Zhang Meaney 10 AnnRPsychol Epigenetics Review

Embed Size (px)

Citation preview

  • 8/11/2019 Zhang Meaney 10 AnnRPsychol Epigenetics Review

    1/34

    Epigenetics andthe EnvironmentalRegulation of the Genomeand Its Function

    Tie-Yuan Zhang and Michael J. Meaney

    Sackler Program for Epigenetics and Psychobiology of McGill University, Douglas Ment

    Health University Institute and the Singapore Institute for Clinical Sciences, Montreal,Quebec, H4H 1R3 Canada; email: [email protected]

    Annu. Rev. Psychol. 2010. 61:43966

    TheAnnual Review of Psychologyis online atpsych.annualreviews.org

    This articles doi:10.1146/annurev.psych.60.110707.163625

    Copyright c2010 by Annual Reviews.All rights reserved

    0066-4308/10/0110-0439$20.00

    Key Words

    maternal care, stress responses, DNA methylation, gene x

    environment interactions, glucocorticoid receptor

    Abstract

    There are numerous examples in psychology and other disciplines o

    the enduring effects of early experience on neural function. In this article, we review the emerging evidence for epigenetics as a candidate

    mechanism for these effects. Epigenetics refers to functionally relevanmodifications to the genome that do not involve a change in nucleotid

    sequence. Such modifications include chemical marks that regulate th

    transcription of the genome. There is now evidence that environmental events can directly modify the epigenetic state of the genome. Thu

    studies with rodent models suggest that during both early develop

    ment and in adult life, environmental signals can activate intracellulapathways that directly remodel the epigenome, leading to changes ingene expression and neural function. These studies define a biologica

    basis for the interplay between environmental signals and the genomin the regulation of individual differences in behavior, cognition, an

    physiology.

    439

  • 8/11/2019 Zhang Meaney 10 AnnRPsychol Epigenetics Review

    2/34

    Phenotype: any

    observablecharacteristic or traitof an organism, such asits morphology,development,biochemical orphysiologicalproperties, or behavior

    Contents

    INTRODUCTION .. . . . . . . . . . . . . . . . . 440GENE TRANSCRIPTION .. . . . . . . . . 441

    Chromatin Modifications. . . . . . . . . . . 442Regulation of Glucocorticoid

    Receptor Expression . . . . . . . . . . . . 443

    ENVIRONMENTALPROGRAMMING OF

    GENE EXPRESSION..... . . . . . . . . 444EPIGENETIC REGULATION

    OF THE GENOME . . . . . . . . . . . . . . 446Epigenetics and the

    Social Environment . . . . . . . . . . . . . 449THE FUNCTIONAL

    IMPORTANCE OF THESOCIAL IMPRINT . . . . . . . . . . . . . . . 451

    ACTIVITY-DEPENDENT

    REGULATION OFTHE EPIGENOME . . . . . . . . . . . . . . 452Summary (and Perhaps

    Some Constraints) . . . . . . . . . . . . . . 455

    EPIGENETICS ANDMENTAL HEALTH . . . . . . . . . . . . . . 456

    CONCLUSIONS . . . . . . . . . . . . . . . . . . . . 458

    INTRODUCTION

    Psychology has seen a major transition in

    personality theory. Personality traits were oncethought to emerge under the dominion of

    influences associated with nurture. The post-natal family environment was considered as the

    primary candidate force in the developmentof individual differences in personality. This

    perspective changed dramatically in responseto integration of the biological sciences into

    personality psychology. First, evolutionary ap-proaches established the idea that the brain and

    its development, like any other organ, are sub-

    ject to evolutionary forces. Second, behavioralgenetics (Ebstein 2006, Kendler 2001, Plomin

    & Rutter 1998) provided evidence for a relationbetween variation at the level of the genome

    and that in personality and mental health.Although efforts to quantify the independent

    contribution of genetic and environmental

    influences are fraught with complications (e

    geneenvironment interactions, nongenommechanisms of inheritance), measures of co

    cordance in specific traitsbetween monozygo

    and dizygotic twins, among other approachsuggest a pervasive influence of genetic var

    tion. Indeed, it is impossible to imagine thatfunction of brain cells could occur independ

    of variations in the genes that encode proteins that regulate neuronal functions.

    Genomic variation at the level of nucleotsequence is associated with individual diff

    ences in personality and thus with vulnerabity and resistance to a wide range of chro

    illness (Ebstein 2006, Meyer-LindenbergWeinberger 2006, Rutter 2007). Such var

    tions can take multiple forms, including vaation at the level of (a) a single nucleotide (i

    single-nucleotide polymorphisms or SNP

    (b) variation in the number of nucleotide peats (i.e., variable number of tandem repeat

    VNTRs), or (c) chromosomal reorganizatiEach form of variation can potentially alter

    nomic function and thus phenotype. The chlenge for psychology is that of conceptually

    tegrating findings from genetics into the stuof personality and our understanding of t

    pathophysiology of mental illness. Howand uder what conditions does genomic variation

    fluence brain development and function? Hmight relevant findings from the field of g

    netics influence the design of public policy a

    therapies in psychology?It is important to note the simple f

    that genes encode for protein, not functiThus, as described below, the effects of gene

    variation are contextually determined and bconsidered as probabilistic. Cellular functi

    can only be understood in terms of the constdialogue that occurs between the genome a

    its environment. The environment regulathe cellular signals that control the operat

    of the genome. The objective of this reviis to describe recent advances in molecu

    biology, notably in the field of epigenetics, a

    to suggest that epigenetic mechanisms areideal candidate mechanism for the effects

    environmental signals, including events su

    440 Zhang Meaney

  • 8/11/2019 Zhang Meaney 10 AnnRPsychol Epigenetics Review

    3/34

    as social interactions, on the structure and

    function of the genome (Harper 2005). Theintent is to first consider the processes by which

    cellular signals, referred to as transcription

    factors, regulate the activity (or expression)of a gene. The biological primacy of gene

    environment interactions is apparent from thesimple realization that the levels and the activ-

    ity of these transcription factors is controlledby environmental signals. Thus the operation

    of the genome is dependent upon context. Thequestion concerns the mechanisms responsible

    for such contextual influences. We suggestthat epigenetics is one such process and can

    account, in part, for instances in which envi-ronmental events occurring at any time over

    the lifespan exert a sustained effect on genomic

    function and phenotype.Epigenetics signals refer to a series of chem-

    ical modifications to the DNA or to regionssurrounding the DNA. The transcriptional

    activity of the genome is regulated by signals,transcription factors, that physically bind to

    specific DNA sites. The importance of epige-netics mechanisms lies in the ability to regulate

    the ease with which transcription factors canaccess the DNA. Epigenetic signals can thus

    determine the capacity for environmentalregulation of the genome. There is emerging

    evidence for the idea that epigenetic marks are

    directly altered in early life by environmentalevents and thus influence the development

    of individual differences in specific neuralfunctions that underlie cognition and emotion.

    More recent studies suggest that dynamicalterations in these same epigenetic signals are

    crucial for the synaptic remodeling that medi-ates learning and memory. Thus, epigenetics

    provides a remarkable insight into the biologythat governs the function of the genome in

    response to environmental signals.

    GENE TRANSCRIPTION

    The most compelling evidence for the pre-

    dominance of geneenvironment interactionsin cellular function emerges from the study of

    gene transcription [Gilbert (2006) provides a

    Gene transcriptionthe synthesis of RNAunder the direction DNA. RNA synthesis the process oftranscribing DNA

    nucleotide sequenceinformation into RNsequence informatio

    Receptor: a proteinembedded in eitherthe plasma membranor cytoplasm of a ceto which a mobilesignaling molecule(ligand) may bind. Tsignaling molecule cbe a peptide, ahormone, a

    pharmaceutical drugor a toxin, and whensuch binding occursthe receptor goes inta conformationalchange that ordinariinitiates a cellularresponse

    Chromatin: thecombination of DNARNA, and protein thmakes upchromosomes. The

    major components ochromatin are DNAand histone proteins

    very clear and well-illustrated description]. The

    transcription of the genome is a highly regu-lated event. At the heart of this process lies a

    class of proteins referred to as transcription fac-

    tors. As the name implies, these proteins havethe ability to bind to regulatory elements of the

    gene and to activate or repress gene transcrip-tion. Importantly, the expression and activation

    of the transcription factors themselves are dy-namically regulated by environmental signals.

    Many of the earliest cellular responses to envi-ronmental stimuli involve either the activation

    of pre-existing transcriptional signals throughchemical modifications such as phosphoryla-

    tion (i.e., the addition of a phosphate) of spe-cific amino acids of the protein, or an increase

    in gene expression that results in the rapid syn-

    thesis of proteins (e.g., immediate early geneproducts) that then serve to regulate the ac-

    tivity of other genes. This includes genes thatare involved in synaptic plasticity. The bind-

    ing of transcription factors to DNA sites is thebiological machinery for the dynamic gene

    environment interactions that result in alteredrates of gene transcription.

    Figure 1 portrays the organization of theglucocorticoid receptor gene as an example of

    genomic organization and a target for discus-sion below. The schema is actually somewhat

    misleading. For reasons of graphic simplicity,

    we often describe the organization of a geneor the interactions between transcription

    factors and DNA as if the DNA were a linearmolecule to which transcription factors gain

    unimpeded access. The reality of proteinDNA interactions is very different. Figure 2

    presents the classic crystallographic analysis ofthe organization of DNA (Luger et al. 1997).

