. Behav. 59:315–20
148. Edwards JR, O’Donnell AH, Rollins RA, Peckham HE, Lee C, et al. 2010. Chromatin and sequence
features that define the fine and gross structure of genomic methylation patterns. Genome Res. 20:972–80
149. Levine A, Worrell TR, Zimnisky R, Schmauss C. 2012. Early life stress triggers sustained changes
in histone deacetylase expression and histone H4 modifications that alter responsiveness to adolescent
antidepressant treatment. Neurobiol. Dis. 45:488–98
150. Zhang TY, Meaney MJ. 2010. Epigenetics and the environmental regulation of the genome and its
function. Annu. Rev. Psychol. 61:439–66
151. Weaver IC, Cervoni N, Champagne FA, D’Alessio AC, Sharma S, et al. 2004. Epigenetic programming
by maternal behavior. Nat. Neurosci. 7:847–54
152. Weaver IC, D’Alessio AC, Brown SE, Hellstrom IC, Dymov S, et al. 2007. The transcription factor
nerve growth factor-inducible protein A mediates epigenetic programming: altering epigenetic marks
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

by immediate-early genes. J. Neurosci. 27:1756–68


by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

153. Conway-Campbell BL, George CL, Pooley JR, Knight DM, Norman MR, et al. 2011. The HSP90
molecular chaperone cycle regulates cyclical transcriptional dynamics of the glucocorticoid receptor and
its coregulatory molecules CBP/p300 during ultradian ligand treatment. Mol. Endocrinol. 25:944–54
154. Zeng L, Zhang Q, Gerona-Navarro G, Moshkina N, Zhou MM. 2008. Structural basis of site-specific
histone recognition by the bromodomains of human coactivators PCAF and CBP/p300. Structure 16:643–
52
155. Weaver IC, Champagne FA, Brown SE, Dymov S, Sharma S, et al. 2005. Reversal of maternal program-
ming of stress responses in adult offspring through methyl supplementation: altering epigenetic marking
later in life. J. Neurosci. 25:11045–54
156. Bagot RC, van Hasselt FN, Champagne DL, Meaney MJ, Krugers HJ, Joëls M. 2009. Maternal care
determines rapid effects of stress mediators on synaptic plasticity in adult rat hippocampal dentate gyrus.
Neurobiol. Learn. Mem. 92:292–300
157. Champagne FA. 2008. Epigenetic mechanisms and the transgenerational effects of maternal care. Front.
Neuroendocrinol. 29:386–97
158. McGowan PO, Sasaki A, D’Alessio AC, Dymov S, Labonte B, et al. 2009. Epigenetic regulation of the
glucocorticoid receptor in human brain associates with childhood abuse. Nat. Neurosci. 12:342–48
159. Champagne FA, Meaney MJ. 2007. Transgenerational effects of social environment on variations in
maternal care and behavioral response to novelty. Behav. Neurosci. 121:1353–63
160. McGowan PO, Suderman M, Sasaki A, Huang TC, Hallett M, et al. 2011. Broad epigenetic signature
of maternal care in the brain of adult rats. PLoS ONE 6:e14739
161. Weaver IC, Meaney MJ, Szyf M. 2006. Maternal care effects on the hippocampal transcriptome and
anxiety-mediated behaviors in the offspring that are reversible in adulthood. Proc. Natl. Acad. Sci. USA
103:3480–85
162. McGowan PO, Sasaki A, Huang TC, Unterberger A, Suderman M, et al. 2008. Promoter-wide hyper-
methylation of the ribosomal RNA gene promoter in the suicide brain. PLoS ONE 3:e2085
163. Maccari S, Morley-Fletcher S. 2007. Effects of prenatal restraint stress on the hypothalamus-pituitary-
adrenal axis and related behavioural and neurobiological alterations. Psychoneuroendocrinology 32(Suppl.
1):S10–15
164. Weinstock M. 2008. The long-term behavioural consequences of prenatal stress. Neurosci. Biobehav. Rev.
32:1073–86
165. Weinstock M. 2010. Intrauterine factors as determinants of depressive disorder. Isr. J. Psychiatry Relat.
Sci. 47:36–45
166. Oberlander TF, Weinberg J, Papsdorf M, Grunau R, Misri S, Devlin AM. 2008. Prenatal exposure to
maternal depression, neonatal methylation of human glucocorticoid receptor gene (NR3C1) and infant
cortisol stress responses. Epigenetics 3:97–106
167. Yehuda R, Bierer LM. 2008. Transgenerational transmission of cortisol and PTSD risk. Prog. Brain Res.
167:121–35

