ChapterPDF AvailableLiterature Review

Anxiety and Epigenetics

Authors:

Abstract

Anxiety disorders are highly prevalent psychiatric disorders often comorbid with depression and substance abuse. Twin studies have shown that anxiety disorders are moderately heritable. Yet, genome-wide association studies (GWASs) have failed to identify gene(s) significantly associated with diagnosis suggesting a strong role for environmental factors and the epigenome. A number of anxiety disorder subtypes are considered " stress related. " A large focus of research has been on the epigenetic and anxiety-like behavioral consequences of stress. Animal models of anxiety-related disorders have provided strong evidence for the role of stress on the epigenetic control of the hypothalamic-pituitary-adrenal (HPA) axis and of stress-responsive brain regions. Neuroepigenetics may continue to explain individual variation in susceptibility to environmental perturbations and consequently anxious behavior. Behavioral and pharmacological interventions aimed at targeting epigenetic marks associated with anxiety may prove fruitful in developing treatments.
145
© Springer International Publishing AG 2017
R. Delgado-Morales (ed.), Neuroepigenomics in Aging and Disease, Advances
in Experimental Medicine and Biology, DOI 10.1007/978-3-319-53889-1_8
Anxiety and Epigenetics
Andrew A. Bartlett, Rumani Singh, and Richard G. Hunter
Abstract
Anxiety disorders are highly prevalent psychiatric disorders often comorbid with
depression and substance abuse. Twin studies have shown that anxiety disorders
are moderately heritable. Yet, genome-wide association studies (GWASs) have
failed to identify gene(s) significantly associated with diagnosis suggesting a
strong role for environmental factors and the epigenome. A number of anxiety
disorder subtypes are considered “stress related.” A large focus of research has
been on the epigenetic and anxiety-like behavioral consequences of stress.
Animal models of anxiety-related disorders have provided strong evidence for
the role of stress on the epigenetic control of the hypothalamic-pituitary-adrenal
(HPA) axis and of stress-responsive brain regions. Neuroepigenetics may con-
tinue to explain individual variation in susceptibility to environmental perturba-
tions and consequently anxious behavior. Behavioral and pharmacological
interventions aimed at targeting epigenetic marks associated with anxiety may
prove fruitful in developing treatments.
Keywords
Anxiety • Epigenetic • Stress • Glucocorticoid • Plasticity • Hippocampus
• Amygdala • Prefrontal cortex • Histone • DNA methylation • Noncoding RNA
A.A. Bartlett • R. Singh, Ph.D. • R.G. Hunter, Ph.D. (*)
Department of Psychology, University of Massachusetts,
100 Morrissey Blvd, Boston, MA 02125, USA
e-mail: Andrew.Bartlett001@umb.edu; Rumani.Singh@umb.edu; Richard.Hunter@umb.edu
8
146
8.1 Introduction
Anxiety disorders (ADs) are among the most common psychiatric disorders, occur-
ring in roughly a third of the US population. They are also highly comorbid with
depression and substance abuse disorders, and the pathogenesis of AD is likely
highly interrelated [1]. While anxiety disorders are heritable and genetic factors
play a role in anxiety disorders, most of the risk of these disorders is environmental
in nature [2]. Stress, particularly in early life, substance abuse, circadian, and micro-
biota have all been shown to have an influence on risk of anxiety disorders [36].
Further, it is likely that anxious phenotypes are influenced by more subtle factors
such as the interplay between an anxious parent and a child whose early life is
defined in part by adapting to that parent’s behavior [7, 8]. Indeed the latter case is
emblematic of one of the important distinctions between heritability, which can
include epigenetic mechanisms, both behavioral and molecular, and the strictly
genetic inheritance with which heritability, in general, is often conflated. ADs are
moderately heritable with most of the disorders in the classification showing herita-
bility in the range of 30% [9]. GWASs have generally not met the criterion of
genome-wide significance, and candidate gene approaches have also been relatively
unsuccessful [10]. However some genetic polymorphisms do show replicable asso-
ciations with AD, for example, the glucocorticoid receptor chaperone FKBP5 has
been associated with risk of post-traumatic stress disorder (PTSD) in individuals
with a history of child abuse in an African–American sample, and the same sample
also demonstrated a female-specific association with PTSD and the ADCYPAP1R1
receptor for the neuropeptide PACAP [11, 12]. The catechol-O-methyltransferase
(COMT) valine158methionine polymorphism has been repeatedly implicated in
risk of panic disorder, though with different alleles imparting risk in European ver-
sus Asian populations [13]. Even these findings point to the role of other contextual
factors like ancestry and sex as influences on the underlying genetics, developmen-
tal context also appears to influence the expression of genetic risk, as a study in a
Swedish cohort has shown that different risk factors act at different times across
adolescence and early adulthood [14].
