Neuroscience Research: Jerry Guintivano, Zachary A. Kaminsky

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Neuroscience Research 102 (2016) 56–66

Contents lists available at ScienceDirect

Neuroscience Research
journal homepage: www.elsevier.com/locate/neures

Review article

Role of epigenetic factors in the development of mental illness


throughout life
Jerry Guintivano, Zachary A. Kaminsky ∗
The Mood Disorders Center, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA

a r t ic l e in f o a b s t r a c t

Article history: Psychiatric disease is believed to result from a combination of genetic vulnerability and environmen-
Received 28 February 2014 tal influence. At the crux are epigenetic modifications, which mediate the influence of environment
Received in revised form 31 July 2014 on the genome. Twin and genome-wide association studies demonstrate a wide range of heritabilities
Accepted 4 August 2014
across psychiatric disorders, while epidemiological and animal models implicate distinct developmental
Available online 20 August 2014
windows where environmental factors may interact with genetic vulnerability to confer risk. Certain
developmental periods appear to be more prone to these influences including during gestation, in the
Keywords:
early postnatal period, and during periods of major hormonal rearrangement. Here we review the role of
Epigenetics
DNA methylation
environmental factors capable of epigenetic reprogramming during these periods and present evidence
Psychiatric disease for the link between these modifications and disease. The cross tissue relevance of environmentally
Heritability induced epigenetic change and its utility for identifying peripheral biomarkers is discussed.
Environmental influence © 2014 Elsevier Ireland Ltd and the Japan Neuroscience Society. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
1.1. Nature and nurture in psychiatric disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
1.2. Heritability in psychiatric disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
1.3. Epigenetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 58
1.4. The possibility for epigenetic inheritance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 58
1.5. The intersection of epigenetics and the environment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
2. Maternal diet and epigenetically mediated effects on neurodevelopment outcomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
2.1. Gestational consequences of maternal stress . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 60
2.2. Early life adversity mediated epigenetic reprogramming . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 60
2.3. The epigenetic intersect of gonadal hormonal fluctuation and mood . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 61
2.4. Evidence for epigenetic associations in post mortem brain DNA methylation studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 61
2.5. The utility of epigenetic disease etiology: biomarkers and personalized medicine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 62
3. Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 62
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 62

1. Introduction unknown, it is thought their onset is due to an underlying genetic


predisposition in combination with environmental insults. Many
Psychiatric disorders are complex diseases classified into clinical studies indicate that the genetic component for these disorders can
syndromes with little known etiology. Those who have psychiatric vary, with heritability ranges from 0.37 in major depression (MDD)
disorders live longer with the burden of their disease compared to to 0.81 in schizophrenia (SCZ) (Sullivan et al., 2012). Despite the
any other (Whiteford et al., 2013). While the etiologies of these are amount of information generated from genetic association studies,
recent studies have focused on the role of epigenetics, specifically
DNA methylation, and how it may contribute to disease risk. At the
∗ Corresponding author at: 720 Rutland Avenue Ross Research Building 1070 intersection of genes and environment, DNA methylation provides
Baltimore, MD 21205, USA. Tel.: +1 443 287 0093. a mechanism for different environmental factors to alter genetic
E-mail address: zkamins1@jhmi.edu (Z.A. Kaminsky). expression. In this paper, we discuss evidence for the contribution

http://dx.doi.org/10.1016/j.neures.2014.08.003
0168-0102/© 2014 Elsevier Ireland Ltd and the Japan Neuroscience Society. All rights reserved.
J. Guintivano, Z.A. Kaminsky / Neuroscience Research 102 (2016) 56–66 57

Fig. 1. Developmental periods sensitive to epigenetic change. An individual’s risk to psychiatric disease may be modified by epigenetic changes induced by environmental
insults. Depicted here are three particularly vulnerable timepoints: gestation, early childhood development, and reproductive years.

of both genetic and environmental factors in the etiology of psychi- as frequent in MZ twins than DZ twins (Torgersen, 1983) suggest-
atric disease across key developmental windows (gestation, early ing that the disease is caused purely by genetic factors. Alzheimer’s
childhood development, and during reproductive years) for epige- disease exhibits a high heritability (Kringlen, 1999), while neurode-
netic reprogramming (Fig. 1). velopmental disorders such as autism spectrum disorder (ASD) and
attention deficit hyperactivity disorder (ADHD) exhibit high heri-
1.1. Nature and nurture in psychiatric disease tabilities when assessed in childhood (Posthuma and Polderman,
2013). On the other end of the scale, MDD usually exhibits a gener-
Like most complex diseases, psychiatric diseases are believed ally low heritability around 30% (Bierut et al., 1999; Kendler et al.,
to result from the combined influences of ‘nature’ and ‘nurture’. 1993; McGuffin et al., 1991; Middeldorp et al., 2005; Sullivan et al.,
These two terms respectively refer to those inherited factors influ- 2000; Torgersen, 1986), while most reviewed heritability values for
encing one’s development and later life outcomes as opposed to suicidality range from 26 to 70% (Pedersen and Fiske, 2010). Other
those factors external to one’s self. In more modern terms, nature psychiatric disorders such as anxiety, somatoform disorders, and
refers to genetic factors while nurture refers to environmental alcohol abuse exhibit lower heritabilities according to twin stud-
influences. Much of what is known about the degree to which these ies (Kringlen, 1999). The results and interpretation of such modern
two influences play a role in disease manifestation comes from the twin studies have fueled the search for the implicated genetic and
employment of the classical twin design. environmental factors.
The classical twin design relies on a comparison of the concord- In recent years, this search has culminated in genetic screening
ance rates for a given trait between monozygotic (MZ) to dizygotic technologies that have enabled comprehensive investigations of
(DZ) twins. This design has long been regarded as one of the most common genetic variation on the genome-wide level. The degree
elegant systems through which to infer the influence of inherited to which the pool of genome-wide significant results from genome-
factors on a trait, as the degree of both genetic and environmental wide association studies (GWAS) has grown across psychiatric
variation occurring in both groups is known. MZ twins arise from diseases and has generally reflected initial heritability estimates
a single fertilized egg, while DZ twins result from two eggs being (Ripke et al., 2013); although it should be kept in mind that those
fertilized by two separate sperm (Gringras and Chen, 2001). These diseases with the highest heritabilities are currently also the best
alternate origins result in MZ twins sharing approximately 100% powered to date (Ripke et al., 2013). The comparative deluge
DNA sequence identity while DZ twins share about 50% of all seg- of genome-wide significant results in MDD, however, stand as a
regating DNA polymorphisms, on average (Boomsma et al., 2002; marked contrast to the results of SCZ and support the notion that
Martin et al., 1997; Wong et al., 2005). Importantly, the degree of psychiatric diseases with low heritability estimates may therefore
shared environment in both MZ and DZ twins is high, so in the tra- be more under the control of environmental influences. What is
ditional model, if the DZ twin group is more variable for a trait, it not attributed to genetics in heritability studies falls into the cat-
is said to be under the influence of heritable factors. Heritability egory of non-shared environmental influence and, in the case of
(Boomsma et al., 2002; Gottesman, 2004) is the degree to which MDD, this portion represents a relatively large (∼70%) piece of the
a trait is due to genetic differences, but does not indicate what pie. In a recent review of the current state of genetic evidence
specific genetic factors are involved in influencing trait develop- in MDD, Cohen-Woods et al. (2013) suggest that the conflicting
ment. Conversely, the remainder of the variation for a trait not genetic results in MDD may be a consequence of a failure to account
accounted for by heritability estimates is generally attributed to for environmental influence.
the effects of non-shared environmental influence. A recent large scale analysis of available GWAS data across a
range of psychiatric diseases including SCZ, BP, MDD, ASD, ADHD,
1.2. Heritability in psychiatric disease among others demonstrates a striking overlap in the amount of
common genetic risk contributing across various combinations
Using the classical twin design, relatively large ranges of her- of these disorders (Lee et al., 2013a,b). For example, SCZ was
itabilities and environmental influences have been identified for most genetically similar to BP, but also shared genetic etiology
numerous psychiatric diseases (Wong et al., 2005). For example with MDD and to a much lesser extent, ASD. Cumulatively, the
SCZ and bipolar disorder (BP) exhibit relatively high levels of inher- data suggest that a common genetic etiology is shared across
ited risk, between 60 and 80% (Bertelsen et al., 1977; Cardno and a range of phenotypically distinct psychiatric disorders. GWAS
Gottesman, 2000; Kieseppa et al., 2004; Lee et al., 2013a,b). An eval- techniques have also enabled the quantification of heritability
uation of 36 MZ and 53 DZ twins found panic disorder to be twice based on genetic variation at single nucleotide polymorphisms
58 J. Guintivano, Z.A. Kaminsky / Neuroscience Research 102 (2016) 56–66

(SNP), known as “SNP heritability”. In the case of MDD, this value genetic control. One such example observed in various human
estimated to be approximately 23%, which is lower than the 30% leukemias (Boumber et al., 2008) is ‘methylation encroachment’,
suggested by classical twin studies (Lee et al., 2013a,b). These which occurs when SNPs influencing transcription factor binding
specific types of “SNP heritability” speak only to the heritability result in the spreading of highly methylated CpG island shores into
accounted for by the measured genetic polymorphisms, while twin relatively unmethylated CpG islands (Mummaneni et al., 1998).
studies will identify the presence of all heritable factors indepen- Somatic mutations in the epigenetic modifiers may also influence
dent of their source. In general, the lower estimate when accounting epigenetic patterns. For example, a SNP in the EZH2 gene result
for a majority of common genetic variation suggests the intriguing in increased histone 3 lysine 27 (H3K27) tri-methylation marks in
possibility that additional sources of non-genetic variation may be other cancers (Yap et al., 2010). Genome-wide studies have shown a
affecting the measurement of heritability. Additionally, the data majority of sequence associated epigenetic change occurs in cis and
suggest the possibility that risk to psychiatric disease is conferred are often referred to as allele specific methylation (ASM) (Bell et al.,
through the interaction of an inherited genetic susceptibility that 2010; Gertz et al., 2011; Numata et al., 2012; Schalkwyk et al., 2010;
must interact with contributing environmental risk factors to result Tycko, 2010). In one multigenerational study, ASM patterns were
in disease and that specific combinations of interactions may dis- consistent across six family members spanning three generations
tinguish between disorders. This interpretation is consistent with (Gertz et al., 2011), suggesting that the inherited genetic template
the observations of non-Mendelian features in psychiatric disease, may result in the passage of epigenetic signatures to subsequent
which include sexual dimorphism (Kaminsky et al., 2006), discor- generations.
dance of identical twins (Kaminsky et al., 2009), and non-Mendelian Importantly, as GWAS studies implicate a shared genetic eti-
modes of inheritance (Petronis, 2001). ology across psychiatric disorders, these findings suggest the
possibility that deleterious contribution to disease etiology is on
1.3. Epigenetics the epigenetic level. For example, a portion of heritable DNA
methylation loci identified in one of the genome-wide heritabil-
Epigenetic factors include methylation and histone protein ity twin studies of DNA methylation above were found to be
modifications. In this review, we will primarily discuss DNA associated with genetic variations in cis (Boks et al., 2009). A
methylation, as we focus mainly on human studies, which have select list of top GWAS hits in BP were shown to be significantly
concentrated more on this epigenetic mark because of its stability associated with cerebellar DNA methylation changes in proxi-
and relative ease of measurement relative to histone modifications. mal regions (Gamazon et al., 2013), while an alternative BP study
In the mammalian genome, DNA methylation occurs on the 5′ car- identified significant enrichment of ASM associated SNPs in gene
bon of the cytosine ring in cytosine-guanine dinucleotides (CpG). ontologies of gated channel and neurotransmitter receptor func-
DNA methylation is associated with a heterochromatic state that tion (Chuang et al., 2013). These studies highlight the fact that
can limit gene transcription as well as regulate numerous other inherited genetic risk may provide a DNA template that is primed
genomic functions (Brown, 1981; Dulac, 2010; Ehrlich and Wang, for interacting with the environment in a dichotomous fashion,
1981; Razin and Riggs, 1980; Schwartz and Ast, 2010). Epigenetic promoting either a resilience or vulnerability in the face of envi-
marks are stably inherited following cell division via DNA methyl- ronmental insults mediated by the reprogramming of epigenetic
transferase enzymes (Chen and Li, 2006), which can recognize the factors.
hemi-methylated duplex strand of DNA following replication and Beyond the influence of genetic factors resulting in inherited
facilitate the addition of a methyl-group to the corresponding loca- epigenetic patterns, the possibility for a transgenerational pas-
tion on the newly synthesized daughter strand. Importantly, this sage of epigenetic signals, conventionally defined as an epigenetic
inheritance refers to retaining the epigenetic code during mitotic change that persists across three generations, would offer yet
cell replication. Additionally, stochastic rearrangements, hormonal another explanation to the high heritabilities observed in numer-
influences (Kangaspeska et al., 2008) and environmental influences ous psychiatric diseases and the relatively low degree of identified
(Jaenisch and Bird, 2003; Razin and Cedar, 1993; Riggs et al., 1998) genetic findings. Can epigenetic inheritance account for this “Miss-
can result in a drift of these regulatory molecular signals over time. ing Heritability” as it has been termed (Manolio et al., 2009)?
It is conventionally believed that there is no passage of epige-
1.4. The possibility for epigenetic inheritance netic information from parent to offspring generations due to
the massive epigenetic rearrangements occurring during game-
Until recently, no studies had investigated evidence for a her- togenesis and at fertilization (Allegrucci et al., 2005; Li, 2002;
itable component to DNA methylation on the genomic scale. The Morgan et al., 2005; Santos and Dean, 2004; Santos et al., 2005),
first such study was published in 2009 in Nature Genetics, whereby including a global demethylation of DNA accompanied by mas-
a comparison of 20 pairs of MZ twins and to 20 same sex matched sive histone modification rearrangement (Hajkova et al., 2008). In
DZ twin pairs from peripheral blood, and a similarly sized sam- mice; however, there are clear exceptions to this rule that often
ple from buccal swab samples demonstrated significantly higher involve the interaction of the parental generation with environ-
DNA methylation variation between matched co-twins in the DZ mental influences that result in epigenetically mediated inherited
as opposed to the MZ group (Kaminsky et al., 2009). This finding phenotypes in the offspring (Morgan et al., 1999; Rakyan et al.,
represented the first evidence for a heritable component to DNA 2003). Other experiments demonstrate clear evidence for a direct
methylation in both the blood and buccal samples on the genomic effect of the environment on the germline resulting in epigenetic
scale. This result has subsequently been replicated by numerous changes that may be transmitted to the next generation (Anway
additional studies performed by different laboratories (Bocklandt et al., 2008; Dias and Ressler, 2014). Few examples of epigenetic
et al., 2011; Boks et al., 2009; Gervin et al., 2011; Gordon et al., changes across generations exist in humans, possibly because it
2011). Cumulatively, it appears that all epigenomic twin studies is difficult to study. Critically, if the molecular mechanisms that
performed to date are detecting at least some degree of a heritable enable environmentally induced germline epigenetic reprogramm-
influence on DNA methylation patterns across tissues in humans, ing in mice to pass to the next generation are also seen in humans,
the substrate of which may include both genetic and non-genetic the implications for complex psychiatric disease is that a portion
inheritance. of the heritability implicated by twin and family studies may,
A larger degree of epigenetic variation in genetically non- in part, be influenced by environmental exposures in the parent
identical twins implies that epigenetic factors are, in part, under generation.
J. Guintivano, Z.A. Kaminsky / Neuroscience Research 102 (2016) 56–66 59