www.annualreviews.org • Epigenetic Mechanisms of Depression 85


PA53CH04-Nestler ARI 5 December 2012 12:48

168. Yehuda R, Teicher MH, Seckl JR, Grossman RA, Morris A, Bierer LM. 2007. Parental posttraumatic
stress disorder as a vulnerability factor for low cortisol trait in offspring of Holocaust survivors. Arch.
Gen. Psychiatry 64:1040–48
169. Jaenisch R, Bird A. 2003. Epigenetic regulation of gene expression: how the genome integrates intrinsic
and environmental signals. Nat. Genet. 33(Suppl.):245–54
170. Feng J, Chang H, Li E, Fan G. 2005. Dynamic expression of de novo DNA methyltransferases Dnmt3a
and Dnmt3b in the central nervous system. J. Neurosci. Res 79:734–46
171. Katz LC, Shatz CJ. 1996. Synaptic activity and the construction of cortical circuits. Science 274:1133–38
172. Youngson NA, Whitelaw E. 2008. Transgenerational epigenetic effects. Annu. Rev. Genomics Hum. Genet.
9:233–57
173. Franklin TB, Russig H, Weiss IC, Graff J, Linder N, et al. 2010. Epigenetic transmission of the impact
of early stress across generations. Biol. Psychiatry 68:408–15
174. Mueller BR, Bale TL. 2008. Sex-specific programming of offspring emotionality after stress early in
pregnancy. J. Neurosci. 28:9055–65
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

175. Mueller BR, Bale TL. 2007. Early prenatal stress impact on coping strategies and learning performance
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

is sex dependent. Physiol. Behav. 91:55–65


176. Morgan CP, Bale TL. 2011. Early prenatal stress epigenetically programs dysmasculinization in second-
generation offspring via the paternal lineage. J. Neurosci. 31:11748–55
177. Dietz DM, LaPlant Q, Watts EL, Hodes GE, Russo SJ, et al. 2011. Paternal transmission of stress-
induced pathologies. Biol. Psychiatry 70:408–14
178. Curley JP, Mashoodh R, Champagne FA. 2011. Epigenetics and the origins of paternal effects. Horm.
Behav. 59:306–14
179. He F, Lidow IA, Lidow MS. 2006. Consequences of paternal cocaine exposure in mice. Neurotoxicol.
Teratol. 28:198–209
180. Dunn GA, Bale TL. 2009. Maternal high-fat diet promotes body length increases and insulin insensitivity
in second-generation mice. Endocrinology 150:4999–5009
181. Dunn GA, Bale TL. 2011. Maternal high-fat diet effects on third-generation female body size via the
paternal lineage. Endocrinology 152:2228–36
182. Anway MD, Cupp AS, Uzumcu M, Skinner MK. 2005. Epigenetic transgenerational actions of endocrine
disruptors and male fertility. Science 308:1466–69
183. Skinner MK, Anway MD, Savenkova MI, Gore AC, Crews D. 2008. Transgenerational epigenetic
programming of the brain transcriptome and anxiety behavior. PLoS ONE 3:e3745
184. Stouder C, Paoloni-Giacobino A. 2010. Transgenerational effects of the endocrine disruptor vinclozolin
on the methylation pattern of imprinted genes in the mouse sperm. Reproduction 139:373–79
185. Brykczynska U, Hisano M, Erkek S, Ramos L, Oakeley EJ, et al. 2010. Repressive and active histone
methylation mark distinct promoters in human and mouse spermatozoa. Nat. Struct. Mol. Biol. 17:679–87
186. Daxinger L, Whitelaw E. 2010. Transgenerational epigenetic inheritance: more questions than answers.
Genome Res. 20:1623–28
187. Franklin TB, Mansuy IM. 2010. Epigenetic inheritance in mammals: evidence for the impact of adverse
environmental effects. Neurobiol. Dis. 39:61–65
188. Hammoud SS, Nix DA, Zhang H, Purwar J, Carrell DT, Cairns BR. 2009. Distinctive chromatin in
human sperm packages genes for embryo development. Nature 460:473–78
189. Cuzin F, Rassoulzadegan M. 2010. Non-Mendelian epigenetic heredity: gametic RNAs as epigenetic
regulators and transgenerational signals. Essays Biochem. 48:101–6
190. Rassoulzadegan M, Cuzin F. 2010. The making of an organ: RNA mediated developmental controls in
mice. Organogenesis 6:33–36
191. Feng S, Jacobsen SE, Reik W. 2010. Epigenetic reprogramming in plant and animal development. Science
330:622–27
192. Delaval K, Feil R. 2004. Epigenetic regulation of mammalian genomic imprinting. Curr. Opin. Genet.
Dev. 14:188–95
193. Reik W, Walter J. 2001. Genomic imprinting: parental influence on the genome. Nat. Rev. Genet. 2:21–32
194. Davies W, Isles AR, Humby T, Wilkinson LS. 2007. What are imprinted genes doing in the brain?
Epigenetics 2:201–6