While environmental factors like stress are clearly significant in many anxiety
disorders, their effects can vary wildly across individuals. The role of the environ-
ment is most clear with PTSD, where most individuals are resilient and only a frac-
tion go on to develop the disorder after a trauma exposure [15, 16]. The question of
differential susceptibility in AD is another to which genetic explanations thus far
fall short.
8.2 The Neuroanatomy of Anxiety Disorders
Human anxiety is defined by emotional symptoms as well as behavioral and physi-
ological phenotypes. Much of the work in understanding the underlying neuroanat-
omy involved in anxiety-related pathology has been done using animal models.
Specifically, the focus has been on conserved endocrine systems and brain regions
A.A. Bartlett et al.
147
that identify or respond to environmental threats. For example, noxious stimuli may
result in freezing behavior, sympathetic nervous system activation, and subsequent
endocrine response in both the rat and the human. Across species, the limbic system
and the prefrontal cortex appear to be crucial for regulating threat recognition and
response. The hormonal response to threat appears, likewise, remarkably similar
and feature highly conserved signaling pathways.
Within the limbic system are a number of structures necessary for threat response
and assessment. The amygdala, for instance, appears to be necessary for fear
responses. Patients with Urbach-Wiethe disease have compromised amygdala func-
tion and report loss of feelings of fear [17]. In rodents, fear conditioning models pair
a benign stimulus, the conditioned stimulus (CS), with a noxious stimulus, the
unconditioned stimulus (US). A frequently used example of a US is a foot shock
which elicits freezing behavior, an unconditioned response (UR). After pairing of
the CS with the US, the CS alone can elicit this freezing behavior. This freezing is
referred to as the conditioned response (CR). Lesioning the amygdala has been
shown to obliterate freezing behavior, the CR, in conditioned rats [18, 19].
Stimulation of the amygdala during CS presentation produces subsequent freezing
behavior to the CS without US pairing [20]. During encoding of fear memories,
hippocampal inputs to the amygdala appear to be necessary for CS-US pairing for
contextual clues [21]. Within the amygdala, various subnuclei have been shown to
regulate different processes. The central amygdala (CeA) appears to regulate CR
expression through projections to the periaqueductal gray (PAG) [22]. The lateral
amygdala (LA) appears to receive CS and US inputs through cortical and thalamic
innervations [23]. The stimulation of a subpopulation of neurons in the LA, when
paired with presentation of the CS, appears to be sufficient to generate a CR. The
basal amygdala (BA) appears to have a dual role in both CR expression and suppres-
sion [24]. Two distinct populations of neurons were identified in the BA, one inner-
vated by the hippocampus and the other innervated by the PFC [24, 25]. During
extinction of the CS-US pairing, the PFC appears to inhibit the BA and attenuate
freezing CR [2628]. The pairing of the CS-US improves prediction of the US
allowing for rapid behavioral response. However, when the CS fails to correctly
predict the US, the association must not continue to persist else anxiety or avoid-
ance for the US has now generalized to benign stimuli. These regions are critical to
avoidant and anxiety-like behavior. The dysregulation of these circuits may lead to
recurrent avoidance or anxious behavior to inappropriate stimuli similar to the defi-
nition of human anxiety.
8.3 The Neuroendocrine Axis in Anxiety Disorders
The HPA axis is a critical component of the acute stress response. In response to a
stressor, the body must divert resources appropriately in order to efficiently address
the challenge at hand. In part, this tentative balance is achieved through the activa-
tion of the HPA axis. The HPA axis is a negative feedback loop that begins with the
release of arginine-vasopressin (AVP) and corticotrophin-releasing factor (CRF)
8 Anxiety and Epigenetics
148
into the pituitary portal from the paraventricular nucleus (PVN) in the hypothala-
mus. This release promotes the production of proopiomelanocortin (POMC) in the
pituitary. POMC is subsequently converted to adrenocorticotropic hormone (ACTH)
and released into the bloodstream. The adrenal gland produces corticosteroids in the
adrenal cortex as a consequence of ACTH. Corticosteroids are released into the
blood and bind to the mineralocorticoid (MR) and glucocorticoid receptor (GR). In
the PVN, pituitary and hippocampus GRs inhibit the production of CRF resulting in
negative feedback loop.