    DNA is organized into units referred to asnucleosomes, each of which contains about

    145150 base pairs wrapped around a coreregion of histone proteins (Turner 2001). The

    histones and DNA together are referred to as

    chromatin; the nucleosome is the organizationof chromatin. Under normal conditions there

    is a tight physical relation between the histoneproteins and its accompanying DNA, resulting

    in a rather closed nucleosome configuration.

    www.annualreviews.org Epigenetics and the Environment 441

  • 8/11/2019 Zhang Meaney 10 AnnRPsychol Epigenetics Review

    4/34

    ..tggg16cggggg17cgggag.

    11 14 15 1617 18 19 110 111 2

    NGFI-A

    GR Promoter 17 Sequence

    9

    Regulatory (non-coding) region Coding region

    Figure 1

    A schema describing the organization of the rat glucocorticoid receptor gene including 9 exon regions.Exons 29 participate in the coding for the glucocorticoid receptor protein. Exon 1 is composed of multipregulatory regions, each of which is capable of activating gene transcription (i.e., promoter sequences). Tactivity of the various exon 1 promoters is tissue-specific, with evidence suggesting that certain promotersare more active in areas such as liver or thymus, and others are more active in brain (e.g., exon 1 7; based oMcCormick et al. 2000; see Turner & Muller 2005 for comparable data in humans). The use of multiplepromoters permits regulation in one tissue independently from other regions (i.e., increased glucocorticoreceptor in pulmonary tissues prior to birth that is necessary for respiratory competency at parturition, whmaintaining reduced glucocorticoid receptor levels in brain, where glucocorticoid effects might inhibitneurogenesis). The consensus binding site for nerve growth factorinducible factor A (NGFI-A) lying witthe exon 17promoter is highlighted. The reader should note that this organization is not necessarily typicRegulatory elements (promoters or enhancers) can exist between exons (i.e., within introns) or at sites thaare either 5 or 3 to the coding region, sometimes at considerable distances.

    Enzyme: a molecule,usually protein, thatcatalyzes (i.e., increasesthe rate of ) a specificchemical reaction

    Histone deacetylases(HDACs): a class ofenzymes that removeacetyl groups from an-N-acetyl lysineamino acid on a

    histone. The action ofHDACs is thus theopposite to that ofhistoneacetyltransferases, andHDACs are associatedwith transcriptionalsilencing

    This restrictive configuration is maintained,

    in part, by electrostatic bonds between thepositively charged histones and the negatively

    charged DNA. The closed configurationimpedes transcription factor binding and

    is associated with a reduced level of geneexpression. An increase in transcription factor

    binding to DNA and the subsequent activationof gene expression commonly requires chem-

    ical modification of the chromatin that occurson thehistone proteins. The primary targets for

    such events are the amino acids that form thehistone tails extending from the nucleosome

    (Figure 2). These modifications alter chro-matin in a manner that either increases or

    decreases the ability of transcription factorsto access regulatory sites on the DNA thatcontrol gene transcription.

    Chromatin Modifications

    The dynamic alteration of chromatin structure

    is achieved through modifications to thehistone

    proteins at the amino acids that form the

    stone protein tails that extend out from tnucleosome (Figure 2). These modificatio

    are achieved through a series of enzymes tbind to the histone tails and modify the

    cal chemical properties of specific amino ac(Grunstein 1997, Hake & Allis 2006, Jenuw

    & Allis 2001). For example, the enzyme histoacetyltransferase transfers an acetyl group on

    specific lysines on the histone tails. The adtion of the acetyl group diminishes the posit

    charge, loosening the relation between the stones and DNA, opening the chromatin a

    improving the ability of transcription factorsaccess DNA sites. Thus, histone acetylation

    specific lysine sites is commonly associatedwactive gene transcription.

    The functional antagonists of the histo

    acetyltransferases are a class of enzymes knoas histone deacetylases (HDACs). These e

    zymes remove acetyl groups and prevent fther acetylation, thus maintaining a clo

    442 Zhang Meaney

  • 8/11/2019 Zhang Meaney 10 AnnRPsychol Epigenetics Review

    5/34

    chromatin structure, decreasing transcription

    factor and gene expression. Both the acety-lation and deacetylation of histones are dy-

    namic processes that are regulated by environ-

    mental signals. Indeed, a number of proteinsthat were known to be associated with tran-

    scriptional activation (e.g., transcriptional co-factors) have been identified as histone acetyl-

    transferases. These factors enhance the efficacyof transcription factors by opening chromatin

    and thus increasing the binding of the factor tothe regulatory regions of the gene.

    The reader should note that there are ac-tually multiple modifications to histone tails,

    including methylation (in this case on the hi-stones), phosphorylation, and ubiquitination.

    For the sake of simplicity, the discussion is lim-

    ited to histone acetylation/deacetylation.

    Regulation of GlucocorticoidReceptor Expression

    The neurotransmitter serotonin (5-

    hydroxytryptamine; 5-HT) regulates glu-cocorticoid receptor gene transcription in

    hippocampal neurons (Figure 3; Mitchell et al.1990, 1992; Weaver et al. 2007). This effect

    is dependent upon the binding of the tran-

    scription factor nerve growth factorinduciblefactor A (NGFI-A) to a specific binding site

    on the exon 17 glucocorticoid (GR) promoter(Figure 1). The importance of this interaction

    can be precisely defined. For example, mutatinga single nucleotide, in this case simply exchang-

    ing a cytosine for an adenine, in the regionof the promoter that normally binds NGFI-A

    abolishes the ability of NGFI-A to associatewith the exon 17 promoter and eliminates

    the effect of NGFI-A on gene transcription(Weaver et al. 2007). However, the ability of

    NGFI-A to bind to the exon 17 promoter is

    regulated by another protein, a transcriptionalcofactor, the CREB-binding protein, that is

    activated by the same 5-HT-cyclic adenosinemonophosphate (cAMP)/cyclic nucleotide

    dependent kinases (PKA)-signaling cascadethat results in the increased levels of NGFI-A

    (Figure 3). The CREB-binding protein is a

    Histoneacetyltransferases:enzymes that acetylalysine amino acids ohistone proteins bytransferring an acety

    group from acetyl Cto form -N-acetyllysine. Histoneacetylation isassociated with theactivation of genetranscription

    Promoter: a regionDNA that facilitatesthe transcription of particular gene.Promoters aretypically located nea

    the genes theyregulate, on the samstrand and upstreamfrom the coding regi

    histone acetyltransferase. The association of

    the CREB-binding protein with the exon 17promoter is accompanied by an increase in

    the acetylation of a specific lysine on the tail

    of histone 3 of the exon 17 promoter (Weaveret al. 2004, 2007). Thus, 5-HT activates both

    NGFI-A and the CREB-binding protein.Interestingly, NGFI-A and the CREBbinding

    protein physically associate with one anotherprior to DNA binding. The CREBbinding

    protein acetylates histones associated withthe exon 17 promoter, enhancing the abil-

    ity of NGFI-A to bind and activate genetranscription.

    Environmental signals alter 5-HT activity.Indeed, the effect of 5-HT on glucocorticoid

    receptor expression reflects the dependency

    of gene transcription on signals that derivefrom environmental events (note that the

    relevant environmental event may be internalor external to the organism; e.g., a change

    in the availability of glucose, an electricalimpulse, or a social interaction). Such effects

    underlie the dynamic interdependence of geneand environment. However, psychologists, and

    in particular developmental psychologists, arefamiliar with more enduring environmental

    influences; instances where experience inearly life has shaped neural development and

    function in a manner that is sustained into

    adulthood. Such effects are considered as thebasis for environmental influences over the de-

    velopment of individual differences. In certaincases, the sustained effects of early experience

    have been associated with structural alterationsin neural circuits that mediate specific func-

    tions. The process of sexual differentiationamong vertebrates provides excellent examples

    where environmental signals lead to differencesin morphology and thus to gender. However,

    more recent studies suggest another form ofenvironmentally regulated plasticity that exists

    at the level of genome itself. Such effects ap-

    pear to involve the modification of epigeneticmarks on the DNA. These studies suggest

    that environmental events alter the activity ofspecific intracellular signals that modify the

    nature of the epigenetic marks at specific sites

    www.annualreviews.org Epigenetics and the Environment 443

  • 8/11/2019 Zhang Meaney 10 AnnRPsychol Epigenetics Review

    6/34

    in the genome, leading to sustained effects on

    gene expression and thus neural function.

    ENVIRONMENTALPROGRAMMING OFGENE EXPRESSION

    Studies in developmental psychobiology and

    physiology are replete with examples of theenvironmental programming of gene expres-

    sion. Such studies commonly report that a vari-ation in the early environment associates with

    changes in gene expression and biological func-tion that persists into adulthood and thus well

    beyond the duration of the relevant environ-mental event. In the rat, for example, prena-

    tal nutrient deprivation or enhanced exposure

    to hormonal signals associated with stress sta-bly alter, or program, the activity of genes in

    the liver and other sites that are associatedwith glucose and fat metabolism, including the

    gene for the glucocorticoid receptor (Batesonet al. 2004; Gluckman & Hanson 2004, 2007;

    Jirtle & Skinner 2007; Meaney et al. 2007;Seckl & Holmes 2007). These findings are as-

    sumed to represent instances in which the op-eration of a genomic region in adulthood varies

    as a function of early environmental influences.The results of recent studies suggest that such

    programming effects can derive from gene

    environment interactions in early life that leadto a structural alteration of the DNA, which

    in turn mediates the effects on gene expressionas well as more complex levels of phenotype

    ( Jirtle & Skinner 2007, Meaney 2007, Meaney& Szyf 2005). These studies were performed

    in rodents but were inspired by the vast lit-erature reporting the pervasive effects of fam-

    ily environment on health outcomes in humans(Repetti et al. 2002). No less compelling are the

    results of studies on maternaleffects in plants,insects, reptiles, and birds showing that varia-

    tions in nongenomic signals of maternal ori-

    gin associate with sustained effects on the phe-notype of the offspring (Cameron et al. 2005,