86 Vialou et al.
PA53CH04-Nestler ARI 5 December 2012 12:48

195. Isles AR, Davies W, Wilkinson LS. 2006. Genomic imprinting and the social brain. Philos. Trans. R. Soc.
Lond. B 361:2229–37
196. Smith FM, Garfield AS, Ward A. 2006. Regulation of growth and metabolism by imprinted genes.
Cytogenet. Genome Res. 113:279–91
197. Gimelbrant A, Hutchinson JN, Thompson BR, Chess A. 2007. Widespread monoallelic expression on
human autosomes. Science 318:1136–40
198. Schalkwyk LC, Meaburn EL, Smith R, Dempster EL, Jeffries AR, et al. 2010. Allelic skewing of DNA
methylation is widespread across the genome. Am. J. Hum. Genet. 86:196–212
199. Meaburn EL, Schalkwyk LC, Mill J. 2010. Allele-specific methylation in the human genome: implica-
tions for genetic studies of complex disease. Epigenetics 5:578–82
200. Gregg C, Zhang J, Butler JE, Haig D, Dulac C. 2010. Sex-specific parent-of-origin allelic expression in
the mouse brain. Science 329:682–85
201. Gregg C, Zhang J, Weissbourd B, Luo S, Schroth GP, et al. 2010. High-resolution analysis of parent-
of-origin allelic expression in the mouse brain. Science 329:643–48
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

202. Borgel J, Guibert S, Li Y, Chiba H, Schubeler D, et al. 2010. Targets and dynamics of promoter DNA
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

methylation during early mouse development. Nat. Genet. 42:1093–100


203. Cheung I, Shulha HP, Jiang Y, Matevossian A, Wang J, et al. 2010. Developmental regulation and
individual differences of neuronal H3K4me3 epigenomes in the prefrontal cortex. Proc. Natl. Acad. Sci.
USA 107:8824–29
204. Ernst J, Kheradpour P, Mikkelsen TS, Shoresh N, Ward LD, et al. 2011. Mapping and analysis of
chromatin state dynamics in nine human cell types. Nature 473:43–49
205. Muotri AR, Zhao C, Marchetto MC, Gage FH. 2009. Environmental influence on L1 retrotransposons
in the adult hippocampus. Hippocampus 19:1002–7
206. Muotri AR, Marchetto MC, Coufal NG, Oefner R, Yeo G, et al. 2010. L1 retrotransposition in neurons
is modulated by MeCP2. Nature 468:443–46
207. Kuwabara T, Hsieh J, Muotri A, Yeo G, Warashina M, et al. 2009. Wnt-mediated activation of NeuroD1
and retro-elements during adult neurogenesis. Nat. Neurosci. 12:1097–105
208. Maze I, Feng J, Wilkinson MB, Sun H, Shen L, Nestler EJ. 2011. Cocaine dynamically regulates
heterochromatin and repetitive element unsilencing in nucleus accumbens. Proc. Natl. Acad. Sci. USA
108:3035–40
209. Singer T, McConnell MJ, Marchetto MC, Coufal NG, Gage FH. 2010. LINE-1 retrotransposons:
mediators of somatic variation in neuronal genomes? Trends Neurosci. 33:345–54
210. Pastor WA, Pape UJ, Huang Y, Henderson HR, Lister R, et al. 2011. Genome-wide mapping of 5-
hydroxymethylcytosine in embryonic stem cells. Nature 473:394–97
211. Covington HI, Berton O. 2010. Chromatin-based treatments for affective disorders—insight or utopia.
In Depression: From Psychopathology to Pharmacotherapy, ed. JF Cryan, BE Leonard, pp. 182–98. Basel,
Switz.: Karger

www.annualreviews.org • Epigenetic Mechanisms of Depression 87


PA53-FrontMatter ARI 14 December 2012 9:33

Annual Review of
Pharmacology and
Toxicology

Contents Volume 53, 2013

A Conversation with Paul Greengard


Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

Paul Greengard and Eric J. Nestler p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 1