The adrenal gland also produces two other hormones, epinephrine and norepi-
nephrine, from the adrenal medulla in response to ACTH. These hormones do not
engage in a self-regulating negative feedback loop but are indirectly regulated
through the actions of GRs. The role of these hormones is to control the response of
the body and the peripheral nervous system, for instance, reducing digestion and
immune function while increasing heart rate and blood pressure acutely. Interestingly,
pharmacological interventions targeting norepinephrine receptors have proved
effective reducing phobias during fear memory reconsolidation [29]. These findings
suggest that the autonomic nervous system may remain a potential area for research
and intervention in stress-related anxiety disorders such as phobias and PTSD.
8.4 Epigenetic Factors
The relative prominence of the environment and the moderate contribution of
genetic factors to the pathogenesis of anxiety disorders have made the study of these
disorders through the lens of epigenetics a fruitful avenue of research in recent
years. Many molecular epigenetic mechanisms have now been implicated in AD,
including DNA and histone modification as well as noncoding RNA (ncRNA).
Epigenetics, in the strict molecular sense, refers to regulation of DNA sequences
that does not involve alteration of actual base composition. Transcription and other
genomic functions are regulated directly through epigenetic modifications that typi-
cally annotate DNA and its associated histones via acetylation, methylation, and
phosphorylation. These epigenetic marks are tightly linked to chromatin state as
complex of DNA, RNA, and protein. Open chromatin is associated with active tran-
scription, whereas closed chromatin is associated with transcriptional silencing.
Epigenetic marks that define the epigenotype include DNA methylation and various
modifications (e.g., methylation, acetylation) of histone proteins that are complexed
with DNA. DNA methylation occurs at cytosines of CpG dinucleotides and is cata-
lyzed by enzymes of the DNA methyltransferase family. DNA methylation may
inhibit gene expression by direct interaction with factors that repress transcription
or, indirectly, through recruitment of methyl-CpG binding proteins (MeCP2 and
MBDs) complexed with enzymes that modify histone proteins. These modifications
can transform chromatin from an active to a repressed state, or vice versa.
The role of the epigenome in etiology of anxiety disorders and variations in
behavior and neurological status can now be investigated. Of particular importance
in epigenetics research is the fact that epigenetic marks are modifiable both in the
A.A. Bartlett et al.
149
germ line and in somatic tissues by genetic, environmental, and stochastic factors.
Each cell in the human body possesses not only a genotype, identical in all somatic
cells of an organism, but also an epigenotype that is highly variable among the dif-
ferent tissues of an individual. Errors or alterations in epigenotype can occur as
primary stochastic events or secondarily in response to either genetic mutations (e.g.
transposition events) or environmental exposures. Therefore a discussion of poten-
tial epigenetic etiologies of anxiety disorders necessarily involves both genetic and
environmental factors. Dysregulation of genes that control epigenetic mechanisms
leads to a number of “epigenetic syndromes” falling into two groups. Those with
changes in genes regulating epigenetic marks include enzymes such as DNA meth-
yltransferases, methyl-binding proteins, and enzymes that affect histone modifica-
tion. The second category involves genes that are regulated by epigenetic marks, for
example, imprinted genes.
8.5 Epigenetics in Animal Models of Anxiety
Twin studies of generalized anxiety disorder have failed to identify either a genetic
basis for or strongly heritable component of the disorder [30]. This class of mental
health disorders is often comorbid with addiction [31]. Both involve pathological
behaviors that have a neurobiological basis. Over the last decade, increasing focus
has been placed on how gene-environment interactions mediated by epigenetic
molecular mechanisms might improve our understanding of the disease. Though
environmental influences including trauma and substance abuse are known con-
tributors to anxiety, it is difficult or impossible at present to examine molecular
epigenetic changes in the central nervous system of clinical populations, and given
the tissue-specific nature of epigenetic mechanisms, accessible peripheral tissues
such as blood or epithelial cells may not reflect the changes present in the brain. For
these reasons, animal models have been employed to mimic the signs of anxiety.
While symptoms, such as intrusive thoughts, are impossible to model in rodents or
nonhuman primates, sophisticated paradigms have been used to model aspects of
social anxiety, general anxiety, and more broadly anxious temperament. In rodent
models, common behavioral paradigms to assess anxiety-like behaviors include the
elevated plus maze (EPM), light/dark box (LD), open field test (OFT), social defeat
(SD), and the social interaction test (SIT). The EPM consists of two arms of open
platforms and two arms of closed platforms featuring three walls. The EPM is based
on an innate fear of heights and open spaces such that rodents prefer the closed
platforms to the open platforms. After quickly equilibrating to the testing arena, less
anxious rodents will explore and spend increasing time on the open platforms. The
LD box consists of two connected chambers, one illuminated while the other is not.