    Mousseau & Fox 1998, Rossiter 1998).The objective of these studies is to examine

    the biological mechanisms whereby variations

    in motherinfant interactions might directly

    fluence gene expression and behavior (Mean2001). Such studies focus on variations

    maternal behavior that lie within the normrange for the species, in this case the Norw

    rat, and that occur in the absence of a

    experimental manipulations (i.e., naturally curring variations in motherpup interaction

    Variations on maternal care in the rat are stuied with simple, albeit very time-consumi

    observations on animals in their home ca(Champagne 2008, Champagne et al. 200

    One behavior, pup licking/grooming (Lemerges as highly variable across mothers. P

    LG is a major source of tactile stimulatifor the neonatal rat that regulates endocr

    and cardiovascular function in the pup (Ho2005, Levine 1994, Schanberg et al. 1984). T

    question then was whether such variations

    pup LG might directly alter the developmof individual differences in behavior a

    physiology. For the studies reviewed hethe focus is on the development of individ

    differences in defensive responses.Subsequent findings revealed considera

    evidence for the effect of maternal care the behavioral and endocrine responses

    stress in the offspring. The male or femadult offspring of mothers that natur

    exhibit increased levels of pup LG (i.e., toffspring of high-LG mothers) show m

    modest behavioral and endocrine responses

    stress compared to animals reared by low-Lmothers (Caldji et al. 1998, Francis et al. 19

    Liu et al. 1997, Menard et al. 2004, Toki et2007, Weaver et al. 2004). Specifically,

    offspring of high-LG mothers show reducfearfulness and more modest hypothalam

    pituitary-adrenal (HPA) responses to strCross-fostering studies, where pups born

    high-LG mothers are fostered at birth low-LG mothers (and vice versa), revea

    direct relationship between maternal cand the postnatal development of individ

    differences in behavioral and HPA respon

    to stress (Caldji et al. 2000, 2003; Francis et1999; Weaver et al. 2004). In these studies,

    rearing mother determined the phenotype

    444 Zhang Meaney

  • 8/11/2019 Zhang Meaney 10 AnnRPsychol Epigenetics Review

    7/34

    Hypothalamus

    Pituitary

    Adrenals

    Hippocampus Hippocampus

    Glucocorticoids

    () ()

    ()

    ()

    ()

    ()

    ()

    CRF

    ACTH

    ()

    Hypothalamus

    Pituitary

    AdrenalsGlucocorticoids

    ()

    CRF

    ACTH

    ()

    Figure 4

    A schema outlining the function of the hypothalamic-pituitary-adrenal axis, the nexus of which are the

    corticotropin-releasing factor (CRF) neurons of the paraventricular nucleus of the hypothalamus. CRF isreleased into the portal system of the anterior pituitary, stimulating the synthesis and release ofadrenocorticotropin (ACTH), which then stimulates adrenal glucocorticoid release. Glucocorticoids act onglucocorticoid receptors in multiple brain regions, including the hippocampus, to inhibit the synthesis andrelease of CRF (i.e., glucocorticoid negative feedback). The adult offspring of high-LG mothers, bycomparison to those of low-LG dams, show (a) increased glucocorticoid receptor expression, (b) enhancednegative-feedback sensitivity to glucocorticoids, (c) reduced CRF expression in the hypothalamus, and(d) more modest pituitary-adrenal responses to stress.

    the offspring. Thus variations within a normal

    range of parental care can dramatically alterphenotypic development in the rat.

    The effects of maternal care on the devel-opment of defensive responses to stress in the

    rat involve alterations in the function of thecorticotrophin-releasing factor (CRF) systems

    in selected brain regions (Figure 4). The CRF

    system furnishes the critical signal for the acti-vation of behavioral, emotional, autonomic, and

    endocrine responses to stressors (Bale & Vale2004, Koob et al. 1994, Plotsky et al. 1989). As

    adults, the offspring of high-LG mothers showdecreased CRF expression in the hypothalamus

    as well as reduced plasma ACTH and gluco-

    corticoid responses to acute stress by compar-ison to the adult offspring of low-LG mothers(Francis et al. 1999; Liu et al. 1997; Weaveret al.

    2004, 2005). Circulating glucocorticoids act atglucocorticoid receptor sites in corticolimbic

    structures, such as thehippocampus, to regulate

    HPA activity (Figure 4). Such feedback effects

    commonly inhibit hypothalamic CRF expres-

    sion. The high-LG offspring showed signif-icantly increased hippocampal glucocorticoid

    receptor expression, enhanced glucocorticoidnegative feedback sensitivity, and decreased hy-

    pothalamic CRF levels. Indeed, the magnitudeof the glucocorticoid response to acute stress

    is significantly correlated with the frequency of

    pup LG during the first week of life, as is thelevel of both hippocampal glucocorticoid re-

    ceptor andhypothalamicCRF expression (all rs>0.70; Liu et al. 1997). Importantly, pharmaco-

    logicalmanipulationsthatblocktheeffectoftheglucocorticoid receptor eliminate the maternal

    effect on the HPA response to stress, suggesting

    that the differences in hippocampal glucocorti-coid receptor expression are directly related tothose at the level of HPA function.

    Pup LG is a major source of tactile stimula-tion for the neonate. Experimental models that

    directly apply tactile stimulation, through the

    stroking of the pup with a brush, provide direct

    www.annualreviews.org Epigenetics and the Environment 445

  • 8/11/2019 Zhang Meaney 10 AnnRPsychol Epigenetics Review

    8/34

    evidence for the importance of tactile stimu-

    lation derived from pup LG. Thus, strokingpups over the first week of life increases hip-

    pocampal glucocorticoid receptor expression

    ( Jutapakdeegul et al. 2003) and dampens be-havioral and HPA responses to stress (Bur-

    ton et al. 2007, Gonzalez et al. 2001). Like-wise, manipulations of lactating mothers that

    directly increase the frequency of pup LGalso increase hippocampal glucocorticoid re-

    ceptor expression and decrease HPA responsesto stress (Francis et al. 1999, Toki et al. 2007).

    Manipulations, notably stressors imposed onthemother, that decrease pupLG areassociated

    with increased behavioral and HPA responsesto stress and are associated with decreased hip-

    pocampal glucocorticoid receptor expression

    and increased hypothalamic expression of CRF(Champagne & Meaney 2006, Fenoglio et al.

    2005).The offspring of the high-LG and low-LG

    mothers also differ in behavioral responses tonovelty (Caldji et al. 1998, Francis et al. 1999,

    Zhang et al. 2004). As adults, the offspringof the high-LG mothers show decreased star-

    tle responses, increased open-field exploration,and shorter latencies to eat food provided in

    a novel environment. There are also behav-ioral differences in response to more precise

    forms of threat. Thus, the offspring of low-

    LG mothers show greater burying of an elec-trified probe in the defensive burying paradigm

    (Menard et al. 2004), which involves an activeresponse to a threat. These differences in be-

    havioral responses to stress are associated withaltered activity in the CRF system that links

    the amygdala (and bed nucleus of the stria ter-minalis) to the noradrenergic cells of the locus

    coeruleus (Caldji et al. 1998, Zhang et al. 2004).The results of these studies suggest that the

    behavior of the mother toward her offspringcan program stable changes in gene expres-

    sion that then serve as the basis for individual

    differences in behavioral and neuroendocrineresponses to stress in adulthood. The maternal

    effects on phenotype are associated with sus-tained changes in the expression of genes in

    brain regions that mediate responses to stress

    and form the basis for stable individual diff

    ences in stress reactivity. These findings pvide a potential mechanism for the influen

    of parental care on vulnerability/resistancestress-induced illness over the lifespan. But

    critical issue is simply that of how mater

    care might stably affect gene expression. Hare variations in the social interactions betw

    the mother and her offspring biologically ebedded so as to stably alter the activity of s

    cific regions of the genome? The answersthese questions appear to involve the abil

    of social interactions in early developmentstructurally modify relevant genomic regio

    For the sake of this review, we focus on the mternal effect on the regulation of hippocam

    glucocorticoid receptor expression.

    EPIGENETIC REGULATIONOF THE GENOME

    The molecular processes that lead to the inittion of gene transcription involve modificatio

    to the histone proteins that form the core ofnucleosome (Figure 2). Such modificati

    open chromatin, permitting transcriptfactor binding and the activation of g

    transcription. A second level of regulat

    occurs not on the histone proteins, but rathdirectly on the DNA. Indeed, the classic e

    genetic alteration is that of DNA methylatiwhich involves the addition of a methyl gro

    (CH3) onto cytosines in the DNA (Bird 19Holliday 1989, Razin & Riggs 1980). DN

    methylation is associated with the silencof gene transcription. This effect appears

    be mediated in one of two ways (Bird 200First, wide swaths of densely methylated DN

    preclude transcription factor binding to DNsites, thus silencing gene expression. The s

    ond manner is subtler and probably far mo

    prevalent in regions with more dynamic vaations in gene transcription, such as the bra

    In this case, selected cytosines are methylatand the presence of the methyl group attra

    a class of proteins know as methylated-DNbinding proteins (Klose & Bird 2007). Th

    proteins, in turn, attract an entire cluster

    446 Zhang Meaney

  • 8/11/2019 Zhang Meaney 10 AnnRPsychol Epigenetics Review

    9/34

    proteins that form a repressor complex, which

    includes active mediators of gene silencing.The HDACs are a critical component of the

    repressor complex. HDACs prevent histone

    acetylation and favor a closed chromatin statethat constrains transcription factor binding

    and gene expression (Figure 2and see above).Compounds that inhibit HDACs can thus

    increase transcription from methylated DNA.When we think of genomic influences, we

    most commonly imagine effects associated withvariation in nucleotide sequence. Yet this is only

    one form of information contained within thegenome. Despite the reverence afforded DNA,

    a gene is basically like any other molecule inthe cell; it is subject to physical modifications.