1.5. The intersection of epigenetics and the environment the developing fetus is protected from stress hormones and inhibit
the delivery of serotonin, brain derived neurotrophic factor (BDNF),
Epigenetic signatures are transmitted through cell division to and leptin, which play important roles in fetal brain development
daughter cells; however, several environmental factors can change (Grissom and Reyes, 2013).
epigenetic patterns in nonspecific ways across the genome, or SGA has been implicated as a risk factor for a number of psy-
at specific sites, which may result in altered phenotypes. In this chiatric disorders. A comparison of a 4650 SCZ cases across three
way, changes to epigenetic patterns represent a molecular medium population-based registries identified an increased SCZ incidence
through which the environment can interact with genetic predispo- ratio associating with cases of SGA (Nielsen et al., 2013). A com-
sition and influence phenotype. Numerous environmental factors parison to case-sibling registries confirmed this effect was due
have been associated with psychiatric disease risk. For example, primarily to gestational factors and not other factors shared across
SCZ onset has been linked to prenatal risk factors including, mater- siblings, such as genetic or postnatal environmental factors. An
nal infection and malnutrition, and advanced paternal age. Other investigation of 724 BP cases and 1147 controls from the Finnish
risk factors associated with SCZ include migrant status, growing up Prenatal Study of Bipolar Disorders identified a significant associa-
in an urban setting, parental loss, substance abuse, and adverse life tion of Caesarian section with BP (Chudal et al., 2014). A significant
events (Bratlien et al., 2014; McDonald and Murray, 2000; McGrath, association of SGA was found with ASD in an analysis of the New
2011; Oher et al., 2014; Sorensen et al., 2014). Environmental risks Jersey Population (Maramara et al., 2014), while an MRI based study
for MDD include both low as well as high birth weight relative to of brain morphology demonstrated an association of low in birth
gestational age (Grissom and Reyes, 2013; Tamashiro and Moran, weight with cortical surface area and reductions in cognitive per-
2010) early life adversity, trauma, or sexual abuse (Klengel and formance (Haukvik et al., 2014).
Binder, 2013), hormonal fluctuation (Kessler et al., 1993, 1995; Dysregulated cell proliferation in the hypothalamus is a con-
Weissman and Olfson, 1995), and stress (Stankiewicz et al., 2013). sequence of both SGA and LGA, which affects nutrient sensing
The development of ASD has been linked to early developmental pathways in the developing organism and may be due, in part,
environmental factors such as maternal weight gain but not preg- to the restricted placental hormone delivery mentioned previ-
nancy BMI (Bilder et al., 2013), maternal migrant status (Lehti et al., ously. In animal models, fetal hypothalamic neuronal progenitor
2013), advanced maternal age (35 years or older), low birth weight, cells exposed to leptin showed a neuronal cell fate commitment,
multiple gestation, gestational age at birth, breech presentation, while insulin had a similar effect driving to glial cell fate commit-
prolonged labor, low Apgar score at 5 min, hypoxia, and parental ment as a function of DNA methylation changes (Desai et al., 2011;
psychiatric status (Larsson et al., 2005; Maramara et al., 2014). Hirabayashi and Gotoh, 2010). Both leptin and insulin also medi-
Each of the environmental risk factors above are associated ate development of the ventral tegmental area (VTA), the origin of
with specific developmental time periods where their effects can dopaminergic neurons in the mesocorticolimbic reward pathway.
be most influential. During these periods, environmental expo- The VTA drives hedonic feeding behavior (Grissom and Reyes, 2013;
sures alone or in combination may lead to epigenetic changes that Narayanan et al., 2010) and may be responsible for obesity changes
can reprogram key genes, altering developmental trajectories and seen in the Dutch famine offspring mentioned previously (Ravelli
increasing the risk for psychiatric disease. Over the course of devel- et al., 1999). Animal models also support the mechanism of altered
opment these subtle changes may lead to a greater vulnerability to BDNF signaling in the brain and placenta of SGA and LGA offspring
stress and culminate in disease, all mediated by epigenetic mech- (Wang and Xu, 2007), which may be linked with observed deficits
anisms. The sections below highlight the influence of different in spatial learning and memory that are associated with reduced
developmental risk factors relevant to epigenetic change. hippocampal volumes (Ranade et al., 2008).
Changes in neuronal development seen in SGA and LGA offspring
may be a result of epigenetic alterations in the brain due to mater-
2. Maternal diet and epigenetically mediated effects on nal diet. Using animal models, Vucetic et al. (2010) reported that
neurodevelopment outcomes in the nucleus accumbens (NAc) and hypothalamus of offspring
maternal high fat diet reduced DNA methylation in the promoter of
The environmental exposures during gestation represent the genes involved in the mesocorticolimbic reward pathway, includ-
earliest non-genetically mediated source of variation potentially ing the dopamine reuptake transporter (DAT), !-opioid receptor
conferring risk to disease. The mother’s womb is meant to direct (MOR), and preproenkephalin (PENK) genes. Subsequently, Carlin
and support early fetal development, with maternal diet influ- et al. (2013) reported that supplementing the maternal diet with
encing developmental outcome of her offspring. One of the first methyl-donors reversed the observed high fat diet induced changes
studies introducing the idea of maternal diet and in utero pro- in DNA global hypomethylation and the associated weight gain.
gramming of offspring found a significant increase in BMI in female In humans, maternal diet induced alterations to DNA methyla-
offspring who were exposed during gestation to the Dutch famine tion may result in MDD associated VTA hyperactivity observed
of 1945–1955 (Ravelli et al., 1999). While this example relates in neuroimaging studies (Kumar et al., 2008; Rive et al., 2013).
specifically to obesity, it illustrates the effect of stress on mater- Further evidence that epigenetic patterns may be affected by pro-
nal environment on offspring. Also, it should be noted that there cesses affecting gestational growth come from the aforementioned
is a high comorbidity between MDD and obesity (Levitan et al., twin studies identifying epigenetic heritability. A second consis-
2012), and that being in the bottom tenth percentile or upper tenth tently replicated finding from those works was twins that share a
percentile of gestational weight is associated with obesity, MDD, placenta, known as monochorionic twins, have significantly more
and other psychiatric disorders (Grissom and Reyes, 2013). Infants epigenetic variation than those with separate placentas, or dichori-
born into the bottom tenth percentile are referred to as small for onic twins. Sharing a placenta between twins results in unequal
gestational age (SGA), while those in the top tenth are large for blood flow between the developing siblings. In extreme cases, this
gestational age (LGA)(ibid). Low maternal nutrition and restricted can result in a condition known as twin-to-twin transfusion syn-
blood flow to the placenta give rise to cases of SGA (Godfrey et al., drome (TTTS), whereby one twin is essentially nutritionally starved
2011; Grissom and Reyes, 2013; Resnick et al., 1982), while LGA during development (Lopriore et al., 2011). This suggests that ges-
is seen in mothers with obesity and increased weight gain during tational inequality can induce epigenetic disruption and has the
pregnancy (Drake and Reynolds, 2010). In addition, maternal obe- potential to affect key systems leading to the development of psy-
sity can restrict development of the placenta, which can alter how chiatric outcomes. Importantly, approximately 70% of twin pairs
60 J. Guintivano, Z.A. Kaminsky / Neuroscience Research 102 (2016) 56–66