The natural preference of the rodent is the dark chamber; however, given time less
anxious rodents again will explore and spend increasing time in the light chamber.
The OFT is a square testing arena with four walls. In novel settings, rodents prefer
to remain unexposed to predators, in this case, close to the wall. After exposure, less
anxious animals will cross the arena exploring and spend increasingly more time in
8 Anxiety and Epigenetics
150
the center. SD paradigms vary to some extent but primarily involve repeated expo-
sure of a rodent to another dominating rodent. The exposed rodents display
depressive- like symptoms but also social avoidance. Social avoidance is most com-
monly measured using SIT. SIT is conducted in a two-chamber arena separated by
a wall to prevent contact but allow for other sensory exchanges, i.e., visual cues,
odor, and ultrasonic vocalizations. More anxious, socially avoidant rodents will
spend less time in the area closest to the neighboring chamber after habituation.
These consist of the major testing paradigms used to proximate anxiety in the rodent
and have allowed for a more comprehensive understanding of the neuroepigenetic
regulation of anxiolytic behavior.
Natural variation in susceptibility to clinical anxiety has been subject to increased
scrutiny in recent years. Early animal work suggested that gene-environment inter-
actions likely mediated anxiety outcomes as SD paradigms among other stressors
produced anxious phenotypes. Notably, an early study showed that susceptibility to
SD, as measured by reduced interaction time in the SIT, was correlated with DNA
methylation of CpG islands in the promoter of the CRH gene in paraventricular
nucleus (PVN) [32]. Natural variation in maternal care during the first week of life
was shown to differentially pattern the methylation of nr3c1 promoter of offspring,
modification that persisted into adulthood and corresponded to reduced glucocorti-
coid receptor expression and enhanced HPA axis activation to an acute stressor [33].
These offspring were later characterized as displaying differential anxiety-like
behaviors as a consequence of maternal care received as measured by the EPM and
OFT [34, 35]. In adult mice, voluntary exercise has been demonstrated to increase
nr3c1 expression while reducing miRNA-124, known to inhibit nr3c1, expression
[36]. Though in contradiction to other findings, voluntary exercise decreased time
in the open arms of the EPM suggesting an increasingly anxious phenotype.
Recently, long noncoding RNA (lncRNA) expression of gomafu in the prefrontal
cortex (PFC) has been shown to regulate time spent in the center of the OFT and
grooming time to suggest that expression of this lncRNA is necessary for reducing
anxiety-like behaviors [37]. Likewise, loss-of-function l3mbtl1, null mice show
reduced latency to enter the light chamber in the LD box and increased time spent
in the center of the OFT [38]. As l3mbtl1 codes for a methylated lysine domain
histone-binding protein, a so-called chromatin reader, this suggests that histone
lysine methylation is required for regulating anxiety-like behavior. In this vein, tlr4
null mice did not show increased synaptic enrichment of NR1 following in the short
term following repeated ethanol exposure nor increased GluR1 enrichment in the
long term in the mPFC compared to similarly treated wild-type controls. These tlr4
mice failed to show mPFC enrichment of acetylated-H4 at the promoter of fosB and
BDNF in response to ethanol exposure. This observation suggests that tlr4 is neces-
sary for histone H4 acetylation at fosB and BDNF following ethanol exposure and
appears to be necessary for ethanol-induced increases in anxiety-like behavior as
indicated by time spent in the open arms of the EPM [39]. In contrast, others have
shown that acute ethanol exposure reduces amygdalar miRNA-494 subsequently
increasing Cited2, CBP, and p300 expression. These changes were associated with
increased H3 acetylation in the central amygdala and anxiolysis [40].
A.A. Bartlett et al.
151
8.6 Transgenerational Epigenetics
Transgenerational epigenetic can be either direct inheritance of mRNAs, protein, or
DNA modification via the germline or indirect “inheritance” such that the feed-
forward phenotypic profile of the parent can lead to changes in either noncoding
RNA expression, histone modification, or DNA methylation. Indirect inheritance
was shown by Weaver et al. (2004) where cross-fostering experiments suggested
that maternal care alone determined GR 1-7 promoter methylation in offspring hip-
pocampi [33]. Morgan and Bale (2011), in a case of direct inheritance, showed that
prenatal stress can lead to alterations in stress sensitivity and miRNA expression in
the brains of male offspring [41]. These effects persist for several generations sug-
gesting direct inheritance of paternal miRNAs or DNA methylation via sperm.