    As described above, these modifications alter

    the structure and chemical properties of theDNA and thus gene expression. Collectively,

    themodifications to the DNA andits chromatinenvironment can be considered as an additional

    layer of information that is contained within thegenome. This information is thus epigenetic in

    nature (the name derives from the Greek epimeaning upon and genetics). The acetylation

    of histone proteins or the methylation of DNAare examples of epigenetic modifications. Epi-

    genetic modifications do not alter the sequencecomposition of the genome. Instead, these epi-

    genetic marks on the DNA and the histone pro-

    teins of the chromatin regulate the operation ofthe genome. Thus, epigenetics has been de-

    fined as a functional modification to the DNAthat does not involve an alteration of sequence

    (Waddington 1957). Although this definitionhas been subjected to revision (Bird 2007, Hake

    & Allis 2006), the essential features of epige-netic mechanisms are (a) structural modifica-

    tions to chromatin either at the level of the hi-stone proteins (Figure 2) or the DNA, (b) the

    associated regulation of the structure and func-tion of chromatin, (c) the downstream effects

    on gene expression, and (d) the fact that these

    effects occur in the absence of any change innucleotide sequence.

    The methylation of DNA in mammals is anactive biochemical modification that selectively

    targets cytosines and is achieved through the

    actions of enzymes, DNA methyltransferases,

    that transfer the methyl groups from methyldonors. There are two critical features to DNA

    methylation: First, it is a stable chemical modi-fication, and second, it is associated with the si-

    lencing of gene transcription (Bestor 1998, Bird

    2002, Bird & Wolffe 1999, Razin 1998).Until very recently, it wasthought that DNA

    methylation patterns on the genome were over-laid upon the genome only during early peri-

    ods in embryonic development. Indeed, DNAmethylation is considered as a fundamental

    feature of cell differentiation. It is importantto consider a simple feature of cell biology:

    All cells in the body generally share the sameDNA. Thus, the processes of cell specializa-

    tion, whereby liver cells specialize in functionsrelated to energy metabolism and brain cells

    establish the capacity for learning and memory,

    involve silencing certain regions of the genomein a manner that is specific for each cell type.

    Genes associated with gluconeogenesis are si-lenced in brain cells but remain active in liver

    cells. Such processes define the function of thecell type (e.g., Fan et al. 2005). DNA methyla-

    tion is considered as a mechanism for the ge-nomic silencing that underlies cell specializa-

    tion. Such events occur early in developmentand are considered to be highly stable,such that

    dedifferentiation (whereby a cell loses its spe-cialization) is rare and often is associated with

    organ dysfunction.Thus DNA methylation was considered

    both unique to early periods in develop-

    ment and irreversible. Experimental modelscommonly used to study DNA methyla-

    tion further reinforced this view. DNAmethylation-induced gene silencing mediates

    two of the most commonly studied examplesof the epigenetic silencing of genes, namely

    X-chromosome inactivation and gene im-printing. Mammalian females bear two copies

    of the X-chromosome. The inactivation ofone copy of the X-chromosome occurs in

    all mammalian females and is essential fornormal function (i.e., maintaining a constant

    gene dosage in males and females). The

    silencing of the X-chromosome is associated

    www.annualreviews.org Epigenetics and the Environment 447

  • 8/11/2019 Zhang Meaney 10 AnnRPsychol Epigenetics Review

    10/34

    with DNA methylation (Mohandas et al. 1981,

    Riggs & Pfieffer 1992; but also see Hellman& Chess 2007 for a more current update).

    The second example of epigenetic-mediated

    gene silencing is that of gene imprinting (daRocha & Ferguson-Smith 2004, Reik 2001),

    a remarkable subject in its own right and onewith considerable implications for growth and

    development (Charalambous et al. 2007). Forhumans and other mammals, the expression of

    specific genes is determined by the parent oforigin. For certaingenes, the copy derived from

    the mother is active while that emanating fromthe father is silenceda maternally imprinted

    gene. In other cases, it is the reverse: The copyof the gene inherited from the father is active

    while that from the mother is silenceda

    paternally imprinted gene. The silent copyis methylated in DNA regions that regulate

    gene expression and thus is inactive. Again,the epigenetic marks associated with gene

    imprinting are established very early in life.These marks, as well as those associated with

    X-chromosome inactivation, are largely stable.Collectively, these models have left biolo-

    gists with the impression that under normalconditions, DNA methylation occurs early in

    embryonic life and is irreversible. DNA methy-lation was considered to be an actively dy-

    namic process only during periods of cell di-

    vision and differentiation (see above) such thatin mature postmitotic cells, further alteration

    of methylation patterns was improbable. More-over, the extensive loss of cytosine methylation

    in the models described above is associated withpathology. This perspective was further rein-

    forced by findings showing that an alterationof DNA methylation at critical genomic tar-

    gets (i.e., tumor suppressors) is associated withcancer (Eden et al. 2003, Feinberg 2007, Laird

    2005).At this point, dynamic changes in DNA

    methylation were of considerable interest for

    developmental biologists but somewhat less sofor psychologists, who study the aftermath of

    more subtle variations in neuronal differentia-tion that occur in later periods of development

    or even in the fully mature brain. The issue for

    developmental psychologists concerns less

    process by which cells specialize as neuroand more the issues related to why neurons

    one individual function differently from thof another, or how neurons might dynamica

    later alter functional properties in relation

    experience (i.e., activity-dependent neuroplasticity). The studies reviewed below prov

    an importantrevision to this perspective. This now considerable evidence in neuroscien

    and other fields, including immunology aendocrinology/metabolism, that the state

    DNA methylation at specific genomic siteindeed dynamic even in adult animals (B

    2007, Jirtle & Skinner 2007, Meaney & S2005). Moreover, alterations in DNA methy

    tion are emerging as a candidate mechanismthe effects of early experience in individual d

    ferences in neural function as well as in learn

    and memory. Thus, although the assumptioconcerning DNA methylation appear valid

    the examples cited above, recent studies revthat DNA methylation patterns are activ

    modified in mature (i.e., fully differentiatcells including, and perhaps especially, n

    rons, and that such modifications can occuranimals in response to cellular signals driv

    by environmental events ( Jirtle & Skin2007, Meaney & Szyf 2005, Sweatt 2009). F

    example, variations in the diet of mice durgestation or later in development, such as

    early postweaning period, can stably alter methylation status of the DNA (Cooney et

    2002, Waterland & Jirtle 2003, Waterla

    et al. 2006, Whitelaw & Whitelaw 200Likewise, both mature lymphocytes (Bruniq

    & Schwartz 2003, Murayama et al. 2006) aneurons (e.g., Champagne 2008, Champag

    et al. 2006, Lubin et al. 2008, Martinowet al. 2003, Sweatt 2009) show changes in t

    DNA methylation patterns at critical genomregions in response to environmental stim

    that stably alter cellular function. The abiof environmental signals to actively remo

    epigenetic marks that regulate gene expressis a rather radical change in our understand

    of the environmental regulationof gene expr

    sion. Such epigenetic modifications are th

    448 Zhang Meaney

  • 8/11/2019 Zhang Meaney 10 AnnRPsychol Epigenetics Review

    11/34

    a candidate mechanism for the environmental

    programming of gene expression.

    Epigenetics and theSocial Environment

    The section below describes studies of themolecular basis for the effects of maternal care

    on the development of individual differencesin gene expression and stress responses. The

    mechanism for this interaction is epigenetic, in-volving alterations in DNA methylation at spe-

    cific sites in the genome. In summary, variationsin motherinfant interactions in theratalter the

    extra- and intracellular environment of neuronsin selected brain regions. Such alterations di-

    rectly modify the epigenetic marks on regions

    of the DNA that regulate the transcription ofthe glucocorticoid receptor, which in turn reg-

    ulates the HPA response to stress. These epige-neticmarksarestable,enduringwellbeyondthe

    period of maternal care, and provide a molec-ular basis for a stable maternal effect on the

    phenotype of the offspring. Thus, the behaviorof the mother directly alters cellular signals that

    then actively sculpt the epigenetic landscape ofthe offspring, influencing the activity of specific

    regions of the genome and the phenotype of theoffspring.

    The critical feature of the maternal effects

    described above is that of persistence. The dif-ferences in the frequency of pup LG between

    high- and low-LG mothers are limited to thefirst week of postnatal life. And yet the differ-

    encesingeneexpressionandneuralfunctionareapparent well into adulthood. How might the

    effects of an essentially social interaction stablyalter the expression of the genes that regulate

    the activity of neural systems that mediate en-docrine and behavioral responses to stress? To

    address this question, we focused on the sus-tained effect of maternal care on glucocorticoid

    receptor gene transcription in the hippocampus

    as a model system for the environmental pro-gramming of gene expression.

    The focus of the epigenetic studies isthe NGFI-A consensus sequence in the

    exon 17 promoter (Figure 1) that activates

    glucocorticoid receptor expression in hip-

    pocampal neurons. The tactile stimulationassociated with pup LG increases 5-HT activ-

    ity in the hippocampus. In vitro studies with

    cultured hippocampal neurons show that 5-HTacts on 5-HT7 receptors to initiate a series of

    intracellular signals that culminate with an in-crease in the expression of NGFI-A as well as in

    the CREB-binding protein (Figure 3). Com-parable effects occur in vivo. Manipulations

    that increase pup LG by lactating rats result inan increased level of cAMP as well as NGFI-A

    (Meaney et al. 2000). Pups reared by high-LGmothers show increased NGFI-A expression

    in hippocampal neurons as well as an increasedbinding of NGFI-A to the exon 17promoter se-quence (Weaver et al. 2007, Zhang et al. 2009).