are monochorionic and are likely to have experienced some degree pregnancy associate with DNA methylation levels in the cord blood
of gestational inequality (Hall, 2003). While few epigenomic twin of offspring at the low affinity glucocorticoid receptor (GR) gene,
studies are available in the psychiatric literature, the handful of NR3C1, which is activated by the binding of cortisol. Similarly,
those performed to date demonstrate evidence for epigenetic drift Bromer et al. (2012) demonstrate a significant association between
between genetically identical yet discordant co-twins. Peripheral placental NR3C1 DNA methylation and NICU Network Neurobehav-
blood DNA methylation was assessed genome-wide in a sample ioral Scales (NNNS), an indicator of early neurobehavioral outcome.
of twins discordant for BP and SCZ, resulting in the identification In addition, placental epigenetic changes at the 11"-HSD2 gene
of over 100 CpG dinucleotides differentially methylated between are also associated with altered NNNS (Marsit et al., 2012). This
affected and unaffected MZ twins (Dempster et al., 2011). Pathway finding was replicated in a prenatal stress rat model where similar
analysis identified enrichment of DNA methylation changes asso- changes to placental 11"-HSD2 were observed and were correlated
ciated with genes related to ‘psychological disorders’, ‘dopamine with methylation changes in the cortex of offspring at the same
receptor signaling’, and ‘nervous system development and func- gene (Jensen Pena et al., 2012). The stress-exposed offspring were
tion’. Another genome-wide investigation of DNA methylation in shown to have elevations in cortical DNMT1, which may regulate
MDD discordant twins identified a higher epigenetic variation in changes observed in 11"-HSD2 DNA methylation (ibid). The role of
affected co-twins relative to their unaffected siblings (Byrne et al., 11"-HSD2 is to metabolize maternal cortisol in the placenta to limit
2013), leading the authors to posit that affected twins may be fetal exposure, further supporting the hypothesis that neurobehav-
subjected to environmentally induced epigenetic instability. Taken ioral outcomes in the offspring may be mediated through hormone
together, the results suggest that variations in maternal diet may induced epigenetic reprogramming. Other studies suggest that ges-
alter the developmental trajectories through epigenetic mecha- tational factors influence adult depression-like behavior through
nisms, potentially leading to psychiatric disease. methylation of the #CGRP gene (Jiao et al., 2012).
The above mechanisms represent a means for epigenetic change
2.1. Gestational consequences of maternal stress to be transmitted from one generation to another. Those offspring
exposed to maternal cortisol in utero may in turn subject their own
Along with the epigenetic alterations induced by maternal diet offspring to increased levels of cortisol when they are pregnant.
on the neural development of offspring, there is mounting evidence The epigenetic foundations of cortisol dysregulation laid down dur-
that suggest offspring of women suffering from depression during ing pregnancy in offspring may be perpetuated. But this epigenetic
pregnancy are at a greater risk for various negative outcomes and mechanism along with diet-associated changes in DNA methyla-
future psychiatric illness. In a comprehensive review examining tion may affect how the results of previous heritability studies have
the outcomes of antenatal depression on offspring, Davalos et al. guided research into psychiatry over the past 30 years. These trans-
(2012) states that both maternal depression during pregnancy and generational epigenetic changes can be viewed as a component of
negative offspring outcomes are highly prevalent yet understudied. heritability outside of pure genetic component, though the mech-
Research in this field is limited, in part, by a lack of consideration of anisms presented here only represent a portion of the potential
antidepressant treatment during the antenatal period. For example, risks for developing psychiatric disorders. While the current data
maternal but not paternal depression was significantly associated supporting these hypotheses is limited, future work should be
with the incidence of ASD in a sample of 4429 individuals with aimed at studying the relationship between maternal stress during
autism and 43,277 matched controls (Rai et al., 2013); however, it gestation and offspring outcomes.
is unclear from this study how much of the effect was mediated
by prenatal antidepressant use. For this reason, the most informa- 2.2. Early life adversity mediated epigenetic reprogramming
tive studies limit their inclusion criteria to treatment naïve cases
of antenatal depression and the subsequent offspring effects. In Following birth the brain continues to undergo rapid and
the Davalos et al. (2012) review, only 14 studies meet this crite- dynamic change as neurodevelopment continues (Knickmeyer
rion. From these, numerous observations of low birth weight and et al., 2008), however environmental factors have been thought
preterm birth associate with offspring of treatment naïve mothers to increase risk for the development of psychiatric illness such as
who were antenatally depressed. In some cases, these metrics were SCZ (Rapoport et al., 2012; Stachowiak et al., 2013) ASD (O’Hearn
correlated with maternal cortisol during pregnancy. Cumulatively, et al., 2008), and affective disorders (Ansorge et al., 2007; North
these studies propose that risk for psychiatric disease later in life et al., 2013; Sigurdsson et al., 1999). More specifically, environmen-
could begin in utero, via maternal anxiety and depression during tal influences such as mothering behavior and early life traumas
pregnancy leading to altered HPA-axis function and development can alter trajectories of cognitive development and stress response
of brain structures related to emotional procession. later on in life (Odgers and Jaffee, 2013). It is not only widely stud-
There is growing evidence that maternal affective changes ied and associated with extreme events like childhood sexual abuse
coincide with hormonal alterations that cross the placenta and or assault that convey risk for disease, but also chronic and more
mediate epigenetic reprogramming of the offspring hypothalamic– common stressors such as familial hostility, financial and food inse-
pituitary–adrenal (HPA) axis (Dulawa, 2014). Stressful life events curity, and under-resourced schools and neighborhoods (Odgers
during pregnancy, thought to be moderated by anxiety levels, were and Jaffee, 2013). Epigenetic changes, such as DNA methylation
moderately associated with elevated risk of psychosis in offspring reprogramming in the brain, represent the “molecular medium”
at 12 years (Dorrington et al., 2014). Maternal depression antena- through which these environmental stressors may alter genetic
tally, measured by the Edinburgh Postnatal Depression Scale (EPDS) pathways. Weaver et al. (2004) published the seminal study that
was associated with microstructure changes in the right amygdala demonstrates how maternal care can impact epigenetic patterns.
of their offspring (Rifkin-Graboi et al., 2013). Some studies have They showed that differences in arched back nursing and groom-
identified higher cortisol and stress behaviors in neonates born to ing of rat pups altered DNA methylation in the NGFI-A transcription
mothers with antenatal depression. While increased maternal anx- factor-binding site of the NR3C1 gene. These changes to the DNA
iety was associated with decreased dopamine and norepinephrin methylation led to an altered stress response in the offspring rats
levels in offspring (Field et al., 2003), both of which may be later in life. Similarly in mice, early life stress has demonstrated
downstream consequences of placental alteration in estrogen and an increase in arginine vasopressin gene transcription driven by
cortisol (Shansky and Lipps, 2013). Hompes et al. report that a decrease in DNA methylation within hypothalamic paraven-
maternal anxiety and serum cortisol measurements taken during tricular nucleus neurons, a critical region in the regulation of
J. Guintivano, Z.A. Kaminsky / Neuroscience Research 102 (2016) 56–66 61

neuroendocrine stress response (Murgatroyd et al., 2009). 2013; Zhou et al., 2014). Post mortem studies have shown that there
McGowan et al. showed that in humans epigenetic variation at the may be sex-specific influences driving DNA methylation differences
exon 1F promoter of NR3C1 in post mortem hippocampal tissue from seen in SCZ and BP (Mill et al., 2008).
suicide victims, syntenic to the Weaver et al. locus, associated with For women, the reproductive years are punctuated with periods
early life trauma in the form of abuse. Using genome-wide microar- of hormonal fluctuation that coincide with increased incidences of
rays, Mehta et al., identified distinct epigenetic profiles associated psychiatric disease. Specifically, estrogen fluctuations are thought
with gene transcription changes in blood from those with PTSD to contribute to times of psychiatric risk given estrogen’s role as an
coinciding with childhood abuse status (Mehta et al., 2013). Path- antidepressant, anxiolytic, and neuroprotectant (Galea et al., 2001;
way analysis of the DNA methylation induced gene changes suggest Romano-Torres and Fernandez-Guasti, 2010). Estrogen, like other
an alteration in “central nervous system”, “tolerance induction”, steroid hormones, binds to and activates its respective hormone
and “apoptosis and growth rate” pathways between those with receptors which have been shown to alter chromatin structure and
childhood trauma and those without. Importantly, this study shows DNA methylation through various interacting protein partners (Fu
that early childhood trauma can result in persistent changes in DNA et al., 2003, 2004; Kaminsky et al., 2006; Kangaspeska et al., 2008;
methylation and that these early environmental influences are both Metivier et al., 2003, 2008). A combination of these sex and tissue
widespread and detectable across tissues. specific epigenetic differences, along with underlying genetic pre-
One implication of these findings is that the passage of HPA axis disposition, has been suggested to influence the level of response
sensitivity and GR mediated stress response phenotypes associ- to gonadal hormone. Major changes in the hormonal milieu may
ated with psychiatric disease risks could be influenced by mood induce epigenetic changes in vulnerable women resulting in phe-
disorders affecting the early postnatal period, such as postpartum notypic changes such as premenstrual dysphoric disorder (PMDD),
depression (PPD). Only a handful of heritability studies have been premenstrual syndrome (PMS), perimenapausal depression, PPD
performed in PPD; however, a systematic review of the literature (Payne et al., 2009), and an increased probability in hospitalization
suggests that treatment of PPD is insufficient to affect the long- for women with SCZ during the leuteal phase of their menstrual
term outcomes in child health (Gunlicks and Weissman, 2008). This cycle (Riecher-Rossler and Hafner, 2000). Further, the role of estro-
finding could be related to either an underlying genetic susceptibil- gen in psychiatric risk is supported by work done by Joinson et al.
ity or to risk conferred from the above intrauterine environmental (2012) where they found depressive symptoms among girls during
mechanisms that are likely to be comorbid with PPD risk. adolescence were more strongly influenced by breast development
stage, rather than timing of menarche. This suggests that the onset
2.3. The epigenetic intersect of gonadal hormonal fluctuation and of depression during puberty may be due to increasing levels of
mood estrogen and may account for the sex differences seen in depression
rates during the reproductive years.
The reproductive years have been shown to be a time of PPD represents a psychiatric phenotype typifying the poten-
increased vulnerability to psychiatric illness, potentially due to tial intersection between epigenetics and gonadal hormones. PPD
the effects of gonadal hormones and their role in psychiatric risk. occurs following parturition, when there is a rapid withdrawal of
During the reproductive years, there are varying degrees of sexual estrogen and progesterone. Importantly, numerous studies have
dimorphism among psychiatric diseases, an observable phenotypic demonstrated that levels of these hormones do not vary between
difference between the sexes, including SCZ, Alzheimer’s disease, PPD and non-PPD women (Studd, 2011). Numerous studies have
anxiety disorders, ASD, and alcohol dependence (Aleman et al., implicated that risk is conferred through an increased sensitiv-
2003; Menger et al., 2010; Petronis, 2001; Seeman, 1997). However, ity to estrogen (Bloch et al., 2000; Mehta et al., 2014; Mitsushima
sex differences among those disorders have been understudied. et al., 2003), and more recently, that risk is conferred through an
The best examples of sex differences are widely observed in MDD. increased sensitivity to estrogen induced DNA methylation change
Prior to puberty and the onset of gonadal hormones, the preva- in the hippocampus (Guintivano et al., 2013a). While extensive
lence for MDD is equal between the sexes. Following puberty, evidence implicates the hippocampus as the site for estrogen’s
however, the rate of MDD more than doubles in females com- anxiolytic and antidepressant effects on mood (Green and Galea,
pared with males (Nolen-Hoeksema and Girgus, 1994; Wade et al., 2008; MacLusky et al., 2005; Suda et al., 2008; ter Horst, 2010;
2002), suggesting that an interaction of genetic susceptibility with Walf et al., 2008), altered sensitivity to estrogen may also result in
major hormonal changes may contribute to the manifestation of the down stream alterations in HPA-axis function and vulnerability to
psychiatric phenotype. This sex difference appears in twin studies stress (Bloch et al., 2000, 2005; Mehta et al., 2014; Mitsushima et al.,
where proband-wise MZ concordance for MDD differs by gender 2003; Shansky and Lipps, 2013; Skalkidou et al., 2012; Tarantino
with 31% for male-MZ twins and 48% for female-MZ twins (Kendler et al., 2011). Shansky and Lipps (2013) posit that estrogen may
and Prescott, 1999), suggesting an increased heritability of depres- exacerbate the effects of stress on glucocorticoid release, which is
sion risk in the female population. Similarly, an analysis of 6265 consistent with higher cortisol response in PPD cases observed dur-
twin pairs from the Netherlands Twin Registry demonstrated a ing simulated pregnancy (Skalkidou et al., 2012). Finally, women
higher heritability for thoughts of self harm and suicidality for with PPD have been shown to have exacerbated stress-induced cor-
females (H = 0.74) as compared to males (H = 0.45) (Althoff et al., tisol and corticotropin releasing hormone (CRH) responses during
2012). The divergent amounts of circulating hormones between pregnancy (Nierop et al., 2006; Yim et al., 2009), which results in
the sexes are thought to contribute to the manifestation of psychi- reductions following childbirth (Skalkidou et al., 2012; Tsigos and
atric phenotypes. It is known that androgens and estrogens have Chrousos, 2002), a period already characterized by decreased HPA-
differential effects on neural development, affecting programmed axis function (Groer and Morgan, 2007; Harris et al., 1996; Jolley
cell death, cellular migration, synaptogenesis and axonal migra- et al., 2007; Taylor et al., 2009).
tion, and formation of sexually distinct neuronal circuits (Simerly,
2002, 2005). While perhaps an over simplification of our current 2.4. Evidence for epigenetic associations in post mortem brain
understanding of hormonal effects on the developing brain, var- DNA methylation studies
ious studies have shown that androgens are related to neurite
arborization and neuronal survival, while estrogens are responsi- One of the challenges in the study of psychiatric epigenetics
ble for synapse formation and initiation of cellular communication is that DNA methylation is tissue and cell type specific, meaning
(Brandt et al., 2013; Hamson et al., 2013; Lustig, 1994; Ottem et al., that to understand the role of epigenetic variation in the brain,
62 J. Guintivano, Z.A. Kaminsky / Neuroscience Research 102 (2016) 56–66