Transgenerational effects have been consistently observed in the offspring of
Holocaust survivors [4244]. Maternal PTSD of these survivors has been predictive
of offspring PTSD risk and increased corticosteroid sensitivity. In specific impor-
tance to this chapter, offspring of Holocaust survivors were found to be at a far
greater risk of developing an anxiety disorder compared to control, age-matched
offspring born to Jewish parents [42]. At this date, the number of generations out to
which this inheritance persists and affects offspring of survivors remains unknown.
Transgenerational non-genomic transmission of both maternal behavior and HPA
axis activation in rats was initially demonstrated by Meaney et al. [45]. The same
group showed that glucocorticoid sensitivity and anxiety-like behavior are patterned
by maternal care and can persist out for several generations [33, 35]. The level of
maternal care during the first week of life patterned the methylation of the GR 1-7
promoter and subsequently GR expression in the hippocampus. These phenotypes
can be reversed however by cross-fostering offspring of low-licking and grooming
dams with high-licking and grooming dams. In anxious adults of low-licking and
grooming dams, the phenotype can be reversed by supplication of an HDAC inhibi-
tor to the hippocampus [34, 35]. Conversely, in low-anxiety adults of high-licking
and grooming dams, the phenotype can be reversed by infusion of a methyl donor
to the hippocampus [35]. Interestingly, maternal care has also been shown to affect
peripheral oxytocin receptor (OXTR) methylation status in rats [46]. A recent clini-
cal study also found that peripheral OXTR methylation was associated with increased
frequency of anxiety and depression [47]. Genome-wide methylation analysis in
infants of mothers with depression and/or anxiety revealed a number of CpG islands
to be differentially methylated [48]. Similarly, increased methylation of the BDNF
gene in blood of adults has been linked to lower maternal care and interpersonal
violence-related PTSD [49, 50]. In addition, poor maternal care and anxiety has
been linked to risk of diabetes and metabolic syndrome in bonnet macaque off-
spring [51, 52]. In high- and low-anxiety bred rats, increased H3K9me3 accumula-
tion was found at both the GR and FGF2 promoters in the hippocampus [53]. This
group also found differences in DNA methylation of the FGF2 promoter in the
hippocampus between high- and low-anxiety rats. High-anxiety rats had reduced
DNA methylation and methyl-binding protein association at the FGF2 promoter,
which presumably was permissive for increased FGF2 expression [53]. This group
8 Anxiety and Epigenetics
152
also showed that FGF2 increases H3K9me3 association with both the GR promoter
and its own. This demonstrates a potential mechanism by which early-life perturba-
tions independent of maternal care can contribute to anxiety-like behavior across
generations.
8.7 Neuroepigenetic Effects of Early Stress on Anxious
Behaviors
Early-life stress has been demonstrated repeatedly to pattern stress reactivity and
anxious behavior. These changes persist beyond the time frame of the initial stressor
and often long into adulthood. The prenatal effects of stress lead to dysregulation of
the HPA axis associated mainly with changes GR expression [35]. Though these
findings were first reported in animal studies. Recently, these findings have been
recapitulated in longitudinal human studies. For instance, maternal prenatal anxiety
has been shown to predict internalizing and anxiety scores on the child behavior
checklist in the infant [49]. Further, differences in global DNA methylation were
observed at a number of CpG sites in neonatal cord blood of mothers affected by
anxiety during gestation [48]. Likewise, maternal PTSD has been shown to associ-
ate with both increased glucocorticoid sensitivity in the offspring of Holocaust sur-
vivors and increased offspring diagnosed with anxiety disorders [42]. Maternal
PTSD has also been demonstrated to be predictive of offspring PTSD and presum-
ably through inherited stress reactivity [43, 44]. These findings suggest that both the
prenatal environment and stress/trauma history may recruit epigenetic processes in
the intergenerational transmission of HPA axis dysregulation and anxiogenic conse-
quences. However, consideration of allostatic load must be of concern as severe and
mild stress have opposing roles on physiology and behavior. Allostatic load is the
cumulative effect of multiple stressors taking into consideration severity, duration,
and ability to cope with stressors [54, 55]. Consider the effects of a severe uncon-
trollable stressor, for example, maternal separation, on stress sensitivity in contrast
to a mild controllable stressor such as voluntary exercise. While maternal separation
sensitizes the HPA axis of the infant, voluntary exercise can promote resiliency to
future stressors [56, 57].