    Moreover, the binding of NGFI-A to the exon17 promoter sequence is actively regulated by

    motherpup interactions, such that there isincreased NGFI-A bound to the exon 17 pro-

    moter immediately following a nursing bout,but not at a period that follows 25 minuteswith-

    out motherpup contact (Zhang et al. 2009).NGFI-A and the CREB-binding protein

    form a complex that binds directly to the exon17 promoter sequence and actively redesigns

    the methylation pattern at this region of the

    genome (Weaver et al. 2004, 2007). Thus, asadults, the offspring reared by high-LG moth-

    ers show very modest levels of methylationat the 5 CpG of the NGFI-A consensus se-

    quence (Figure 5). This effect on methylationis very precise. Located only a few nucleotides

    removed from this site is the 3 CpG site(Figures 1 and 5), the methylation status of

    which is unaffected by maternal care.A rather novel aspect of the effect of ma-

    ternal care on DNA methylation was apparentin the results of a simple developmental study

    examining the methylation status of the 5 and

    3 CpG sites from late in fetal life to adulthood(Weaver et al. 2004). Neither the 5 nor the

    3 CpG sites within the NGFI-A bindingregion is methylated in hippocampal neurons

    from fetal rats, whereas both sites are heavilymethylated on the day following birth, with

    no difference as a function of maternal care.

    www.annualreviews.org Epigenetics and the Environment 449

  • 8/11/2019 Zhang Meaney 10 AnnRPsychol Epigenetics Review

    12/34

    These findings reflect what is referred to as

    de novo methylation, whereby a methyl groupis applied to previously unmethylated sites.

    However, between the day following birth and

    the end of the first week of life, the 5 CpGis demethylated in pups reared by high-LG,

    but not low-LG, mothers. This differencethen persists into adulthood. Importantly, the

    period over which the demethylation occursfalls precisely within that time when high- and

    low-LG mothers differ in the frequency of pupLG; the difference in pup LG between high-

    and low-LG mothers is not apparent in thesecond week of postnatal life (Caldji et al. 1998,

    Champagne 2008, Champagne et al. 2003).The demethylation of the 5 CpG siteoccurs

    as a function of the same 5-HT-activated sig-

    nals that regulate glucocorticoid receptor geneexpression in cultured hippocampal neurons

    (Weaver et al. 2007). Thus, when hippocam-pal neurons of embryonic origin are placed in

    culture and treated with 5-HT, which mimicsthe extracellularsignal associated with maternal

    LG,the5 CpG site is demethylated; there is noeffectatthe3 CpG site. The binding of NGFI-

    A to the exon 17 site is critical. Hippocampalneurons that are rendered incapable of increas-

    ing NGFI-A expression through antisense or

    siRNA treatment show neither the demethyla-tion of the 5 CpG site nor the increase in glu-

    cocorticoid receptor expression (Weaver et al.2007). Likewise, a mutation of the NGFI-A

    site (exchanging a C for an A at the 3 CpGsite) that completely abolishes the binding of

    NGFI-A to the exon 17 promoter also pre-vents the demethylation of the 5 CpG. Finally,

    the infection of hippocampal neurons with avirus containing a nucleotide construct engi-

    neered to express high levels of NGFI-A pro-duces demethylation of the 5 CpG of the exon

    17 promoter sequence and increases glucocor-

    ticoid receptor expression.These findings suggest that maternal lick-

    ing of pups increases NGFI-A levels in thehippocampal neurons of the offspring, thus al-

    tering DNA methylation. But there is a compli-cation. If DNA methylation blocks transcrip-

    tion factor binding and the 5 CpG site of

    the exon 17 promoter is heavily methylated

    neonates, then how might maternally activaNGFI-A bind to and remodel the exon 17

    gion? And why is the effect apparent at the

    but not the 3 CpG? The answer to these qutions appears to involve other transcriptio

    signals that are affected by maternal care. Lels of the transcription factorspecific protein

    (SP-1) and the CREB-binding protein are aincreased in the hippocampus of pups reared

    high-LG mothers (Weaver et al. 2007, Zhaet al. 2009). The exon 17 promoter contain

    DNA sequence that binds SP-1, and this gion overlaps with that for NGFI-A. SP-1 c

    actively target both methylation and demethlation of CpG sites (Brandeis et al. 1994). T

    5CpG site is the region of overlap in the bin

    ing sites. The CREB-binding protein, on tother hand, acts as a histone acetyltransfera

    an enzyme capable of acetylating histone taincluding the exon 17 region, opening ch

    matin and permitting the binding of transcrtion factors such as NGFI-A and SP-1. Incre

    ing histone acetylation can lead to transcriptfactor binding at previously methylated si

    and the subsequent demethylation of these gions (Fan et al. 2005, Szyf et al. 2005). Th

    we suggest that the binding of this complex

    proteins, NGFI-A, the CREB-binding proteand SP-1 is critical in activating the process

    demethylation. The results to date are certaiconsistent with this model, but we should n

    that we have yet to firmly establish the idetity of the enzyme that is responsible for t

    demethylation of the 5 CpG site.These findings suggest that materna

    induced increases in hippocampal NGFlevels can initiate the remodeling of DN

    methylation at the regions of the DNA thregulate glucocorticoid receptor expressi

    The NGFI-A transcription factor binds

    multiple sites across the genome. If NGFI-related complexes target demethylation, th

    one might assume that other NGFI-A-sensitregions should show a maternal effect on DN

    methylation and gene expression comparato that observed with the glucocorticoid rec

    tor. Zhang and colleagues (2009) showed t

    450 Zhang Meaney

  • 8/11/2019 Zhang Meaney 10 AnnRPsychol Epigenetics Review

    13/34

    the hippocampal expression of theGAD1gene

    that encodes for glutamic acid decarboxylase,an enzyme in the production of the neuro-

    transmitter GABA, is increased in the adult

    offspring of high-LG mothers. This effect isassociated with altered DNA methylation of

    an NGFI-A response element in a mannercomparable to that for the glucocorticoid

    receptor gene. Moreover, as with the effecton the glucocorticoid receptor, an in vitro

    increase in NGFI-A expression mimics theeffects of increased pup LG. The function of

    GABAergic neurons in the limbic system is alsoregulated by maternal care (Caldji et al. 1998,

    2000, 2003) and is a major target for anxiolytic

    agents. These findings are therefore likelyrelevant for the decreased fearfulness observed

    in the adult offspring of high-LG mothers.In summary, the maternallyinduced changes

    in specific intracellular signals in hippocampalneurons can physically remodel the genome.

    The increased binding of NGFI-A that derivesfrom pup LG appears critical for the demethy-

    lation of the exon 17promoter. We suggest thatthis process involves accompanying increases in

    SP-1 and the CREB-binding protein, and thatthe combination of these factors results in the

    active demethylation of the exon 17 promoter.

    It should be noted that there are important fea-tures of this model that remain to be clearly de-

    fined, includingthe identificationof theenzymethat is directly responsible for the demethyla-

    tion. Nevertheless, the events described to daterepresent a model by which the biological path-

    ways activated by a social event may become im-printed onto the genome. This imprint is then

    physically apparent in the adult genome, result-ing in stable alterations (or programming) of

    gene expression.

    THE FUNCTIONALIMPORTANCE OF THESOCIAL IMPRINT

    A critical issue is that of relating the epige-

    netic modifications at specific DNA regionsto function. The presence of a methyl group

    on the 5 CpG of the NGFI-A binding site is

    functionally related to glucocorticoid receptor

    gene expression in adult animals. In vitro stud-ies reveal that the methylation of the 5 CpG

    site reduces the ability of NGFI-A to bind

    to the exon 17 promoter and activate gluco-corticoid receptor transcription (Weaver et al.

    2007). These findings are consistent with themodel described above, whereby DNA methy-

    lation impedes transcription factor binding andthus the activation of gene expression. The

    next question concerns the in vivo situationand function at a level beyond that of gene

    expression.In contrast to the situation with neonates,

    there is no difference in NGFI-A expressionas a function of maternal care among adult

    animals: Hippocampal levels of NGFI-A are

    comparable in the adult offspring of high-and low-LG mothers. However, the altered

    methylation of the exon 17 promoter wouldsuggest differences in the ability of NGFI-A to

    access its binding site on the exon 17promoter.Chromatin-immunoprecipitation assays, which

    permit measurement of the interaction be-tween a specific protein and a defined region of

    the DNA, reveal increased NGFI-A associationwith the exon 17promoter in hippocampi from

    adult offspring of high- compared to low-LG

    mothers (Weaver et al. 2004, 2005). This dif-ference occurs despite the comparable levels of

    NGFI-A. These findings show that in the livinganimal, under normal conditions, there is more

    NGFI-A associated with the exon 17promoterin hippocampal neurons of adult animals reared

    by high- compared with low-LG mothers.There is also evidence that directly links

    the maternal effect on the epigenetic state ofthe exon 17 promoter to the changes in glu-

    cocorticoid receptor expression and HPA re-sponses to stress. Recall that the methylation

    of specific CpG sites can diminish transcrip-

    tion factor binding through the recruitmentof repressor complexes that include HDACs.