one must study the brain. This fact has limited psychiatric studies blood (Guintivano et al., 2013a). This altered sensitivity to estrogen
in humans to post mortem studies, which confer numerous chal- could be due to changes in DNA methylation of the estrogen recep-
lenges including a difficulty to assess the cause vs. effect nature tors. The first month of postnatal development is associated with
of any identified changes and to exclude the influence of various increased methylation of the estrogen receptor alpha promoter
confounders such as disease specific differences in psychiatric med- (Westberry et al., 2010; Westberry and Wilson, 2012). Alterations
ication, post mortem interval, brain pH, and neuronal relative to to this normal developmental process, such as differences in rear-
glial proportions. Variation in the detailed recording of medication ing (Edelmann and Auger, 2011; Pena et al., 2013), may result in
history and other substances complicates findings. Until recently, DNA methylation changes that persist into adulthood and affect the
no methods were available to account for cellular heterogeneity downstream signaling of estrogen. Could similar scenarios be pos-
without the separation of neuronal and glial nuclei by fluorescence sible in response to systemic exposure to other hormones? While
activated sorting (Jiang et al., 2008). More recently, Guintivano potentially all hormones may be candidates for such a mechanism,
et al. (2013b) developed the first bioinformatic methods to con- a good example is the stress hormone cortisol because dysregu-
trol for cellular heterogeneity based on DNA methylation proxies lated HPA axis function is a common feature to many psychiatric
in genome-wide data after which various other methods have been diseases. Etiological insults resulting in altered cortisol response
developed (Jaffe and Irizarry, 2014). As analytical methods improve such as those observed in MDD may be expected to leave proxy
to make the study of post mortem tissues easier and more devoid epigenetic reprogramming in peripheral tissues to some degree
of confounding factors, it is possible that novel epigenetic associa- mirroring patterns that might be expected in the brain. In the
tions will be discovered with the analysis of new brain samples or above developmental time points reviewed in this paper, systemic
the re-analysis of existing data using novel methods. Unfortunately, influences of diet and hormonal factors are generally implicated in
the availability of post mortem brains are rare relative to what is epigenetic reprogramming. Such early environmental influences
available from peripheral tissues. The ability to assess and iden- may therefore leave early yet stable epigenetic proxies of brain
tify epigenetic associations in peripheral blood would open up vast related deleterious gene regulation. These epigenetic biomarkers
opportunities for biomarker discovery and personalized medicine may then be used to develop better prospective diagnostics for
and greatly add to the statistical power available for psychiatric disease and allow for the testing of prophylactic psychiatric treat-
epigenetic studies. ments as well as pharmacoepigenetic status, allowing treatments
to be tailored to the response profile of the individual.
2.5. The utility of epigenetic disease etiology: biomarkers and
personalized medicine
3. Summary
The degree to which peripheral tissues like blood or saliva
may contain brain relevant epigenetic signatures is a topic of con- Risk to psychiatric disease is likely mediated by the complex
tinued study and has been reviewed extensively by Gladkevich interaction between underlying genetic susceptibility, environ-
et al. (2004). Some initial reports in small sample sizes have mental exposures, and their downstream epigenetic consequences
investigated this issue (Davies et al., 2012). Davies et al. performed to mediate long lasting changes in gene regulation. Twin studies
MeDIP sequencing on blood, prefrontal cortex, and cerebellum implicate a range of evidence for the contribution of inherited fac-
from the same post mortem individuals and identified numerous tors and environmental influences across numerous psychiatric
regions of tissue specific epigenetic variation; however, they noted disorders. Distinct developmental windows during gestation, in
that some inter-individual variation was reflected across brain the early postnatal environment, and during periods of major hor-
and blood. These inter-individual associations across tissues were monal rearrangement may result in epigenetic changes that confer
observed between blood, which is composed of multiple cell types risk to disease. The system wide nature of the majority of these
each of which may have unique changes in DNA methylation, and influences, be it dietary or hormonal, leave the potential for cross
brain, which may undergo varying degrees of active demethyla- tissue epigenetic reprogramming. Such an epigenetic record of past
tion in addition to methylation events (Gavin et al., 2013). In the environmental influences opens the door for the use of peripheral
case of these cross tissue findings, what factors might mediate an tissues such as blood to study psychiatric phenotypes.
agreement of epigenetic patterns in a systemic manner? Petronis
has suggested that a shared developmental origin prior to major
tissue differentiation, such that inherited or early developmentally References
acquired epimutations may be reflected across primary germ layers
Aleman, A., Kahn, R.S., Selten, J.P., 2003. Sex differences in the risk of schizophrenia:
(Petronis, 2006). In these cases, developmentally acquired epige- evidence from meta-analysis. Arch. Gen. Psychiatry 60, 565–571.
netic change in response to intrauterine environmental factors like Allegrucci, C., Thurston, A., Lucas, E., Young, L., 2005. Epigenetics and the germline.
maternal anxiety or diet may initiate such early epimutations that Reproduction 129, 137–149.
Althoff, R.R., Hudziak, J.J., Willemsen, G., Hudziak, V., Bartels, M., Boomsma, D.I., 2012.
are propagated across germ layers. Due to the epigenetic repro- Genetic and environmental contributions to self-reported thoughts of self-harm
gramming inherent in the differentiation process, it follows that and suicide. Am. J. Med. Genet. B Neuropsychiatr. Genet. 159B, 120–127.
inherited or developmentally acquired epimutations would stand Ansorge, M.S., Hen, R., Gingrich, J.A., 2007. Neurodevelopmental origins of depressive
disorders. Curr. Opin. Pharmacol. 7, 8–17.
the best chance of being detected in regions not responsible for Anway, M.D., Rekow, S.S., Skinner, M.K., 2008. Comparative anti-androgenic actions
differentiation. of vinclozolin and flutamide on transgenerational adult onset disease and sper-
Alternative mechanisms may be responsible for cross tissue matogenesis. Reprod. Toxicol. 26, 100–106.
Bell, C.G., Finer, S., Lindgren, C.M., Wilson, G.A., Rakyan, V.K., Teschendorff, A.E.,
correlations of epigenetic patterns such as those observed above
Akan, P., Stupka, E., Down, T.A., Prokopenko, I., Morison, I.M., Mill, J., Pidsley, R.,
in PPD where the implicated risk for disease involves a response Deloukas, P., Frayling, T.M., Hattersley, A.T., McCarthy, M.I., Beck, S., Hitman, G.A.,
to systemic hormone exposure capable of reprogramming epige- 2010. Integrated genetic and epigenetic analysis identifies haplotype-specific
methylation in the FTO type 2 diabetes and obesity susceptibility locus. PLoS
netic patterns. Peripheral detection of disease specific epigenetic
ONE 5, e14040.
variation in peripheral blood is therefore a proxy marker of the Bertelsen, A., Harvald, B., Hauge, M., 1977. A Danish twin study of manic-depressive
deleterious hormonal influences on the brain in the disease group. disorders. Br. J. Psychiatry 130, 330–351.
Importantly, in our own work we identified an increased sensitiv- Bierut, L.J., Heath, A.C., Bucholz, K.K., Dinwiddie, S.H., Madden, P.A., Statham, D.J.,
Dunne, M.P., Martin, N.G., 1999. Major depressive disorder in a community-
ity of estrogen induced epigenetic change in PPD above were able based twin sample: are there different genetic and environmental contributions
to prospectively predict PPD status using epigenetic biomarkers in for men and women? Arch. Gen. Psychiatry 56, 557–563.
J. Guintivano, Z.A. Kaminsky / Neuroscience Research 102 (2016) 56–66 63

Bilder, D.A., Bakian, A.V., Viskochil, J., Clark, E.A., Botts, E.L., Smith, K.R., Pimentel, Fu, M., Wang, C., Zhang, X., Pestell, R.G., 2004. Acetylation of nuclear receptors in
R., McMahon, W.M., Coon, H., 2013. Maternal prenatal weight gain and autism cellular growth and apoptosis. Biochem. Pharmacol. 68, 1199–1208.
spectrum disorders. Pediatrics 132, e1276–e1283. Galea, L.A., Wide, J.K., Barr, A.M., 2001. Estradiol alleviates depressive-like symp-
Bloch, M., Rubinow, D.R., Schmidt, P.J., Lotsikas, A., Chrousos, G.P., Cizza, G., 2005. toms in a novel animal model of post-partum depression. Behav. Brain Res. 122,
Cortisol response to ovine corticotropin-releasing hormone in a model of preg- 1–9.
nancy and parturition in euthymic women with and without a history of Gamazon, E.R., Badner, J.A., Cheng, L., Zhang, C., Zhang, D., Cox, N.J., Gershon, E.S.,
postpartum depression. J. Clin. Endocrinol. Metab. 90, 695–699. Kelsoe, J.R., Greenwood, T.A., Nievergelt, C.M., Chen, C., McKinney, R., Shilling,
Bloch, M., Schmidt, P.J., Danaceau, M., Murphy, J., Nieman, L., Rubinow, D.R., 2000. P.D., Schork, N.J., Smith, E.N., Bloss, C.S., Nurnberger, J.I., Edenberg, H.J., Foroud,
Effects of gonadal steroids in women with a history of postpartum depression. T., Koller, D.L., Scheftner, W.A., Coryell, W., Rice, J., Lawson, W.B., Nwulia,
Am. J. Psychiatry 157, 924–930. E.A., Hipolito, M., Byerley, W., McMahon, F.J., Schulze, T.G., Berrettini, W.H.,
Bocklandt, S., Lin, W., Sehl, M.E., Sanchez, F.J., Sinsheimer, J.S., Horvath, S., Vilain, E., Potash, J.B., Zandi, P.P., Mahon, P.B., McInnis, M.G., Zollner, S., Zhang, P., Craig,
2011. Epigenetic predictor of age. PLoS ONE 6, e14821. D.W., Szelinger, S., Barrett, T.B., Liu, C., 2013. Enrichment of cis-regulatory gene