by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

Pharmacology of Iron Transport


Shaina L. Byrne, Divya Krishnamurthy, and Marianne Wessling-Resnick p p p p p p p p p p p p p p17
Impact of Soluble Epoxide Hydrolase and Epoxyeicosanoids
on Human Health
Christophe Morisseau and Bruce D. Hammock p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p37
Epigenetic Mechanisms of Depression and Antidepressant Action
Vincent Vialou, Jian Feng, Alfred J. Robison, and Eric J. Nestler p p p p p p p p p p p p p p p p p p p p p p p p59
The PI3K, Metabolic, and Autophagy Networks: Interactive Partners
in Cellular Health and Disease
Naval P. Shanware, Kevin Bray, and Robert T. Abraham p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p89
Small Molecule–Based Approaches to Adult Stem Cell Therapies
Luke L. Lairson, Costas A. Lyssiotis, Shoutian Zhu, and Peter G. Schultz p p p p p p p p p p p p p 107
G Protein–Coupled Receptor Deorphanizations
Olivier Civelli, Rainer K. Reinscheid, Yan Zhang, Zhiwei Wang,
Robert Fredriksson, and Helgi B. Schiöth p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 127
Pluripotent Stem Cell–Derived Hepatocytes: Potential and Challenges
in Pharmacology
Dagmara Szkolnicka, Wenli Zhou, Balta Lucendo-Villarin, and David C. Hay p p p p p p 147
Tyrosine Kinase Inhibitors: Views of Selectivity, Sensitivity,
and Clinical Performance
Alexander Levitzki p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 161
Creating Order from Chaos: Cellular Regulation by Kinase Anchoring
John D. Scott, Carmen W. Dessauer, and Kjetil Taskén p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 187
Unnatural Amino Acids as Probes of Ligand-Receptor Interactions
and Their Conformational Consequences
Stephan A. Pless and Christopher A. Ahern p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 211

v
PA53-FrontMatter ARI 14 December 2012 9:33

Cyclic Nucleotide Compartmentalization: Contributions of


Phosphodiesterases and ATP-Binding Cassette Transporters
Satish Cheepala, Jean-Sebastien Hulot, Jessica A. Morgan, Yassine Sassi,
Weiqiang Zhang, Anjaparavanda P. Naren, and John D. Schuetz p p p p p p p p p p p p p p p p p p p 231
One Hundred Years of Drug Regulation: Where Do We Go
from Here?
Raymond L. Woosley p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 255
Autophagy in Toxicology: Cause or Consequence?
Sten Orrenius, Vitaliy O. Kaminskyy, and Boris Zhivotovsky p p p p p p p p p p p p p p p p p p p p p p p p p p p 275
Insights from Genome-Wide Association Studies of Drug Response
Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org

Kaixin Zhou and Ewan R. Pearson p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 299


by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

The Potential of HDAC Inhibitors as Cognitive Enhancers


Johannes Gräff and Li-Huei Tsai p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 311
Molecular Mechanisms Deployed by Virally Encoded
G Protein–Coupled Receptors in Human Diseases
Silvia Montaner, Irina Kufareva, Ruben Abagyan, and J. Silvio Gutkind p p p p p p p p p p p p p 331
Genetic Risk Prediction: Individualized Variability in Susceptibility
to Toxicants
Daniel W. Nebert, Ge Zhang, and Elliot S. Vesell p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 355
microRNAs as Mediators of Drug Toxicity
Tsuyoshi Yokoi and Miki Nakajima p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 377
Role of Nrf2 in Oxidative Stress and Toxicity
Qiang Ma p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 401
Direct-Acting Antiviral Agents for Hepatitis C Virus Infection
Jennifer J. Kiser and Charles Flexner p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 427
Systems Pharmacology to Predict Drug Toxicity: Integration Across
Levels of Biological Organization
Jane P.F. Bai and Darrell R. Abernethy p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 451
Omics and Drug Response
Urs A. Meyer, Ulrich M. Zanger, and Matthias Schwab p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 475
Renal Transporters in Drug Development
Kari M. Morrissey, Sophie L. Stocker, Matthias B. Wittwer, Lu Xu,
and Kathleen M. Giacomini p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 503
Structure-Function of the G Protein–Coupled Receptor Superfamily
Vsevolod Katritch, Vadim Cherezov, and Raymond C. Stevens p p p p p p p p p p p p p p p p p p p p p p p p p p 531

vi Contents
PA53-FrontMatter ARI 14 December 2012 9:33

Design of Peptide and Peptidomimetic Ligands with Novel


Pharmacological Activity Profiles
Victor J. Hruby and Minying Cai p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 557
Hepatic and Intestinal Drug Transporters: Prediction of
Pharmacokinetic Effects Caused by Drug-Drug Interactions
and Genetic Polymorphisms
Kenta Yoshida, Kazuya Maeda, and Yuichi Sugiyama p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 581

Indexes

Contributing Authors, Volumes 49–53 p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 613


Annu. Rev. Pharmacol. Toxicol. 2013.53:59-87. Downloaded from www.annualreviews.org
by NORTH CAROLINA STATE UNIVERSITY on 01/16/13. For personal use only.

Article Titles, Volumes 49–53 p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p p 616

Errata

An online log of corrections to Annual Review of Pharmacology and Toxicology articles


may be found at http://pharmtox.annualreviews.org/errata.shtml

Contents vii

You might also like