    The HDACs deacetylate histone tails, thusfavoring a closed chromatin configuration. In-

    deed, the exon 17 promoter is more promi-nently acetylated in hippocampi from adult off-

    springof high- compared with low-LGmothers

    www.annualreviews.org Epigenetics and the Environment 451

  • 8/11/2019 Zhang Meaney 10 AnnRPsychol Epigenetics Review

    14/34

    (Weaver et al. 2004, 2005). This finding is con-

    sistent with the increased transcription of theglucocorticoid receptor gene in animals reared

    by high- versus low-LG mothers. A subsequent

    study (Weaver et al. 2004) examined the ef-fects of directly blocking the actions of the

    HDACs in the adult offspring of high- and low-LG mothers by directly infusing an HDAC in-

    hibitor into the hippocampus daily for four con-secutive days. The treatment with the HDAC

    inhibitor produces a series of predictable re-sults that reflect a cause-effect relation between

    DNA methylation andgene expression. First, asexpected, HDAC blockade eliminates the dif-

    ferences in the acetylation of the histone tails(open chromatin) of the exon 17 promoter inhippocampal samples from high- and low-LG

    mothers. Second, the increased histone acety-lation of the exon 17promoter in the offspring

    of low-LG mothers is associated with an in-crease in the binding of NGFI- A to the exon

    17promoter in the offspring of low-LG moth-ers, eliminating the maternal effect on NGFI-

    A binding to the exon 17 promoter. Compa-rable levels of NGFI-A binding to the exon

    17 promoter then eliminate the maternal ef-fect on hippocampal glucocorticoid receptor

    expression, such that glucocorticoid receptor

    levels in the adult offspring of low-LG motherstreated with the HDAC inhibitor are compa-

    rable to those in animals reared by high-LGmothers. And most importantly, the infusion of

    the HDAC inhibitor reversed the differences inthe HPA response to stress.

    HDAC inhibition increases NGFI-A bind-ing to the exon 17 promoter in the offspring

    of low-LG mothers. The studies with neonatesreveal that increased NGFI-A binding results

    in the demethylation of the 5 CpG. In vitro,the introduction of a viral tool that leads to the

    increased expression of NGFI-A is sufficient

    to demethylate the exon 17 promoter. Weaveret al. (2007) argue that the binding of NGFI-A

    is critical for the demethylation of the 5CpGsite. The same effect is apparent in vivo and

    even with the adult animals used in the stud-ies described above. HDAC infusion into the

    hippocampus increases NGFI-A binding to the

    exon 17promoter in the adult offspring of lo

    LG mothers and decreases the level of methytionof the 5CpG site onthe exon17promo

    Another study (Weaver et al. 2005) show

    that the reverse pattern of results could be otained in response to the infusion of meth

    nine into the hippocampus. The methioninfusion produced greater methylation of

    5CpG in the offspring of high-LG mothedecreasedNGFI-Abinding andGR expressi

    and increased HPA responses to stress (Weaet al. 2005).

    Although these studies employ rather crupharmacological manipulations, the results

    criticalastheysuggestthatfullymatureneurin an adult animal express the necessary en

    matic machinery to demethylate or remeth

    late DNA. The importance of this plasticat the level of DNA methylation is revea

    in subsequent studies of cognition (see belowhich suggest that dynamic modification

    DNA methylation in critical neuronal poplations in adult animals is involved in spec

    forms of learning and memory.

    ACTIVITY-DEPENDENTREGULATION OF

    THE EPIGENOME

    The maternal effect on the epigenetic state

    the glucocorticoid receptor exon 17 promoand glucocorticoid receptor gene expression

    apparent over the first week of life and occin response to an increased NGFI-A signa

    hippocampal neurons. The increased exprsion of NGFI-A and its binding to the exon

    GR promoter over the first week of life are avated by maternal behavior (Weaver et al. 20

    Zhang et al.2009). An increase in theexpressof NGFI-A is associated with synaptic plast

    ity and with learning and memory (Dragun

    1996, Jones et al. 2001, Knapska & Kaczma2004, Li et al. 2005, ODonovan et al. 199

    Thus it is not surprising that the offspringhigh-LG mothers show increased synaptic d

    sity both in early life (Liu et al. 2000) ain adulthood (Bagot et al. 2009, Bredy et

    2003, Champagne et al. 2008, Liu et al. 200

    452 Zhang Meaney

  • 8/11/2019 Zhang Meaney 10 AnnRPsychol Epigenetics Review

    15/34

    Such events occur as a function of a series of

    activity-dependent changes in neuronal activ-ity triggered by the action of glutamate at the

    NMDA receptor site (Ali & Salter 2001, Bear &

    Malenka 1994, Malenka & Nicoll 1993, Morris& Frey 1997). Thus, it is possible that envi-

    ronmentally driven changes in neuronal tran-scriptional signals could potentially remodel

    the methylation state of specific regions of theDNA (Meaney & Szyf 2005, Sng & Meaney

    2009). These effects could, in turn, prove es-sential for sustained alterations in synaptic

    function.Learning and long-term memory com-

    monly require changes in gene expression andprotein synthesis (Alberini et al. 1995, Kandel

    2001, Lynch 2004). As described above, gene

    transcription is associated with chromatinremodeling engineered by enzymes that

    modify the histone proteins within chromatincomplexes. A number of the intracellular

    signals that are crucial for learning and mem-ory are in fact enzymes that modify histone

    proteins. One example is that of the CREB-binding protein, which functions as a histone

    acetyltransferase and is strongly implicated incognitive function (e.g., Alarcon et al. 2004).

    Thus, contextual fear conditioning, which isa hippocampus-dependent learning paradigm

    whereby an animal associates a novel context

    with an aversive stimulus, is accompanied byincreased acetylation of histone H3 (Levenson

    et al. 2004). Likewise, there is evidence for theimportance of epigenetic modifications of his-

    tones in the amygdala during fear conditioning(Yeh et al. 2004). Interestingly, extinction of

    the conditioned fear response is associatedwith increased histone acetylation in the pre-

    frontal cortex, which mediates the inhibitionof conditioned fear responses (Bredy et al.

    2007). The CREB-binding protein is probablyinvolved in the relevant histone modifications.

    Mice that are heterozygous for a dysfunction

    form of the CREB-binding protein showsignificant impairments in multiple forms of

    hippocampal-dependent, long-term memory(Bourtchouladze et al. 2003, Korzus et al. 2004,

    Wood et al. 2006; also see Guan et al. 2002,

    Vecsey et al. 2007). Importantly, the cognitive

    impairments are reversed with HDAC ad-ministration, suggesting that CREB-binding

    protein-induced histone acetylation mediateseffects on learning and memory.

    There is also evidence for the importance of

    dynamic changes in DNA methylation at spe-cific sites during learning and memory. Fear

    conditioning results in the rapid methylationand transcriptional silencing of the gene for

    protein phosphatase 1 (PP1), which suppresseslearning. The same training results in the

    demethylation and transcriptional activation ofthe synaptic plasticity gene reelin. These find-

    ings imply that both DNA methylation anddemethylation might be involved in long-term

    memory consolidation.BDNF has been implicated in adult neu-

    ral plasticity, including learning and memory

    (West 2001). Thegenomic structure of the Bdnfgene contains multiple promoters that gener-

    ate mRNAs containing different noncoding ex-ons spliced upstream of a common coding exon

    (Timmusk et al. 1993). This organization issomewhat like that described above for the glu-

    cocorticoid receptor (Figure 1). In the case ofBDNF, the exon IV promoter in rat is activated

    upon membrane depolarization in cultured cor-tical and hippocampal neurons by means of

    KCl, which leads to calcium influx, activatingsignaling cascades and inducing the expression

    of an array of genes that are involved in neuralplasticity (West 2001).

    Importantly, the activity-dependent Bdnf

    gene is also regulated through epigenetic modi-fications that involve dynamic changes in DNA

    methylation and the association of methylated-DNA binding proteins to the relevant sites

    on the bdnf promoter. Thus, increased DNAmethylation of the exon IV promoter at sites

    that bind to transcriptional activators is associ-atedwiththepresenceofthemethylated-DNA-

    binding protein, MeCP2, and a decreased levelof bdnf expression. This transcriptionally

    quiescent state prior to depolarization is alsoassociated with the presence of histone deacety-

    lases (i.e., HDAC1) and mSIN3A, which form

    a common repressor complex. Membrane

    www.annualreviews.org Epigenetics and the Environment 453

  • 8/11/2019 Zhang Meaney 10 AnnRPsychol Epigenetics Review

    16/34

    Proteinphosphorylation:a modification ofproteins in which aspecific amino acid(serine, a threonine or

    a tyrosine) isphosphorylated by aprotein kinase by theaddition of a covalentlybound phosphategroup. In such cases ofphosphoregulation,the protein switchesbetween aphosphorylated and anunphosphorylatedstate, with one in anactive form and the

    other inactive

    depolarization of the neuron leads to a de-

    crease in CpG methylation and a dissociationof MeCP2-related repressor complex from the

    exon IV promoter. As described above for the

    glucocorticoid receptor, the decrease in CpGmethylation is then associated with an increase

    in histone acetylation and the binding of thetranscription factor, CREB. CREB is known

    to activatebdnfexpression. These data suggestthat DNA methylation at a particular site can

    suppress activity-dependent transcription of

    Bdnf. These findings also indicate that DNA

    methylation patterns in postmitotic neuronscan undergo dynamic changes in response to

    neuronal activation, and a lower level of DNAmethylation correlates with a higher level of

    Bdnfgene transcription in neurons.