Boks, M.P., Derks, E.M., Weisenberger, D.J., Strengman, E., Janson, E., Sommer, I.E., expression SNPs and methylation quantitative trait loci among bipolar disorder
Kahn, R.S., Ophoff, R.A., 2009. The relationship of DNA methylation with age, susceptibility variants. Mol. Psychiatry 18, 340–346.
gender and genotype in twins and healthy controls. PLoS ONE 4, e6767. Gavin, D.P., Chase, K.A., Sharma, R.P., 2013. Active DNA demethylation in post-
Boomsma, D., Busjahn, A., Peltonen, L., 2002. Classical twin studies and beyond. Nat. mitotic neurons: a reason for optimism. Neuropharmacology 75, 233–245.
Rev. Genet. 3, 872–882. Gertz, J., Varley, K.E., Reddy, T.E., Bowling, K.M., Pauli, F., Parker, S.L., Kucera,
Boumber, Y.A., Kondo, Y., Chen, X., Shen, L., Guo, Y., Tellez, C., Estecio, M.R., Ahmed, K.S., Willard, H.F., Myers, R.M., 2011. Analysis of DNA methylation in a
S., Issa, J.P., 2008. An Sp1/Sp3 binding polymorphism confers methylation pro- three-generation family reveals widespread genetic influence on epigenetic reg-
tection. PLoS Genet. 4, e1000162. ulation. PLoS Genet. 7, e1002228.
Brandt, N., Vierk, R., Rune, G.M., 2013. Sexual dimorphism in estrogen-induced Gervin, K., Hammero, M., Akselsen, H.E., Moe, R., Nygard, H., Brandt, I., Gjessing, H.K.,
synaptogenesis in the adult hippocampus. Int. J. Dev. Biol. 57, 351–356. Harris, J.R., Undlien, D.E., Lyle, R., 2011. Extensive variation and low heritability
Bratlien, U., Oie, M., Haug, E., Moller, P., Andreassen, O.A., Lien, L., Melle, I., 2014. of DNA methylation identified in a twin study. Genome Res. 21, 1813–1821.
Environmental factors during adolescence associated with later development Gladkevich, A., Kauffman, H.F., Korf, J., 2004. Lymphocytes as a neural probe:
of psychotic disorders – a nested case-control study. Psychiatry Res. 215 (3), potential for studying psychiatric disorders. Prog. Neuropsychopharmacol. Biol.
579–585. Psychiatry 28, 559–576.
Bromer, C., Marsit, C.J., Armstrong, D.A., Padbury, J.F., Lester, B., 2012. Genetic and Godfrey, K.M., Inskip, H.M., Hanson, M.A., 2011. The long-term effects of pre-
epigenetic variation of the glucocorticoid receptor (NR3C1) in placenta and natal development on growth and metabolism. Semin. Reprod. Med. 29,
infant neurobehavior. Dev. Psychobiol. 55 (7), 673–683. 257–265.
Brown, D.D., 1981. Gene expression in eukaryotes. Science 211, 667–674. Gordon, L., Joo, J.H., Andronikos, R., Ollikainen, M., Wallace, E.M., Umstad, M.P., Per-
Byrne, E.M., Carrillo-Roa, T., Henders, A.K., Bowdler, L., McRae, A.F., Heath, A.C., mezel, M., Oshlack, A., Morley, R., Carlin, J.B., Saffery, R., Smyth, G.K., Craig, J.M.,
Martin, N.G., Montgomery, G.W., Krause, L., Wray, N.R., 2013. Monozygotic twins 2011. Expression discordance of monozygotic twins at birth: effect of intrauter-
affected with major depressive disorder have greater variance in methylation ine environment and a possible mechanism for fetal programming. Epigenetics
than their unaffected co-twin. Transl. Psychiatry 3, e269. 6, 579–592.
Cardno, A.G., Gottesman II, I.I., 2000. Twin studies of schizophrenia: from bow-and- Gottesman, S.L.T.A.I.I., 2004. Twin studies and the genetics of mental disorders in
arrow concordances to Star Wars Mx and functional genomics. Am. J. Med. Genet. the genomic age. In: Smith, G.A.a.B. (Ed.), Neuroscience Encyclopedia. , 3rd ed.
97, 12–17. Elsevier Science, Amsterdam.
Carlin, J., George, R., Reyes, T.M., 2013. Methyl donor supplementation blocks the Green, A.D., Galea, L.A., 2008. Adult hippocampal cell proliferation is suppressed
adverse effects of maternal high fat diet on offspring physiology. PLOS ONE 8, with estrogen withdrawal after a hormone-simulated pregnancy. Horm. Behav.
e63549. 54, 203–211.
Chen, T., Li, E., 2006. Establishment and maintenance of DNA methylation patterns Gringras, P., Chen, W., 2001. Mechanisms for differences in monozygous twins. Early
in mammals. Curr. Top. Microbiol. Immunol. 301, 179–201. Hum. Dev. 64, 105–117.
Chuang, L.C., Kao, C.F., Shih, W.L., Kuo, P.H., 2013. Pathway analysis using information Grissom, N.M., Reyes, T.M., 2013. Gestational overgrowth and undergrowth affect
from allele-specific gene methylation in genome-wide association studies for neurodevelopment: similarities and differences from behavior to epigenetics.
bipolar disorder. PLOS ONE 8, e53092. Int. J. Dev. Neurosci. 31, 406–414.
Chudal, R., Sourander, A., Polo-Kantola, P., Hinkka-Yli-Salomaki, S., Lehti, V., Groer, M.W., Morgan, K., 2007. Immune, health and endocrine characteristics of
Sucksdorff, D., Gissler, M., Brown, A.S., 2014. Perinatal factors and the risk of depressed postpartum mothers. Psychoneuroendocrinology 32, 133–139.
bipolar disorder in Finland. J. Affect. Disord. 155, 75–80. Guintivano, J., Arad, M., Gould, T.D., Payne, J.L., Kaminsky, Z.A., 2013a. Antenatal
Cohen-Woods, S., Craig, I.W., McGuffin, P., 2013. The current state of play on the prediction of postpartum depression with blood DNA methylation biomarkers.
molecular genetics of depression. Psychol. Med. 43, 673–687. Mol. Psychiatry 19 (5), 560–567.
Davalos, D.B., Yadon, C.A., Tregellas, H.C., 2012. Untreated prenatal maternal depres- Guintivano, J., Aryee, M.J., Kaminsky, Z.A., 2013b. A cell epigenotype specific model
sion and the potential risks to offspring: a review. Arch. Womens Ment. Health for the correction of brain cellular heterogeneity bias and its application to age,
15, 1–14. brain region and major depression. Epigenetics 8, 290–302.
Davies, M.N., Volta, M., Pidsley, R., Lunnon, K., Dixit, A., Lovestone, S., Coarfa, C., Gunlicks, M.L., Weissman, M.M., 2008. Change in child psychopathology with
Harris, R.A., Milosavljevic, A., Troakes, C., Al-Sarraj, S., Dobson, R., Schalkwyk, L.C., improvement in parental depression: a systematic review. J. Am. Acad. Child
Mill, J., 2012. Functional annotation of the human brain methylome identifies Adolesc. Psychiatry 47, 379–389.
tissue-specific epigenetic variation across brain and blood. Genome Biol. 13, R43. Hajkova, P., Ancelin, K., Waldmann, T., Lacoste, N., Lange, U.C., Cesari, F., Lee, C.,
Dempster, E.L., Pidsley, R., Schalkwyk, L.C., Owens, S., Georgiades, A., Kane, F., Almouzni, G., Schneider, R., Surani, M.A., 2008. Chromatin dynamics during
Kalidindi, S., Picchioni, M., Kravariti, E., Toulopoulou, T., Murray, R.M., Mill, J., epigenetic reprogramming in the mouse germ line. Nature 452, 877–881.
2011. Disease-associated epigenetic changes in monozygotic twins discordant Hall, J.G., 2003. Twinning. Lancet 362, 735–743.
for schizophrenia and bipolar disorder. Hum. Mol. Genet. 20, 4786–4796. Hamson, D.K., Wainwright, S.R., Taylor, J.R., Jones, B.A., Watson, N.V., Galea, L.A.,
Desai, M., Li, T., Ross, M.G., 2011. Fetal hypothalamic neuroprogenitor cell culture: 2013. Androgens increase survival of adult-born neurons in the dentate gyrus
preferential differentiation paths induced by leptin and insulin. Endocrinology by an androgen receptor-dependent mechanism in male rats. Endocrinology
152, 3192–3201. 154, 3294–3304.
Dias, B.G., Ressler, K.J., 2014. Parental olfactory experience influences behavior and Harris, B., Lovett, L., Smith, J., Read, G., Walker, R., Newcombe, R., 1996. Cardiff
neural structure in subsequent generations. Nat. Neurosci. 17, 89–96. puerperal mood and hormone study. III. Postnatal depression at 5 to 6 weeks
Dorrington, S., Zammit, S., Asher, L., Evans, J., Heron, J., Lewis, G., 2014. Perinatal postpartum and its hormonal correlates across the peripartum period. Br. J.
maternal life events and psychotic experiences in children at twelve years in a Psychiatry 168, 739–744.
birth cohort study. Schizophr. Res. 152, 158–163. Haukvik, U.K., Rimol, L.M., Roddey, J.C., Hartberg, C.B., Lange, E.H., Vaskinn, A., Melle,
Drake, A.J., Reynolds, R.M., 2010. Impact of maternal obesity on offspring obesity I., Andreassen, O.A., Dale, A., Agartz, I., 2014. Normal birth weight variation is
and cardiometabolic disease risk. Reproduction 140, 387–398. related to cortical morphology across the psychosis spectrum. Schizophr. Bull.
Dulac, C., 2010. Brain function and chromatin plasticity. Nature 465, 728–735. 40, 410–419.
Dulawa, S.C., 2014. Epigenetic programing of depression during gestation. Bioessays. Hirabayashi, Y., Gotoh, Y., 2010. Epigenetic control of neural precursor cell fate
Edelmann, M.N., Auger, A.P., 2011. Epigenetic impact of simulated maternal groom- during development. Nat. Rev. Neurosci. 11, 377–388.
ing on estrogen receptor alpha within the developing amygdala. Brain Behav. Jaenisch, R., Bird, A., 2003. Epigenetic regulation of gene expression: how the genome
Immun. 25, 1299–1304. integrates intrinsic and environmental signals. Nat. Genet. 33 (Suppl), 245–254.
Ehrlich, M., Wang, R.Y., 1981. 5-Methylcytosine in eukaryotic DNA. Science 212, Jaffe, A.E., Irizarry, R.A., 2014. Accounting for cellular heterogeneity is critical in
1350–1357. epigenome-wide association studies. Genome Biol. 15, R31.
Field, T., Diego, M., Hernandez-Reif, M., Schanberg, S., Kuhn, C., Yando, R., Bendell, Jensen Pena, C., Monk, C., Champagne, F.A., 2012. Epigenetic effects of prenatal stress
D., 2003. Pregnancy anxiety and comorbid depression and anger: effects on the on 11beta-hydroxysteroid dehydrogenase-2 in the placenta and fetal brain. PLoS
fetus and neonate. Depress. Anxiety 17, 140–151. ONE 7, e39791.
Fu, M., Rao, M., Wang, C., Sakamaki, T., Wang, J., Di Vizio, D., Zhang, X., Albanese, Jiang, Y., Matevossian, A., Huang, H.S., Straubhaar, J., Akbarian, S., 2008. Isolation of
C., Balk, S., Chang, C., Fan, S., Rosen, E., Palvimo, J.J., Janne, O.A., Muratoglu, S., neuronal chromatin from brain tissue. BMC Neurosci. 9, 42.
Avantaggiati, M.L., Pestell, R.G., 2003. Acetylation of androgen receptor enhances Jiao, J., Opal, M.D., Dulawa, S.C., 2012. Gestational environment programs adult
coactivator binding and promotes prostate cancer cell growth. Mol. Cell. Biol. 23, depression-like behavior through methylation of the calcitonin gene-related
8563–8575. peptide gene. Mol. Psychiatry 18 (12), 1273–1280.
64 J. Guintivano, Z.A. Kaminsky / Neuroscience Research 102 (2016) 56–66