8.7.1 Prenatal Stress
In utero exposure to maternal stress and corticosteroids patterns the HPA axis of
infants ultimately altering synaptic connectivity, function, and behavioral responses
specifically those involved in stress adaptation [5860]. Prenatal restraint stress has
been shown to impair offspring brain function and development reducing HPA axis
feedback and altering neuroplasticity [61]. Prenatal stress and glucocorticoid treat-
ment produce lasting behavioral changes such as spatial learning impairment and
increased anxiety-like behavior [58, 59]. In addition, mild stressors, for instance,
postnatal handling, have been shown to reduce these deficits as well as attenuate
A.A. Bartlett et al.
153
HPA axis sensitivity [58, 59]. Prenatal stress does so by altering synaptic connectiv-
ity, neurogenesis, and chromatin structure in stress-sensitive regions of the brain, for
example, in the PFC where offspring of maternally stressed dams show reduced
dendritic spine complexity and density [60]. Similarly, in both rodent and nonhu-
man primate models, prenatal stress retards hippocampal neurogenesis in the den-
tate gyrus. Prenatal stress has been linked to increased methylation of the GR 1-7
promoter in the hippocampus as well as reduced methylation of the CRF promoter
in the hypothalamus and amygdala of male but not female mice [62]. These sex-
specific changes have been linked to differential expression of DNA methyltransfer-
ase 1 (DNMT1), though the changes responsible for this dichotomized expression
remain unknown. Elliot et al. (2010) first ascribed natural variation in social interac-
tion following social defeat in adults to be due in part to the methylation of the
CRH promoter in the hypothalamus. Mice susceptible to social defeat show
increased social anxiety and reduced CRH promoter methylation in the PVN [32].
The methylation status of the CRF promoter in PVN helps to explain natural vari-
ability in the susceptibility of mice to social defeat and consequently social anxiety.
Prenatal stress had previously been shown to differentially affect CRF release in the
PVN [63]. Interestingly, a subsequent study found that prenatal restraint stress
increased both anxious behavior and corticosterone release in response to stress
while reducing CRF promoter methylation at the same CpG islands noted by Elliot
et al. (2010) [64]. Prenatal restraint stress has also been shown to increase methyla-
tion of the REELIN promoter in the PFC perhaps linking changes in synaptic con-
nectivity observed there to underlying molecular influences [65]. REELIN is an
important neuroplasticity gene, known to be epigenetically regulated by fear condi-
tioning [66]. Similarly, prenatal exposure to maternal depression and anxiety has
been linked to increased NR3C1 1F promoter methylation and increased salivary
cortisol following exposure to a stressor in infants [67, 68]. Maternal anxiety has
been linked to differential methylation of a number of other genes in cord blood
including IGF2 and H19 [69]. In fact, distress during pregnancy has been linked to
placental methylation of a number of stress-related genes including HSD11B2,
NR3C1, and FKBP5 [70]. Other perturbations, including maternal diet and paternal
exposure to drugs of abuse such as cocaine and ethanol, have been shown to alter
cortical gene expression through changes in the epigenetic machinery and affect
anxiolytic behavior in the offspring [7173]. Importantly, mild postnatal stressors
have been shown to reverse the effects of prenatal stress as well as promote resil-
iency [58, 59, 74, 75]. Given the association between maternal stress and anxiety,
these findings provide evidence for the efficacy of behavioral therapy and alike as
an early-life intervention [7, 76].
8.7.2 Early-Life Stress
The vast majority of studies of early-life stress focus on the epigenetic consequences
of the interactions within the mother-infant dyad. Both maternal care and separation
have been demonstrated to both alter HPA axis stress reactivity and adult anxiety
8 Anxiety and Epigenetics
154
behaviors of the infant through lasting changes to the epigenomes [35, 77].
Specifically, maternal separation has been shown to sensitize offspring HPA axis
activation early-life interventions including environmental enrichment attenuate
this effect [78]. Poor rearing conditions have been shown to increase CRF release
from the PVN and amygdala as well as hypermethylate the GR 1-7 promoter in the
hippocampus [33, 79]. Conversely, good maternal care and rearing conditions have
been demonstrated to hypomethylate the GR 1-7 promoter in the hippocampus,
produce efficient stress responses, and reduce anxiety-like behaviors [35, 8082].
The GR 1F promoter is the human ortholog of the rodent 1-7 promoter [83].