    Interestingly, MeCP2 levels increase as neu-rons mature (Zoghbi 2003). The high level of

    MeCP2 protein in mature neurons is consis-tent with a possible role for MeCP2 in synaptic

    remodeling associated with learning and mem-ory (Zhou et al. 2006). Further supporting a

    roleforMeCP2inmaturesynapticfunctionandplasticity, Mecp2-null mice exhibit abnormali-

    ties in dendritic arborization (Chen et al. 2003,Kishi & Macklis 2004), basal synaptic transmis-

    sion (Moretti et al. 2006), presynaptic function(Asaka et al. 2006, Moretti et al. 2006, Nelson

    et al. 2006), excitatory synaptic plasticity (Asaka

    et al. 2006, Moretti et al. 2006), and hippocam-pal and amygdalar learning (Moretti et al. 2006,

    Pelka et al. 2006). Zhou et al. (2006) found thatneuronal activity (membrane depolarization) is

    associated with a phosphorylation of MeCP2 atSerine421 that led to its dissociation from the

    bdnfexon IV promoter and an increase in bdnfexpression (also see Chen et al. 2003). Impor-

    tantly, activity-dependent increases in BDNFlevels are blocked in cells bearing a mutant

    version of MeCP2 that is unable to undergophosphorylation. Glutamate is a primary neural

    signalforsynapticplasticity,andbothglutamate

    as well as the direct activation of its NMDAreceptor produced MeCP2 phosphorylation in

    neurons. Glutamate activates NMDA recep-tors, resulting in a neuronal calcium influx and

    the activation of calcium-modulated kinase II

    (CaMKII), which regulates synaptic plastic

    (Lisman et al. 2002). Zhou et al. (2006) fouthat CaMKII actively phosphorylates MeCP

    The protein phosphorylation occurringMeCP2 in response to neuronal activation

    a transient event. The results described abo

    (Martinowich et al. 2003) suggest that nronal activation can lead to changes in DN

    methylation, which is a potentially more stabepigenetic alteration that could conceiva

    result in a long-term change inbdnfexpressiThus far, this review has considered

    relation between DNA methylation, histoacetylation/deacetylation, transcription fac

    binding, and gene expression. However, this evidence that the chromatin alteratio

    can alter DNA methylation. Thus, HDinhibitors result in an increase in histo

    acetylation, enhanced transcription fac

    binding, and decreased DNA methylatiSuch effects were described above in relation

    DNA methylation and glucocorticoid recepexpression (Weaver et al. 2004, 2005). Thus

    is possible that (a) neuronal activation leadsthe transient phosphorylation of MeCP2 a

    its dissociation from the exon IVbdnfpromoand (b) an increase in histone acetylation a

    CREB binding, producing increased bexpression; and that (c) the histone acetylat

    and CREB binding are also associated wDNA demethylation, as described above

    the case of the glucocorticoid receptor histone acetylation and NGFI-A bindi

    Such events could underlie a common proc

    of activity-dependent modification of DNmethylation (Meaney & Szyf 2005).

    Studies by Sweatt and colleagues suggthat the changes in DNA methylation at t

    exon IVbdnfpromoter are involved in specforms of learning and memory (Sweatt 200

    Bdnf gene expression increases in the hpocampus with contextual and spatial learn

    and appears essential for the synaptic remoeling that accompanies such forms of learni

    and memory (Hall et al. 2000, Linnarsson et1997). NMDA receptor activation is criti

    for both contextual (Maren & Quirk 20

    and spatial (Morris et al. 2003) learning

    454 Zhang Meaney

  • 8/11/2019 Zhang Meaney 10 AnnRPsychol Epigenetics Review

    17/34

    well as for the increase in bdnf expression that

    accompanies such events. Lubin et al. (2008)found that contextual fear conditioning was

    associated with a demethylation of the exon IV

    bdnfpromoter and an increase in bdnfexpres-sion: Both effects were blocked with a gluta-

    mate receptor antagonist.Taken together, thesefindings suggest that the activity-dependent

    changes in neuronal activity that associatewith learning and memory induce a dynamic

    alteration in DNA methylation that, in turn,subserves the sustained changes in gene expres-

    sion critical for long-term memory. Althoughthis remains a working hypothesis, the findings

    discussed above further emphasize the degree

    to which neuronal activation can structurallyremodel the genome and alters its operation.

    Interestingly, there is also evidence that en-vironmental influences prevailing during early

    development may determine the capacity forsuch activity-driven, epigenetic modifications.

    Disruptions to motherinfant interactions dur-ing early development are associated with alter-

    ations in hippocampalbdnfexpression (Branchiet al. 2006; Lippman et al. 2007; Roceri et al.

    2002, 2004; but also see Griesen et al. 2005)and increased DNA methylation at the exon IV

    bdnfpromoter (Roth et al. 2009). Rearing micein a communal nest, with three mothers and

    their litters, increases maternal care toward the

    offspring, which in turn is associated with in-creased BDNF expression (Branchi et al. 2006).

    And in the rat, the offspring of high-LG moth-ers show decreased MeCP2 association with the

    exon IVbdnfpromoter (Weaver et al. 2007) andincreased bdnfexpression (Liu et al. 2000). Such

    maternal effects might bias in favor of reducedcapacity for epigenetic remodeling at this crit-

    ical site and restrain synaptic plasticity associ-ated with learning and memory.

    Summary (and PerhapsSome Constraints)

    Studies over the past five years have created

    considerable enthusiasm for epigenetic mod-els of the effects of early experience, synaptic

    plasticity, and neural function. The hypothesis

    underlying this approach considers epigenetic

    effectsongeneexpressionasacandidatemecha-nism for the effects of environmental signals on

    the future behavior of the organism. This hy-

    pothesis is particularly attractive for those ex-amining the sustained effects of early experi-

    ence or of chronic, biologically relevant eventsin adulthood (e.g., environmental enrichment,

    chronic stress) on gene expression and neuralfunction. Mature neurons undergo consider-

    able changes in phenotype and are thereforean ideal cell population for epigenetic regu-

    lation. Nevertheless, there are constraints onthe influence of epigenetic marks. For exam-

    ple, the effects of DNA methylation on geneexpression are influenced by the organization

    of the relevant genomic region. DNA methy-

    lation appears to have a reduced effect ongene expression in regions that have a very

    high density of cytosine-guanine paired sites(Weber et al. 2007). Moreover, much of the

    DNA within a cell is packed tightly in het-erochromatin (Fraser & Bickmore 2007) and

    is probably inaccessible to environmentally in-duced chromatin remodeling signals. Thus, the

    infusion of an HDAC inhibitor directly intothe adult hippocampus alters the expression of

    only about 2% of all the genes normally ex-pressed in the rat hippocampus (Weaver et al.

    2006). Were the entire genome subject to dy-

    namic epigenetic regulation such as describedabove for thebdnfgene, then we could expect

    this percentage to be substantially higher. Itis likely that there is a pool of genes that re-

    tains the capacity for dynamic environmentalregulation through epigenetic mechanisms. Of

    course this begs questions concerning the fac-tors that determine the nature and contents of

    suchpools.These considerations notwithstand-ing, it appears that with neurons, a number of

    genes are closely related to synaptic plasticityand neural function and are subject to dynamic

    regulation through epigenetic mechanisms, in-

    cluding DNA methylation.Epigenetics refers to a collection of chemi-

    cal modifications that occur to histones or di-rectly on the DNA. These modifications, in

    turn, alter gene transcription. One might argue

    www.annualreviews.org Epigenetics and the Environment 455

  • 8/11/2019 Zhang Meaney 10 AnnRPsychol Epigenetics Review

    18/34

    that in defining such mechanisms we have, in

    effect, simply better defined the processes thatregulate gene transcription. Although the value

    of such findings is obvious for molecular biol-

    ogy, how might such processes revise our think-ing at the level of the systems sciences? We

    suggest that these findings provide researcherswith a renewed appreciation of the environ-

    mental regulation of cellular activity. We nowunderstand the physical basis for the Hebbian

    synapse (Hebb 1958), whereby environmen-tal signals activate intracellular pathways that

    result in the remodeling of synaptic connec-tions in a manner that influences subsequent

    activity at relevant sites. There is a physicalreference for the process of neuroplasticity.

    Epigenetic modifications provide the mecha-

    nism for a comparable level of plasticity at thelevelof the genome. We once thought of synap-

    tic connections as being fixed, immutable tofurther changes beyond some critical period in

    development. Studies of synaptic plasticity re-vised our appreciation of the brain, revealing

    instead a dynamic tissue, subject to constant re-modeling through the environmental activation

    of activity-dependent synaptic plasticity. Thestudy of epigenetics suggests a comparable pro-

    cess at the level of the genome, also once con-sidered a constant, static source of influence.

    Indeed, we must emphasize that epigenetic

    modifications do not alter DNA sequence. Theproduct of the glucocorticoid receptor gene is

    unaffected by epigenetic marks. However, it ap-pears that the operation of thegenomeis indeed

    subject to environmental regulation in a man-ner that may be no less dynamic than that of

    synaptic connections.Recent studies from Nestler and colleagues

    reveal considerable epigenetic modification atspecific genomic sites associated with chronic

    stress or repeated exposure to psychostimulantdrugs, both of which produce sustained influ-

    ences on behavior (Nestler 2009, Renthal et al.

    2009). Although such effects have yet to bereported for DNA methylation, modifications

    of histone proteins are associated with expo-sure to drugs of abuse and stressors in rodent

    models (Renthal et al. 2009, Renthal & Nestler

    2008). These findings suggest that epigene

    states, including DNA methylation, are alteby a wide range of biologically relevant eve

    (Meaney & Szyf 2005, Renthal & Nestler 20Szyf et al. 2005). Such epigenetic modificatio

    might therefore underlie a wide range of s

    ble changes in neural function following expsure to highlysalient events (e.g.,chronic stre

    drugs of abuse, reproductive phases such as penting) and are thus logical mechanisms

    environmentally induced alterations in menhealth (Akbarian & Huang 2009, Jiang et

    2008, Tsankova et al. 2007).