Joinson, C., Heron, J., Araya, R., Paus, T., Croudace, T., Rubin, C., Marcus, M., Lewis, G., M.L., Pericak-Vance, M.A., Pickard, B.S., Pimm, J., Piven, J., Posthuma, D., Potash,
2012. Association between pubertal development and depressive symptoms in J.B., Poustka, F., Propping, P., Puri, V., Quested, D.J., Quinn, E.M., Ramos-Quiroga,
girls from a UK cohort. Psychol. Med. 42, 2579–2589. J.A., Rasmussen, H.B., Raychaudhuri, S., Rehnstrom, K., Reif, A., Ribases, M., Rice,
Jolley, S.N., Elmore, S., Barnard, K.E., Carr, D.B., 2007. Dysregulation of the J.P., Rietschel, M., Roeder, K., Roeyers, H., Rossin, L., Rothenberger, A., Rouleau,
hypothalamic-pituitary-adrenal axis in postpartum depression. Biol. Res. Nurs. G., Ruderfer, D., Rujescu, D., Sanders, A.R., Sanders, S.J., Santangelo, S.L., Sergeant,
8, 210–222. J.A., Schachar, R., Schalling, M., Schatzberg, A.F., Scheftner, W.A., Schellenberg,
Kaminsky, Z., Wang, S.C., Petronis, A., 2006. Complex disease, gender and epigenetics. G.D., Scherer, S.W., Schork, N.J., Schulze, T.G., Schumacher, J., Schwarz, M., Scol-
Ann. Med. 38, 530–544. nick, E., Scott, L.J., Shi, J., Shilling, P.D., Shyn, S.I., Silverman, J.M., Slager, S.L.,
Kaminsky, Z.A., Tang, T., Wang, S.C., Ptak, C., Oh, G.H., Wong, A.H., Feldcamp, L.A., Smalley, S.L., Smit, J.H., Smith, E.N., Sonuga-Barke, E.J., St Clair, D., State, M., Stef-
Virtanen, C., Halfvarson, J., Tysk, C., McRae, A.F., Visscher, P.M., Montgomery, fens, M., Steinhausen, H.C., Strauss, J.S., Strohmaier, J., Stroup, T.S., Sutcliffe, J.S.,
G.W., Gottesman, I.I., Martin, N.G., Petronis, A., 2009. DNA methylation profiles Szatmari, P., Szelinger, S., Thirumalai, S., Thompson, R.C., Todorov, A.A., Tozzi, F.,
in monozygotic and dizygotic twins. Nat. Genet. 41, 240–245. Treutlein, J., Uhr, M., van den Oord, E.J., Van Grootheest, G., Van Os, J., Vicente,
Kangaspeska, S., Stride, B., Metivier, R., Polycarpou-Schwarz, M., Ibberson, D., Car- A.M., Vieland, V.J., Vincent, J.B., Visscher, P.M., Walsh, C.A., Wassink, T.H., Wat-
mouche, R.P., Benes, V., Gannon, F., Reid, G., 2008. Transient cyclical methylation son, S.J., Weissman, M.M., Werge, T., Wienker, T.F., Wijsman, E.M., Willemsen,
of promoter DNA. Nature 452, 112–115. G., Williams, N., Willsey, A.J., Witt, S.H., Xu, W., Young, A.H., Yu, T.W., Zammit,
Kendler, K.S., Neale, M.C., Kessler, R.C., Heath, A.C., Eaves, L.J., 1993. The lifetime S., Zandi, P.P., Zhang, P., Zitman, F.G., Zollner, S., Devlin, B., Kelsoe, J.R., Sklar, P.,
history of major depression in women. Reliability of diagnosis and heritability. Daly, M.J., O’Donovan, M.C., Craddock, N., Sullivan, P.F., Smoller, J.W., Kendler,
Arch. Gen. Psychiatry 50, 863–870. K.S., Wray, N.R., 2013a. Genetic relationship between five psychiatric disorders
Kendler, K.S., Prescott, C.A., 1999. A population-based twin study of lifetime major estimated from genome-wide SNPs. Nat. Genet. 45, 984–994.
depression in men and women. Arch. Gen. Psychiatry 56, 39–44. Lee, S.H., Neale, R.S., Faraone, B.M., Purcell, S.V., Perlis, S.M., Mowry, R.H., Thapar, B.J.,
Kessler, R.C., McGonagle, K.A., Swartz, M., Blazer, D.G., Nelson, C.B., 1993. Sex and Goddard, A., Witte, M.E., Absher, J.S., Agartz, D., Akil, I., Amin, H., Andreassen, F.,
depression in the National Comorbidity Survey: I. Lifetime prevalence chronicity Anjorin, O.A., Anney, A., Anttila, R., Arking, V., Asherson, D.E., Azevedo, P., Back-
and recurrence. J. Affect. Disord. 29, 85–96. lund, M.H., Badner, L., Bailey, J.A., Banaschewski, A.J., Barchas, T., Barnes, J.D.,
Kessler, R.C., Sonnega, A., Bromet, E., Hughes, M., Nelson, C.B., 1995. Posttraumatic Barrett, M.R., Bass, T.B., Battaglia, N., Bauer, A., Bayés, M., Bellivier, M., Bergen, F.,
stress disorder in the National Comorbidity Survey. Arch. Gen. Psychiatry 52, Berrettini, S.E., Betancur, W., Bettecken, C., Biederman, T., Binder, J., Black, E.B.,
1048–1060. Blackwood, D.W., Bloss, D.H., Boehnke, C.S., Boomsma, M., Breen, D.I., Breuer, G.,
Kieseppa, T., Partonen, T., Haukka, J., Kaprio, J., Lonnqvist, J., 2004. High concordance Bruggeman, R., Cormican, R., Buccola, P., Buitelaar, N.G., Bunney, J.K., Buxbaum,
of bipolar I disorder in a nationwide sample of twins. Am. J. Psychiatry 161, W.E., Byerley, J.D., Byrne, W.F., Caesar, E.M., Cahn, S., Cantor, W., Casas, R.M.,
1814–1821. Chakravarti, M., Chambert, A., Choudhury, K., Cichon, K., Cloninger, S., Collier,
Klengel, T., Binder, E.B., 2013. Gene-environment interactions in major depressive C.R., Cook, D.A., Coon, E.H., Cormand, H., Corvin, B., Coryell, A., Craig, W.H., Craig,
disorder. Can. J. Psychiatry 58, 76–83. D.W., Crosbie, I.W., Cuccaro, J., Curtis, M.L., Czamara, D., Datta, D., Dawson, S.,
Knickmeyer, R.C., Gouttard, S., Kang, C., Evans, D., Wilber, K., Smith, J.K., Hamer, Day, G., De Geus, R., Degenhardt, E.J., Djurovic, F., Donohoe, S., Doyle, G.J., Duan,
R.M., Lin, W., Gerig, G., Gilmore, J.H., 2008. A structural MRI study of human A.E., Dudbridge, J., Duketis, F., Ebstein, E., Edenberg, R.P., Elia, H.J., Ennis, J., Etain,
brain development from birth to 2 years. J. Neurosci. 28, 12176–12182. S., Fanous, B., Farmer, A., Ferrier, A.E., Flickinger, I.N., Fombonne, M., Foroud, E.,
Kringlen, E., 1999. Twin studies in psychiatry. Tidsskr. Nor. Laegeforen. 119, Frank, T., Franke, J., Fraser, B., Freedman, C., Freimer, R., Freitag, N.B., Friedl, C.M.,
3322–3328. Frisén, M., Gallagher, L., Gejman, L., Georgieva, P.V., Gershon, L., Geschwind, E.S.,
Kumar, P., Waiter, G., Ahearn, T., Milders, M., Reid, I., Steele, J.D., 2008. Abnormal Giegling, D.H., Gill, I., Gordon, M., Gordon-Smith, S.D., Green, K., Greenwood, E.K.,
temporal difference reward-learning signals in major depression. Brain 131, Grice, T.A., Gross, D.E., Grozeva, M., Guan, D., Gurling, W., De Haan, H., Haines,
2084–2093. L., Hakonarson, J.L., Hallmayer, H., Hamilton, J., Hamshere, S.P., Hansen, M.L.,
Larsson, H.J., Eaton, W.W., Madsen, K.M., Vestergaard, M., Olesen, A.V., Agerbo, E., Hartmann, T.F., Hautzinger, A.M., Heath, M., Henders, A.C., Herms, A.K., Hickie,
Schendel, D., Thorsen, P., Mortensen, P.B., 2005. Risk factors for autism: perinatal S., Hipolito, I.B., Hoefels, M., Holmans, S., Holsboer, P.A., Hoogendijk, F., Hot-
factors, parental psychiatric history, and socioeconomic status. Am. J. Epidemiol. tenga, W.J., Hultman, J.J., Hus, C.M., Ingason, V., Ising, A., Jamain, M., Jones, S.,
161, 916–925, discussion 926-918. Jones, E.G., Jones, I., Tzeng, L., Kähler, J.Y., Kahn, A.K., Kandaswamy, R.S., Keller,
Lee, S.H., Ripke, S., Neale, B.M., Faraone, S.V., Purcell, S.M., Perlis, R.H., Mowry, B.J., R., Kennedy, M.C., Kenny, J.L., Kent, E., Kim, L., Kirov, Y., Klauck, G.K., Klei, S.M.,
Thapar, A., Goddard, M.E., Witte, J.S., Absher, D., Agartz, I., Akil, H., Amin, F., Knowles, L., Kohli, J.A., Koller, M.A., Konte, D.L., Korszun, B., Krabbendam, A., Kra-
Andreassen, O.A., Anjorin, A., Anney, R., Anttila, V., Arking, D.E., Asherson, P., sucki, L., Kuntsi, R., Kwan, J., Landén, P., Långström, M., Lathrop, N., Lawrence, M.,
Azevedo, M.H., Backlund, L., Badner, J.A., Bailey, A.J., Banaschewski, T., Barchas, Lawson, J., Leboyer, W.B., Ledbetter, M., Lee, D.H., Lencz, P.H., Lesch, T., Levin-
J.D., Barnes, M.R., Barrett, T.B., Bass, N., Battaglia, A., Bauer, M., Bayes, M., Bel- son, K.P., Lewis, D.F., Li, C.M., Lichtenstein, J., Lieberman, P., Lin, J.A., Linszen,
livier, F., Bergen, S.E., Berrettini, W., Betancur, C., Bettecken, T., Biederman, J., D.Y., Liu, D.H., Lohoff, C., Loo, F.W., Lord, S.K., Lowe, C., Lucae, J.K., MacIntyre,
Binder, E.B., Black, D.W., Blackwood, D.H., Bloss, C.S., Boehnke, M., Boomsma, S., Madden, D.J., Maestrini, P.A., Magnusson, E., Mahon, P.K., Maier, P.B., Mal-
D.I., Breen, G., Breuer, R., Bruggeman, R., Cormican, P., Buccola, N.G., Buitelaar, hotra, W., Mane, A.K., Martin, S.M., Martin, C.L., Mattheisen, N.G., Matthews, M.,
J.K., Bunney, W.E., Buxbaum, J.D., Byerley, W.F., Byrne, E.M., Caesar, S., Cahn, W., Mattingsdal, K., McCarroll, M., McGhee, S.A., McGough, K.A., McGrath, J.J., McGuf-
Cantor, R.M., Casas, M., Chakravarti, A., Chambert, K., Choudhury, K., Cichon, S., fin, P.J., McInnis, P., McIntosh, M.G., McKinney, A., McLean, R., McMahon, A.W.,
Cloninger, C.R., Collier, D.A., Cook, E.H., Coon, H., Cormand, B., Corvin, A., Coryell, McMahon, F.J., McQuillin, W.M., Medeiros, A., Medland, H., Meier, S.E., Melle,
W.H., Craig, D.W., Craig, I.W., Crosbie, J., Cuccaro, M.L., Curtis, D., Czamara, D., S., Meng, I., Meyer, F., Middeldorp, J., Middleton, C.M., Milanova, L., Miranda, V.,
Datta, S., Dawson, G., Day, R., De Geus, E.J., Degenhardt, F., Djurovic, S., Donohoe, Monaco, A., Montgomery, A.P., Moran, G.W., Moreno-De-Luca, J.L., Morken, D.,
G.J., Doyle, A.E., Duan, J., Dudbridge, F., Duketis, E., Ebstein, R.P., Edenberg, H.J., Morris, G., Morrow, D.W., Moskvina, E.M., Muglia, V., Mühleisen, P., Muir, T.W.,
Elia, J., Ennis, S., Etain, B., Fanous, A., Farmer, A.E., Ferrier, I.N., Flickinger, M., Müller-Myhsok, W.J., Murtha, B., Myers, M., Myin-Germeys, R.M., Neale, I., Nel-
Fombonne, E., Foroud, T., Frank, J., Franke, B., Fraser, C., Freedman, R., Freimer, son, M.C., Nievergelt, S.F., Nikolov, C.M., Nimgaonkar, I., Nolen, V., Nöthen, W.A.,
N.B., Freitag, C.M., Friedl, M., Frisen, L., Gallagher, L., Gejman, P.V., Georgieva, L., Nurnberger, M.M., Nwulia, J.I., Nyholt, E.A., O’Dushlaine, D.R., Oades, C., Olincy,
Gershon, E.S., Geschwind, D.H., Giegling, I., Gill, M., Gordon, S.D., Gordon-Smith, R.D., Oliveira, A., Olsen, G., Ophoff, L., Osby, R.A., Owen, U., Palotie, M.J., Parr,
K., Green, E.K., Greenwood, T.A., Grice, D.E., Gross, M., Grozeva, D., Guan, W., A., Paterson, J.R., Pato, A.D., Pato, C.N., Penninx, M.T., Pergadia, B.W., Pericak-
Gurling, H., De Haan, L., Haines, J.L., Hakonarson, H., Hallmayer, J., Hamilton, Vance, M.L., Pickard, M.A., Pimm, B.S., Piven, J., Posthuma, J., Potash, D., Poustka,
S.P., Hamshere, M.L., Hansen, T.F., Hartmann, A.M., Hautzinger, M., Heath, A.C., J.B., Propping, F., Puri, P., Quested, V., Quinn, D.J., Ramos-Quiroga, E.M., Ras-
Henders, A.K., Herms, S., Hickie, I.B., Hipolito, M., Hoefels, S., Holmans, P.A., Hols- mussen, J.A., Raychaudhuri, H.B., Rehnström, S., Reif, K., Ribasés, A., Rice, M.,
boer, F., Hoogendijk, W.J., Hottenga, J.J., Hultman, C.M., Hus, V., Ingason, A., Ising, Rietschel, J.P., Roeder, M., Roeyers, K., Rossin, H., Rothenberger, L., Rouleau, A.,
M., Jamain, S., Jones, E.G., Jones, I., Jones, L., Tzeng, J.Y., Kahler, A.K., Kahn, R.S., Ruderfer, G., Rujescu, D., Sanders, D., Sanders, A.R., Santangelo, S.J., Sergeant,
Kandaswamy, R., Keller, M.C., Kennedy, J.L., Kenny, E., Kent, L., Kim, Y., Kirov, G.K., S.L., Schachar, J.A., Schalling, R., Schatzberg, M., Scheftner, A.F., Schellenberg,
Klauck, S.M., Klei, L., Knowles, J.A., Kohli, M.A., Koller, D.L., Konte, B., Korszun, W.A., Scherer, G.D., Schork, S.W., Schulze, N.J., Schumacher, T.G., Schwarz, J.,
A., Krabbendam, L., Krasucki, R., Kuntsi, J., Kwan, P., Landen, M., Langstrom, N., Scolnick, M., Scott, E., Shi, L.J., Shilling, J., Shyn, P.D., Silverman, S.I., Slager,
Lathrop, M., Lawrence, J., Lawson, W.B., Leboyer, M., Ledbetter, D.H., Lee, P.H., J.M., Smalley, S.L., Smit, S.L., Smith, J.H., Sonuga-Barke, E.N., St Clair, E.J., State,
Lencz, T., Lesch, K.P., Levinson, D.F., Lewis, C.M., Li, J., Lichtenstein, P., Lieber- D., Steffens, M., Steinhausen, M., Strauss, H.C., Strohmaier, J.S., Stroup, J., Sut-
man, J.A., Lin, D.Y., Linszen, D.H., Liu, C., Lohoff, F.W., Loo, S.K., Lord, C., Lowe, J.K., cliffe, T.S., Szatmari, J.S., Szelinger, P., Thirumalai, S., Thompson, S., Todorov,
Lucae, S., MacIntyre, D.J., Madden, P.A., Maestrini, E., Magnusson, P.K., Mahon, R.C., Tozzi, A.A., Treutlein, F., Uhr, J., van den Oord, M., Van Grootheest, E.J.,
P.B., Maier, W., Malhotra, A.K., Mane, S.M., Martin, C.L., Martin, N.G., Mattheisen, Van Os, G., Vicente, J., Vieland, A.M., Vincent, V.J., Visscher, J.B., Walsh, P.M.,
M., Matthews, K., Mattingsdal, M., McCarroll, S.A., McGhee, K.A., McGough, J.J., Wassink, C.A., Watson, T.H., Weissman, S.J., Werge, M.M., Wienker, T., Wijsman,
McGrath, P.J., McGuffin, P., McInnis, M.G., McIntosh, A., McKinney, R., McLean, T.F., Willemsen, E.M., Williams, G., Willsey, N., Witt, A.J., Xu, S.H., Young, W., Yu,
A.W., McMahon, F.J., McMahon, W.M., McQuillin, A., Medeiros, H., Medland, S.E., A.H., Zammit, T.W., Zandi, S., Zhang, P.P., Zitman, P., Zöllner, F.G., International
Meier, S., Melle, I., Meng, F., Meyer, J., Middeldorp, C.M., Middleton, L., Milanova, Inflammatory Bowel Disease Genetics Consortium (IIBDGC), S., Devlin, B., Kel-
V., Miranda, A., Monaco, A.P., Montgomery, G.W., Moran, J.L., Moreno-De-Luca, soe, J.R., Sklar, P., Daly, M.J., O’Donovan, M.C., Craddock, N., Sullivan, P.F., Smoller,
D., Morken, G., Morris, D.W., Morrow, E.M., Moskvina, V., Muglia, P., Muhleisen, J.W., Kendler, K.S., Wray, N.R., 2013b. Identification of risk loci with shared
T.W., Muir, W.J., Muller-Myhsok, B., Murtha, M., Myers, R.M., Myin-Germeys, effects on five major psychiatric disorders: a genome-wide analysis. Lancet 381,
I., Neale, M.C., Nelson, S.F., Nievergelt, C.M., Nikolov, I., Nimgaonkar, V., Nolen, 1371–1379.
W.A., Nothen, M.M., Nurnberger, J.I., Nwulia, E.A., Nyholt, D.R., O’Dushlaine, C., Lehti, V., Hinkka-Yli-Salomaki, S., Cheslack-Postava, K., Gissler, M., Brown, A.S.,
Oades, R.D., Olincy, A., Oliveira, G., Olsen, L., Ophoff, R.A., Osby, U., Owen, M.J., Sourander, A., 2013. The risk of childhood autism among second-generation
Palotie, A., Parr, J.R., Paterson, A.D., Pato, C.N., Pato, M.T., Penninx, B.W., Pergadia, migrants in Finland: a case-control study. BMC Pediatr. 13, 171.
J. Guintivano, Z.A. Kaminsky / Neuroscience Research 102 (2016) 56–66 65