Hypermethylation of the 1F promoter in the brains of suicide victims was associated
with childhood abuse [84]. The findings of McGowan et al. (2009) were later
expanded to include the 1-B, 1-C, and 1-H promoters as well [85]. Other groups
have failed to replicate some of these findings, however [86]. The McGowan group
has also shown that hippocampal ribosomal RNA expression is reduced in suicide
victims suggesting reduced hippocampal protein synthesis [87]. Childhood adver-
sity has also been linked to increased 1F promoter methylation in peripheral cells as
well [88, 89]. Methylation patterns as a consequence of childhood abuse over-
whelmingly persist into adulthood [90]. Early postnatal stress followed by subse-
quent adult chronic stress has been linked to reduced hippocampal plasticity and
increased anxiety-like behaviors [91]. Maternal separation has been shown to reduce
amygdalar neurotensin receptor 1 (NTSR1) expression through increased methyla-
tion of the NTSR1 promoter. Microinfusion of NTSR1 receptor agonist increased
conditioned freezing responses, while an agonist reduced this behavior suggesting
an epigenetic molecular mechanism sufficient for increasing anxiety-like behavior
[92]. Similarly, maternal separation has been linked to increased HPA activation to
environmental stressors in adult offspring [93]. More recently, however, this finding
was both replicated and associated with hypomethylation of the POMC, the gene
encoding the precursor for ACTH, in the pituitary [94]. As HPA axis dysregulation
has been associated with anxiety-like outcomes, again these findings suggest a criti-
cal role of these molecular influences as a consequence of stress in the context of
anxiety outcomes. Clinical work has recently shown that early childhood trauma
affects CpG methylation in both the promoter and gene proper of the 5-HT3Ar in
blood [95]. Interestingly, this locus is downstream of GR response element which
showed altered CpG methylation associated with emotional neglect and CpG meth-
ylation associated with anxiety-related behaviors.
Adolescence represents another postnatal life stage sensitive to the epigenetic
effects of stress [96100]. For instance, chronic variable stress during adolescence
reduces hippocampal volume and spatial cognition, these effects persisting into
adulthood [101]. Isolation rearing in adolescent mice reduces the expression of
5-α-reductase I, the rate-limiting enzyme for allopregnanolone, a hormone shown
to reduce depressive- and anxiety-like symptoms in rodents [102, 103]. Isolated
juveniles show increased CpG methylation upstream of the transcription start site
of the SRD5A1 gene, which codes for this enzyme; one of these islands was dem-
onstrated to be sufficient to reduce expression in the PFC [102]. In adolescent
rhesus monkeys, anxious temperament is associated with increased methylation
A.A. Bartlett et al.
155
and reduced expression of the BCL11A and JAG1 genes, associated with neuro-
plasticity, in the amygdala [104]. Similarly, these findings have been supported by
recent clinical work identifying a correlation between NR3C1, the gene coding for
the glucocorticoid receptor, 1F promoter methylation in blood, and internalizing
symptoms [105]. Moreover, these adolescents showing increased 1F promoter
methylation and displaying internalizing behavior also had higher concentrations
of cortisol upon waking. These findings, in tandem, indicate a significant role of
neuroplasticity and HPA axis regulation in stress-sensitive regions of the brain,
notably the hippocampus, amygdala, PVN, and PFC, during adolescence and may
underscore potential individual variations that contribute to anxious susceptibility.
These epigenetic predispositions may be compounded by other environmental per-
turbations such as exposure to drugs of abuse. Intermittent alcohol exposure, for
instance, has been shown to increase HDAC activity in the rodent amygdala [106].
These changes were also associated with reduced time spent in the open arms of
the EPM and in the light compartment of the dark/light box into adulthood. Further
alcohol-exposed adults had reductions in the number of spines and increased alco-
hol intake. Conversely, acute alcohol exposure during adolescence produces simi-
lar changes in anxiety-like behaviors while decreasing HDAC activity in the rodent
amygdala [107]. In summation both predisposition and environmental perturbation
may work in synchrony during adolescence to dysregulate both transcription and
synaptic integrity in the amygdala and ultimately help shape entrain anxious
behavior.
8.8 Stress in Adulthood
Stress induces lasting changes in heterochromatin structure, ultimately changing
neuronal plasticity and behavior. The hippocampus, PFC, and amygdala are targets
of glucocorticoids. As these regions help regulate spatial memory, executive func-
tion, and fear responses, respectively, they are of the utmost importance in the con-
text of anxiety. These regions are extremely sensitive to both acute and chronic
stressors and express a large number of epigenetic enzymes and display profound
structural changes at the synaptic level in response to environmental stressors.