    EPIGENETICS ANDMENTAL HEALTH

    Emerging evidence links the alterations in gexpression associated with DNA methylati

    to psychiatric illness. Cortical dysfunctionschizophrenia is associated with changes

    GABAergic circuitry (Benes & Berretta 200This effect is associated with a decrease

    the expression of the GAD1 gene that ecodes for a specific form of glutamic acid

    carboxylase (GAD67), one to two key enzymfor GABA synthesis in cortical interneuro

    There is compelling evidence for the decrea

    expression of GAD67 in cortical tissues frschizophrenic patients (Akbarian & Hua

    2006, Costa et al. 2004). The dysregulaGAD67 expression in the chandelier GA

    neurons is thought to result in disruptionsynchronized cortical activity and impairm

    of executive functions in schizophrenia subje(Lewis et al. 2005). Likewise, allelic variation

    GAD1is associated with schizophrenia (Straet al. 2007).

    In addition to GAD67, there is alsodecrease in cortical expression of reelin

    schizophrenic brains (Eastwood & Harris

    2003); reelin is closely associated with syntic plasticity. The same GABAergic neurons

    the schizophrenic brain that express reelin aGAD67 exhibit an increase in DNA meth

    transferases 1 (DNMT1; Veldic et al. 200DNMT1 is a member of a family of

    zymes that transfers a methyl group from t

    456 Zhang Meaney

  • 8/11/2019 Zhang Meaney 10 AnnRPsychol Epigenetics Review

    19/34

    methyl donor S-adenosyl-methionine (SAM)

    onto cytosines, thus producing DNA methy-lation. The promoter for thereelingene shows

    increased methylation in the brains of patients

    with schizophrenia compared with control sub-jects (Abdolmaleky et al. 2005, Grayson et al.

    2005). Kundakovic et al. (2007) showed that theinhibition of DNMT1 in neuronal cell lines re-

    sulted in the increased expression of both reelinand GAD67. The increase in gene expression

    was associated with a decreased association ofMeCP2, further suggesting that these differ-

    ences are associated with alteration in DNAmethylation. Recall that maternal care directly

    alters DNA methylation of the GAD67 pro-

    moter in the rat (Zhang et al. 2009). This effectis associated with a decrease in DNMT1 ex-

    pression and reduced MeCP2 association withtheGAD1promoter.

    An important question is that of the devel-opmental origins of such differences in DNA

    methylation. A set of recent studies (McGowanet al. 2009) suggests that epigenetic modifi-

    cations might occur in humans in responseto variations in parentoffspring interactions.

    DNA was extracted from hippocampal samplesobtained from victims of suicide or from

    individuals who had died suddenly from othercauses (auto accidents, heart attacks, etc.).

    The samples were obtained from the Quebec

    Suicide Brain Bank, which conducts forensicphenotyping that includes a validated assess-

    ment of psychiatric status and developmentalhistory (e.g., McGirr et al. 2008). The studies

    examined the methylation status of the exon 1Fpromoter of the glucocorticoid receptor, which

    corresponds to the exon 17promoter in the rat(Turner & Muller 2005). The results showed

    increased DNA methylation of the exon 1Fpromoter in hippocampal samples from suicide

    victims compared with controls, but only if

    suicide was accompanied with a developmentalhistory of child maltreatment. Child maltreat-

    ment, independent of psychiatric state, pre-dicted the DNA methylation status of the exon

    1Fpromoter. As in the previous rodent studies,the methylation state of the exon 1F promoter

    also determined the ability of NGFI-A to bind

    to the promoter andactivate gene transcription.

    Although such studies are obviously correla-tional and limited by postmortem approaches,

    the results are nevertheless consistent with

    the hypothesis that variations in parental carecan modify the epigenetic state of selected

    sites of the human genome. Moreover, thefindings are also consistent with studies that

    link childhood abuse to individual differencesin stress responses (Heim et al. 2000). Child-

    hood abuse is associated with an increase inpituitary ACTH responses to stress among

    individuals with or without concurrent majordepression. These findings are particularly

    relevant, since pituitary ACTH directly reflectscentral activation of the HPA stress response,

    and hippocampal glucocorticoid receptor

    activation dampens HPA activity. The findingsin humans are consistent with the rodent

    studies cited above investigating epigeneticregulation of the glucocorticoid receptor

    gene and with the hypothesis that early lifeevents can alter the epigenetic state of relevant

    genomic regions, the expression of which maycontribute to individual differences in the risk

    for psychopathology (Holsboer 2000, Neigh& Nemeroff 2006, Schatzberg et al. 1985).

    Certain limitations need to be considered aswe integrate epigenetics into the study of psy-

    chopathology. The study of epigenetic mech-

    anisms in humans is complicated by the factthat epigenetic marks are often tissue-specific.

    For example, the brain contains some neu-rons that synthesize and release dopamine as

    a neurotransmitter and others that rely onacetylcholine. We might assume that among

    dopaminergic neurons, the genes associatedwith the capacity for acetylcholine production

    are silenced, likely through some level of epi-genetic regulation. Such processes are inherent

    in the specialization of brain cells, as with allother differentiated cells in the body. This pro-

    cess of specialization involves epigenetic reg-

    ulation and implies that the epigenetic marksvary from cell type to cell type. Indeed, there is

    considerable variation in epigeneticmarks fromone brain region to another, perhaps even more

    so than variation within the same brain region

    www.annualreviews.org Epigenetics and the Environment 457

  • 8/11/2019 Zhang Meaney 10 AnnRPsychol Epigenetics Review

    20/34

    across individuals (Ladd-Acosta et al. 2007).

    Brain samples are for the most part beyonddirect examination in the living individualat the

    level of molecular analysis. This often leaves us

    with measures of DNA extracted from bloodor saliva and with the question of whether the

    epigenetic marks within such samples actuallyreflect those within the relevant neuronal pop-

    ulation. Thus, for the time being advances inthe study of neuroepigenetics will rely heavily

    on relevant models with nonhuman species aswell as complementary studies of samples from

    postmortem human brains.

    CONCLUSIONS

    It is now evident that genomic variation at

    the level of nucleotide sequence is associatedwith individual differences in personality and

    thus with vulnerability and resistance to a widerange of chronic illness (Ebstein 2006, Meyer-

    Lindenberg & Weinberger 2006, Rutter 2007).The challenge is how to conceptually integrate

    thefindings from genetics into psychology. Theoperation of the genome is regulated by cellu-

    lar signals that are responsive to environmentalconditions. Thus, the effects of genetic varia-

    tion are contextually determined and therefore

    best considered as probabilistic. Genetic vari-ations influence cellular activity and, depend-

    ing upon current and past environmental con-ditions, will bias toward particular functional

    outcomes. The molecular events that mediategene transcription reveal the interdependence

    of gene and environment (Sokolowski 2001,Sokolowski & Wahlsten 2001). Oddly, what is

    perhaps the most profound comment on thisissue dates back several years. In response to

    a question from a journalist considering therelative importance of nature versus nurture

    in defining individual differences in personal-ity, Hebb responded that such comparisons are

    akin to asking what contributes more to the

    area of rectangle, the length or the width? Therecent flush of studies examining gene x en-

    vironment effects on personality and vulnera-bility/resistance to mental illness (Caspi et al.

    2003, Meaney 2009, Rutter 2007, Suomi 2006)

    reflects the interdependence of genetic and

    vironmental influences, such that the effectone level can only be understood within t

    context of the other. Indeed, developmenprocesses are best considered as the outco

    of a constant dialog between the genome a

    its environment (Bateson 1994; Gottelieb 191998; Lewontin 1974).

    The gene x environment perspective is cical in the establishment of an understand

    of the development of individual differencesneural function and personality. Until recen

    most experimental approaches were limitedidentifying factors that could influence neu

    development. Our own studies of maternal cin the rat are a case in point. This research

    amines the effects of variation in maternal cin animals that are housed from weaning o

    ward under identical conditions. We systema

    cally minimize variation from weaning onwaThis approach permits conclusions as to the p

    tential effects of variations in maternal care bcannot estimate the importance of such effe

    for individual differences in adult function uder natural conditions. Indeed, environmen

    enrichment in the postweaning period can verse effects associated with the variations

    maternal care (Bredy et al. 2004, Champag& Meaney 2006, Zhang et al. 2006). Likew

    studies of monozygotic-dizygotic twins exaine what are, in effect, differences in paren

    gene dosage while minimizing variation in

    early environment. Such approaches have pvided convincing evidence that genetic fact

    caninfluence thedevelopment of individualdferences, but do not identify how. Indeed,

    challenge is to define how, when, and unwhich conditions specific genetic or enviro

    ment factors operate to regulate developmeHerein lies the enormous contribution of t

    gene x environment perspective, particulawhen integrated into longitudinal studies

    development.The excitement concerning the findin

    in the area of epigenetics derives from

    realization that such mechanisms could fothe biological basis for the interplay betwe

    environmental signals and the genome. T

    458 Zhang Meaney

  • 8/11/2019 Zhang Meaney 10 AnnRPsychol Epigenetics Review

    21/34

    studies reviewed here suggest that (a) epi-

    genetic remodeling occurs in response tothe environmental activation of the classic

    activity-dependent cellular signaling path-

    ways that are associated with synaptic plasticity,(b) epigenetic marks, particularly DNA methy-

    lation, are actively remodeled over early devel-opment in response to environmental events

    that regulate neural development and function,and (c) epigenetic marks at histone proteins and

    the DNA are subject to remodeling in responseto environmental influences even at later stages

    in development. We have highlighted examplesof environmental influences that are of obvious

    relevance for psychologists. However, increas-

    ing evidence from animal studies indicates thatprenatal and early postnatal environmental

    factors, including nutritional supplements,xenobiotic chemicals, and reproductive tech-

    nologies, can alter the epigenetic state ofspecific genomic regions ( Jirtle & Skinner

    2007).These findings suggest that epigenetic re-

    modeling might serve as an ideal mechanismfor phenotypic plasticitythe process whereby

    the environment interacts with the genome toproduce individual differences in the expression

    of specific traits. One could easily imagine that

    such processes mediate observed discordancesbetween m