Levitan, R.D., Davis, C., Kaplan, A.S., Arenovich, T., Phillips, D.I., Ravindran, A.V., 2012. Numata, S., Ye, T., Hyde, T.M., Guitart-Navarro, X., Tao, R., Wininger, M., Colantuoni,
Obesity comorbidity in unipolar major depressive disorder: refining the core C., Weinberger, D.R., Kleinman, J.E., Lipska, B.K., 2012. DNA methylation signa-
phenotype. J. Clin. Psychiatry 73, 1119–1124. tures in development and aging of the human prefrontal cortex. Am. J. Hum.
Li, E., 2002. Chromatin modification and epigenetic reprogramming in mammalian Genet. 90, 260–272.
development. Nat. Rev. Genet. 3, 662–673. O’Hearn, K., Asato, M., Ordaz, S., Luna, B., 2008. Neurodevelopment and executive
Lopriore, E., Oepkes, D., Walther, F.J., 2011. Neonatal morbidity in twin-twin trans- function in autism. Dev. Psychopathol. 20, 1103–1132.
fusion syndrome. Early Hum. Dev. 87, 595–599. Odgers, C.L., Jaffee, S.R., 2013. Routine versus catastrophic influences on the devel-
Lustig, R.H., 1994. Sex hormone modulation of neural development in vitro. Horm. oping child. Annu. Rev. Public Health 34, 29–48.
Behav. 28, 383–395. Oher, F.J., Demjaha, A., Jackson, D., Morgan, C., Dazzan, P., Morgan, K., Boydell, J.,
MacLusky, N.J., Luine, V.N., Hajszan, T., Leranth, C., 2005. The 17alpha and Doody, G.A., Murray, R.M., Bentall, R.P., Jones, P.B., Kirkbride, J.B., 2014. The effect
17beta isomers of estradiol both induce rapid spine synapse formation in the of the environment on symptom dimensions in the first episode of psychosis: a
CA1 hippocampal subfield of ovariectomized female rats. Endocrinology 146, multilevel study. Psychol. Med., 1–12.
287–293. Ottem, E.N., Bailey, D.J., Jordan, C.L., Breedlove, S.M., 2013. With a little help from
Manolio, T.A., Collins, F.S., Cox, N.J., Goldstein, D.B., Hindorff, L.A., Hunter, D.J., my friends: androgens tap BDNF signaling pathways to alter neural circuits.
McCarthy, M.I., Ramos, E.M., Cardon, L.R., Chakravarti, A., Cho, J.H., Guttmacher, Neuroscience 239, 124–138.
A.E., Kong, A., Kruglyak, L., Mardis, E., Rotimi, C.N., Slatkin, M., Valle, D., Whitte- Payne, J.L., Palmer, J.T., Joffe, H., 2009. A reproductive subtype of depression: concep-
more, A.S., Boehnke, M., Clark, A.G., Eichler, E.E., Gibson, G., Haines, J.L., Mackay, tualizing models and moving toward etiology. Harv. Rev. Psychiatry 17, 72–86.
T.F., McCarroll, S.A., Visscher, P.M., 2009. Finding the missing heritability of Pedersen, N.L., Fiske, A., 2010. Genetic influences on suicide and nonfatal suicidal
complex diseases. Nature 461, 747–753. behavior: twin study findings. Eur Psychiatry 25, 264–267.
Maramara, L.A., He, W., Ming, X., 2014. Pre- and perinatal risk factors for autism Pena, C.J., Neugut, Y.D., Champagne, F.A., 2013. Developmental timing of the effects
spectrum disorder in a New Jersey Cohort. J. Child Neurol., e0883073813512899. of maternal care on gene expression and epigenetic regulation of hormone
Marsit, C.J., Maccani, M.A., Padbury, J.F., Lester, B.M., 2012. Placental 11-beta hydrox- receptor levels in female rats. Endocrinology 154, 4340–4351.
ysteroid dehydrogenase methylation is associated with newborn growth and a Petronis, A., 2001. Human morbid genetics revisited: relevance of epigenetics.
measure of neurobehavioral outcome. PLoS ONE 7, e33794. Trends Genet. 17, 142–146.
Martin, N., Boomsma, D., Machin, G., 1997. A twin-pronged attack on complex traits. Petronis, A., 2006. Epigenetics and twins: three variations on the theme. Trends
Nat. Genet. 17, 387–392. Genet. 22, 347–350.
McDonald, C., Murray, R.M., 2000. Early and late environmental risk factors for Posthuma, D., Polderman, T.J., 2013. What have we learned from recent twin studies
schizophrenia. Brain Res. Brain Res. Rev. 31, 130–137. about the etiology of neurodevelopmental disorders? Curr. Opin. Neurol. 26,
McGrath J.J.a.M.R.M., 2011. Environmental Risk Factors for Schizophrenia. 111–121.
Wiley–Blackwell, Oxford. Rai, D., Lee, B.K., Dalman, C., Golding, J., Lewis, G., Magnusson, C., 2013. Parental
McGuffin, P., Katz, R., Rutherford, J., 1991. Nature, nurture and depression: a twin depression, maternal antidepressant use during pregnancy, and risk of autism
study. Psychol. Med. 21, 329–335. spectrum disorders: population based case-control study. BMJ 346, f2059.
Mehta, D., Klengel, T., Conneely, K.N., Smith, A.K., Altmann, A., Pace, T.W., Rex- Rakyan, V.K., Chong, S., Champ, M.E., Cuthbert, P.C., Morgan, H.D., Luu, K.V.,
Haffner, M., Loeschner, A., Gonik, M., Mercer, K.B., Bradley, B., Muller-Myhsok, Whitelaw, E., 2003. Transgenerational inheritance of epigenetic states at the
B., Ressler, K.J., Binder, E.B., 2013. Childhood maltreatment is associated with murine Axin(Fu) allele occurs after maternal and paternal transmission. Proc.
distinct genomic and epigenetic profiles in posttraumatic stress disorder. Proc. Natl. Acad. Sci. U. S. A. 100, 2538–2543.
Natl. Acad. Sci. U. S. A. 110, 8302–8307. Ranade, S.C., Rose, A., Rao, M., Gallego, J., Gressens, P., Mani, S., 2008. Different types
Mehta, D., Newport, D.J., Frishman, G., Kraus, L., Rex-Haffner, M., Ritchie, J.C., Lori, A., of nutritional deficiencies affect different domains of spatial memory function
Knight, B.T., Stagnaro, E., Ruepp, A., Stowe, Z.N., Binder, E.B., 2014. Early predic- checked in a radial arm maze. Neuroscience 152, 859–866.
tive biomarkers for postpartum depression point to a role for estrogen receptor Rapoport, J.L., Giedd, J.N., Gogtay, N., 2012. Neurodevelopmental model of
signaling. Psychol. Med., 1–14. schizophrenia: update 2012. Mol. Psychiatry 17, 1228–1238.
Menger, Y., Bettscheider, M., Murgatroyd, C., Spengler, D., 2010. Sex differences in Ravelli, A.C., van Der Meulen, J.H., Osmond, C., Barker, D.J., Bleker, O.P., 1999. Obesity
brain epigenetics. Epigenomics 2, 807–821. at the age of 50 y in men and women exposed to famine prenatally. Am. J. Clin.
Metivier, R., Gallais, R., Tiffoche, C., Le Peron, C., Jurkowska, R.Z., Carmouche, R.P., Nutr. 70, 811–816.
Ibberson, D., Barath, P., Demay, F., Reid, G., Benes, V., Jeltsch, A., Gannon, F., Sal- Razin, A., Cedar, H., 1993. DNA methylation and embryogenesis. In: Jost, J., Saluz,
bert, G., 2008. Cyclical DNA methylation of a transcriptionally active promoter. H. (Eds.), DNA Methylation: Molecular Biology and Biological Significance.
Nature 452, 45–50. Birkhauser Verlag, Basel/Switzerland, pp. 343–356.
Metivier, R., Penot, G., Hubner, M.R., Reid, G., Brand, H., Kos, M., Gannon, F., 2003. Razin, A., Riggs, A.D., 1980. DNA methylation and gene function. Science 210,
Estrogen receptor-alpha directs ordered, cyclical, and combinatorial recruit- 604–610.
ment of cofactors on a natural target promoter. Cell 115, 751–763. Resnick, O., Morgane, P.J., Hasson, R., Miller, M., 1982. Overt and hidden forms of
Middeldorp, C.M., Birley, A.J., Cath, D.C., Gillespie, N.A., Willemsen, G., Statham, D.J., chronic malnutrition in the rat and their relevance to man. Neurosci. Biobehav.
de Geus, E.J., Andrews, J.G., van Dyck, R., Beem, A.L., Sullivan, P.F., Martin, N.G., Rev. 6, 55–75.
Boomsma, D.I., 2005. Familial clustering of major depression and anxiety dis- Riecher-Rossler, A., Hafner, H., 2000. Gender aspects in schizophrenia: bridging the
orders in Australian and Dutch twins and siblings. Twin Res. Hum. Genet. 8, border between social and biological psychiatry. Acta Psychiatr. Scand. Suppl.,
609–615. 58–62.
Mill, J., Tang, T., Kaminsky, Z., Khare, T., Yazdanpanah, S., Bouchard, L., Jia, P., Rifkin-Graboi, A., Bai, J., Chen, H., Hameed, W.B., Sim, L.W., Tint, M.T., Leutscher-
Assadzadeh, A., Flanagan, J., Schumacher, A., Wang, S.C., Petronis, A., 2008. Broekman, B., Chong, Y.S., Gluckman, P.D., Fortier, M.V., Meaney, M.J., Qiu, A.,
Epigenomic profiling reveals DNA-methylation changes associated with major 2013. Prenatal Maternal Depression Associates with Microstructure of Right
psychosis. Am. J. Hum. Genet. 82, 696–711. Amygdala in Neonates at Birth. Biol Psychiatry.
Mitsushima, D., Masuda, J., Kimura, F., 2003. Sex differences in the stress-induced Riggs, A., Xiong, Z., Wang, L., JM, L., 1998. Methylation dynamics, epigenetic fidelity
release of acetylcholine in the hippocampus and corticosterone from the adrenal and X chromosome structure. In: Wolffe, A. (Ed.), Epigenetics. Novartis Founda-
cortex in rats. Neuroendocrinology 78, 234–240. tion Symposium, vol. 214. John Wiley & Sons, Chistester, pp. 214–227.
Morgan, H.D., Santos, F., Green, K., Dean, W., Reik, W., 2005. Epigenetic reprogramm- Ripke, S., O’Dushlaine, C., Chambert, K., Moran, J.L., Kahler, A.K., Akterin, S., Bergen,
ing in mammals. Hum. Mol. Genet. 14 (Spec no. 1), R47–R58. S.E., Collins, A.L., Crowley, J.J., Fromer, M., Kim, Y., Lee, S.H., Magnusson, P.K.,
Morgan, H.D., Sutherland, H.G., Martin, D.I., Whitelaw, E., 1999. Epigenetic inheri- Sanchez, N., Stahl, E.A., Williams, S., Wray, N.R., Xia, K., Bettella, F., Borglum,
tance at the agouti locus in the mouse. Nat. Genet. 23, 314–318. A.D., Bulik-Sullivan, B.K., Cormican, P., Craddock, N., de Leeuw, C., Durmishi, N.,
Mummaneni, P., Yates, P., Simpson, J., Rose, J., Turker, M.S., 1998. The primary func- Gill, M., Golimbet, V., Hamshere, M.L., Holmans, P., Hougaard, D.M., Kendler,
tion of a redundant Sp1 binding site in the mouse aprt gene promoter is to block K.S., Lin, K., Morris, D.W., Mors, O., Mortensen, P.B., Neale, B.M., O’Neill, F.A.,
epigenetic gene inactivation. Nucleic Acids Res. 26, 5163–5169. Owen, M.J., Milovancevic, M.P., Posthuma, D., Powell, J., Richards, A.L., Riley,
Murgatroyd, C., Patchev, A.V., Wu, Y., Micale, V., Bockmuhl, Y., Fischer, D., Holsboer, B.P., Ruderfer, D., Rujescu, D., Sigurdsson, E., Silagadze, T., Smit, A.B., Stefans-
F., Wotjak, C.T., Almeida, O.F., Spengler, D., 2009. Dynamic DNA methyla- son, H., Steinberg, S., Suvisaari, J., Tosato, S., Verhage, M., Walters, J.T., Levinson,
tion programs persistent adverse effects of early-life stress. Nat. Neurosci. 12, D.F., Gejman, P.V., Laurent, C., Mowry, B.J., O’Donovan, M.C., Pulver, A.E., Schwab,
1559–1566. S.G., Wildenauer, D.B., Dudbridge, F., Shi, J., Albus, M., Alexander, M., Campion,
Narayanan, N.S., Guarnieri, D.J., DiLeone, R.J., 2010. Metabolic hormones, dopamine D., Cohen, D., Dikeos, D., Duan, J., Eichhammer, P., Godard, S., Hansen, M., Lerer,
circuits, and feeding. Front. Neuroendocrinol. 31, 104–112. F.B., Liang, K.Y., Maier, W., Mallet, J., Nertney, D.A., Nestadt, G., Norton, N., Papa-
Nielsen, P.R., Mortensen, P.B., Dalman, C., Henriksen, T.B., Pedersen, M.G., Pedersen, dimitriou, G.N., Ribble, R., Sanders, A.R., Silverman, J.M., Walsh, D., Williams,
C.B., Agerbo, E., 2013. Fetal growth and schizophrenia: a nested case-control and N.M., Wormley, B., Arranz, M.J., Bakker, S., Bender, S., Bramon, E., Collier, D.,
case-sibling study. Schizophr. Bull. 39, 1337–1342. Crespo-Facorro, B., Hall, J., Iyegbe, C., Jablensky, A., Kahn, R.S., Kalaydjieva, L.,
Nierop, A., Bratsikas, A., Zimmermann, R., Ehlert, U., 2006. Are stress-induced cortisol Lawrie, S., Lewis, C.M., Linszen, D.H., Mata, I., McIntosh, A., Murray, R.M., Ophoff,
changes during pregnancy associated with postpartum depressive symptoms? R.A., Van Os, J., Walshe, M., Weisbrod, M., Wiersma, D., Donnelly, P., Barroso,
Psychosom. Med. 68, 931–937. I., Blackwell, J.M., Brown, M.A., Casas, J.P., Corvin, A.P., Deloukas, P., Duncanson,
Nolen-Hoeksema, S., Girgus, J.S., 1994. The emergence of gender differences in A., Jankowski, J., Markus, H.S., Mathew, C.G., Palmer, C.N., Plomin, R., Rautanen,
depression during adolescence. Psychol. Bull. 115, 424–443. A., Sawcer, S.J., Trembath, R.C., Viswanathan, A.C., Wood, N.W., Spencer, C.C.,
North, C.R., Wild, T.C., Zwaigenbaum, L., Colman, I., 2013. Early neurodevelopment Band, G., Bellenguez, C., Freeman, C., Hellenthal, G., Giannoulatou, E., Pirinen,
and self-reported adolescent symptoms of depression and anxiety in a National M., Pearson, R.D., Strange, A., Su, Z., Vukcevic, D., Langford, C., Hunt, S.E., Edkins,
Canadian Cohort Study. PLOS ONE 8, e56804. S., Gwilliam, R., Blackburn, H., Bumpstead, S.J., Dronov, S., Gillman, M., Gray, E.,
66 J. Guintivano, Z.A. Kaminsky / Neuroscience Research 102 (2016) 56–66

Hammond, N., Jayakumar, A., McCann, O.T., Liddle, J., Potter, S.C., Ravindrara- Tarantino, L.M., Sullivan, P.F., Meltzer-Brody, S., 2011. Using animal models to
jah, R., Ricketts, M., Tashakkori-Ghanbaria, A., Waller, M.J., Weston, P., Widaa, disentangle the role of genetic, epigenetic, and environmental influences on
S., Whittaker, P., McCarthy, M.I., Stefansson, K., Scolnick, E., Purcell, S., McCar- behavioral outcomes associated with maternal anxiety and depression. Front
roll, S.A., Sklar, P., Hultman, C.M., Sullivan, P.F., 2013. Genome-wide association Psychiatry 2, 44.
analysis identifies 13 new risk loci for schizophrenia. Nat. Genet. 45, 1150–1159. Taylor, A., Glover, V., Marks, M., Kammerer, M., 2009. Diurnal pattern of cortisol
Rive, M.M., van Rooijen, G., Veltman, D.J., Phillips, M.L., Schene, A.H., Ruhe, H.G., output in postnatal depression. Psychoneuroendocrinology 34, 1184–1188.
2013. Neural correlates of dysfunctional emotion regulation in major depressive ter Horst, G.J., 2010. Estrogen in the limbic system. Vitam. Horm. 82,
disorder. A systematic review of neuroimaging studies. Neurosci. Biobehav. Rev. 319–338.
37 (10 Pt 2), 2529–2553. Torgersen, S., 1983. Genetic factors in anxiety disorders. Arch. Gen. Psychiatry 40,
Romano-Torres, M., Fernandez-Guasti, A., 2010. Estradiol valerate elicits 1085–1089.
antidepressant-like effects in middle-aged female rats under chronic mild Torgersen, S., 1986. Genetic factors in moderately severe and mild affective disor-
stress. Behav. Pharmacol. 21, 104–111. ders. Arch. Gen. Psychiatry 43, 222–226.
Santos, F., Dean, W., 2004. Epigenetic reprogramming during early development in Tsigos, C., Chrousos, G.P., 2002. Hypothalamic-pituitary-adrenal axis, neuroen-
mammals. Reproduction 127, 643–651. docrine factors and stress. J. Psychosom. Res. 53, 865–871.
Santos, F., Peters, A.H., Otte, A.P., Reik, W., Dean, W., 2005. Dynamic chromatin Tycko, B., 2010. Allele-specific DNA methylation: beyond imprinting. Hum. Mol.
modifications characterise the first cell cycle in mouse embryos. Dev. Biol. 280, Genet. 19, R210–R220.
225–236. Vucetic, Z., Kimmel, J., Totoki, K., Hollenbeck, E., Reyes, T.M., 2010. Maternal high-
Schalkwyk, L.C., Meaburn, E.L., Smith, R., Dempster, E.L., Jeffries, A.R., Davies, M.N., fat diet alters methylation and gene expression of dopamine and opioid-related
Plomin, R., Mill, J., 2010. Allelic skewing of DNA methylation is widespread across genes. Endocrinology 151, 4756–4764.
the genome. Am. J. Hum. Genet. 86, 196–212. Wade, T.J., Cairney, J., Pevalin, D.J., 2002. Emergence of gender differences in depres-
Schwartz, S., Ast, G., 2010. Chromatin density and splicing destiny: on the cross-talk sion during adolescence: national panel results from three countries. J. Am. Acad.
between chromatin structure and splicing. EMBO J. 29 (10), 1629–1636. Child Adolesc. Psychiatry 41, 190–198.
Seeman, M.V., 1997. Psychopathology in women and men: focus on female hor- Walf, A.A., Koonce, C.J., Frye, C.A., 2008. Estradiol or diarylpropionitrile decrease
mones. Am. J. Psychiatry 154, 1641–1647. anxiety-like behavior of wildtype, but not estrogen receptor beta knockout,
Shansky, R.M., Lipps, J., 2013. Stress-induced cognitive dysfunction: hormone- mice. Behav. Neurosci. 122, 974–981.
neurotransmitter interactions in the prefrontal cortex. Front. Hum. Neurosci. Wang, L., Xu, R.J., 2007. The effects of perinatal protein malnutrition on spatial learn-
7, 123. ing and memory behaviour and brain-derived neurotrophic factor concentration
Sigurdsson, E., Fombonne, E., Sayal, K., Checkley, S., 1999. Neurodevelopmental in the brain tissue in young rats. Asia Pac. J. Clin. Nutr. 16 (Suppl. 1), 467–472.
antecedents of early-onset bipolar affective disorder. Br. J. Psychiatry 174, Weaver, I.C., Cervoni, N., Champagne, F.A., D’Alessio, A.C., Sharma, S., Seckl, J.R.,
121–127. Dymov, S., Szyf, M., Meaney, M.J., 2004. Epigenetic programming by maternal
Simerly, R.B., 2002. Wired for reproduction: organization and development of sex- behavior. Nat. Neurosci. 7, 847–854.
ually dimorphic circuits in the mammalian forebrain. Annu. Rev. Neurosci. 25, Weissman, M.M., Olfson, M., 1995. Depression in women: implications for health
507–536. care research. Science 269, 799–801.
Simerly, R.B., 2005. Wired on hormones: endocrine regulation of hypothalamic Westberry, J.M., Trout, A.L., Wilson, M.E., 2010. Epigenetic regulation of estrogen
development. Curr. Opin. Neurobiol. 15, 81–85. receptor alpha gene expression in the mouse cortex during early postnatal
Skalkidou, A., Hellgren, C., Comasco, E., Sylven, S., Sundstrom Poromaa, I., 2012. development. Endocrinology 151, 731–740.
Biological aspects of postpartum depression. Womens Health (Lond Engl) 8, Westberry, J.M., Wilson, M.E., 2012. Regulation of estrogen receptor alpha gene
659–672. expression in the mouse prefrontal cortex during early postnatal development.
Sorensen, H.J., Nielsen, P.R., Pedersen, C.B., Benros, M.E., Nordentoft, M., Mortensen, Neurogenetics 13, 159–167.
P.B., 2014. Population impact of familial and environmental risk factors for Whiteford, H.A., Degenhardt, L., Rehm, J., Baxter, A.J., Ferrari, A.J., Erskine, H.E., Charl-
schizophrenia: a nationwide study. Schizophr. Res. 153 (1-3), 214–219. son, F.J., Norman, R.E., Flaxman, A.D., Johns, N., Burstein, R., Murray, C.J., Vos, T.,
Stachowiak, M.K., Kucinski, A., Curl, R., Syposs, C., Yang, Y., Narla, S., Terranova, C., 2013. Global burden of disease attributable to mental and substance use dis-
Prokop, D., Klejbor, I., Bencherif, M., Birkaya, B., Corso, T., Parikh, A., Tzanakakis, orders: findings from the Global Burden of Disease Study 2010. Lancet 382,
E.S., Wersinger, S., Stachowiak, E.K., 2013. Schizophrenia: a neurodevelopmental 1575–1586.
disorder–integrative genomic hypothesis and therapeutic implications from a Wong, A.H., Gottesman, I.I., Petronis, A., 2005. Phenotypic differences in genet-
transgenic mouse model. Schizophr. Res. 143, 367–376. ically identical organisms: the epigenetic perspective. Hum. Mol. Genet. 14
Stankiewicz, A.M., Swiergiel, A.H., Lisowski, P., 2013. Epigenetics of stress adapta- (Spec no. 1), R11–R18.
tions in the brain. Brain Res. Bull. 98C, 76–92. Yap, D.B., Chu, J., Berg, T., Schapira, M., Cheng, S.W., Moradian, A., Morin, R.D.,
Studd, J.W., 2011. A guide to the treatment of depression in women by estrogens. Mungall, A.J., Meissner, B., Boyle, M., Marquez, V.E., Marra, M.A., Gascoyne, R.D.,
Climacteric 14 (6), 637–642. Humphries, R.K., Arrowsmith, C.H., Morin, G.B., Aparicio, S.A., 2010. Somatic
Suda, S., Segi-Nishida, E., Newton, S.S., Duman, R.S., 2008. A postpartum model in rat: mutations at EZH2 Y641 act dominantly through a mechanism of selectively
behavioral and gene expression changes induced by ovarian steroid deprivation. altered PRC2 catalytic activity, to increase H3K27 trimethylation. Blood 117 (8),
Biol. Psychiatry 64, 311–319. 2451–2459.
Sullivan, P.F., Daly, M.J., O’Donovan, M., 2012. Genetic architectures of psychi- Yim, I.S., Glynn, L.M., Dunkel-Schetter, C., Hobel, C.J., Chicz-DeMet, A., Sandman, C.A.,
atric disorders: the emerging picture and its implications. Nat. Rev. Genet. 13, 2009. Risk of postpartum depressive symptoms with elevated corticotropin-
537–551. releasing hormone in human pregnancy. Arch. Gen. Psychiatry 66, 162–169.
Sullivan, P.F., Neale, M.C., Kendler, K.S., 2000. Genetic epidemiology of major depres- Zhou, L., Fester, L., Haghshenas, S., de Vrese, X., von Hacht, R., Gloger, S., Brandt,
sion: review and meta-analysis. Am. J. Psychiatry 157, 1552–1562. N., Bader, M., Vollmer, G., Rune, G.M., 2014. Oestradiol-induced synapse for-
Tamashiro, K.L., Moran, T.H., 2010. Perinatal environment and its influences on mation in the female hippocampus: roles of oestrogen receptor subtypes. J.
metabolic programming of offspring. Physiol. Behav. 100, 560–566. Neuroendocrinol. 26, 439–447.

You might also like