Stressors often produce some type of learning, the spatial and contextual compo-
nents of which are presumed to be coded by the hippocampus and the cue-based
components coded by the amygdalar [24, 108]. The reconsolidation and extinction
of these associations are mediated by the PFC. Dysregulation of these memories
may fail to attenuate improper responses to environmental stimuli, much like the
symptoms of anxiety. Fear conditioning is widely used to study learning and neuro-
plastic consequences thereof as well as to model symptoms of a number of anxiety
disorders as well as other stress-related disorders such as post-traumatic stress dis-
order [109, 110]. Epigenetics has been thought to be a potential basis of memory on
the molecular level [111113]. Initially, Sweatt et al. (2004) first demonstrated the
role of hippocampal histone acetylation during fear memory formation [114]. Miller
and Sweatt (2007) later showed that fear conditioning upregulated expression of
8 Anxiety and Epigenetics
156
hippocampal DNMT3A and 3B, and that DNMT activity there was required for fear
memory consolidation [66]. Hippocampal methylation of reelin, PP1, and BDNF
was also changed by fear conditioning [115]. Interestingly, reelin and BDNF have
well-established roles in dendritic remodeling, and PP1 codes for a phosphatase that
acts at histone H3S10 [116, 117]. Presumably these are the grounds for its role in
memory as the dual acetylation-phosphorylation H3 mark was enriched at the BDNF
locus in the hippocampus. Others have found similarly that both histone modifica-
tion and DNA methylation play critical roles in the amygdala in memory reconsoli-
dation and consolidation, respectively [118]. Tsai et al. (2007) established that both
environmental enrichment and HDAC inhibition were sufficient for restoring defi-
cits in memory and synaptic connectivity in a mouse model of neuronal cell loss
[119]. A later study by the same group (2009) identified HDAC2 to be necessary for
the negative impacts on memory [120]. Recall of recent memories results HDAC2
dissociation from the chromatin, which causes increases in H3 acetylation and
increased expression of immediate early genes [121]. Recall of less recent memo-
ries do not produce such profound changes in HDAC activity. Yet, HDAC inhibition
during reconsolidation of remote fear memories allows for H3 acetylation, increased
immediate early gene expression, and neuroplastic changes [121]. This suggests
that epigenetic control of chromatin structure regulates neuroplastic changes under-
pinning behavioral outputs related to fear memories.
Social defeat represents another type of stress-based learning producing an anx-
ious phenotype in the defeated. Social defeat is a well-characterized animal model
of a number of psychiatric disorders including modeling symptoms of depression
and anxiety [122]. The Nestler group was early in demonstrating that social defeat
affects hippocampal chromatin signatures [123]. They showed that chronic social
defeat increased H3K27me3 repression of the BDNF promoter in the hippocampus.
Also, the accumulation of this repressive mark was mitigated by antidepressant
treatment, inhibiting HDAC2, resulting in increases in H3 acetylation and H3K4
methylation, both marks promoting transcription [123, 124]. The same group also
showed that chronic stress or cocaine exposure altered HDAC activity in the nucleus
accumbens [125]. DNMT3A expression increases as a consequence of chronic
defeat and decreases as a consequence of chronic cocaine which were associated
with synaptic changes as well in the same nuclei [126]. Interestingly, natural varia-
tion to susceptibility to social defeat has been associated with distinct methylation
signatures of the CRF promoter in the PVN [32]. Resilient animals also show
increased H3K9me3 and K3K27me3 in the nucleus accumbens [127, 128]. The
levels of accumbal H3K9me3 also change in response to cocaine exposure as well
as dendritic morphology [129]. Acute stress and chronic antidepressant treatment
have also been shown to increase H3K9me3 levels in the hippocampus [130]. This
repressive mark appears to accumulate selectively at repetitive elements, specifi-
cally retrotransposons (for review see Lapp & Hunter, 2016) in the genome [131,
132]. Interestingly, Alu and LINE1 retrotransposons appear upregulated in PTSD
veterans compared to combat deployed controls [133]. Socially defeated animals
also show increased basal corticosterone in circulation, reduced time spent in open
arms of the EPM and in the light component of the light/dark box, as well as reduced
A.A. Bartlett et al.
157
hippocampal H3 acetylation and increased HDAC5 expression [134]. These defi-
cits, however, were rescued by a moderate, involuntary exercise regiment, a mild
stressor [134]. Voluntary exercise, a mild and controllable stressor, alone has been
shown to have anxiolytic effects in addition to reducing hippocampal expression of
the histone H2 variant H2A.z and increasing expression of mitochondrial- related
genes TFAM and NDUFA6 in the same region [135]. These recent findings hark
back to the importance of allostasis and suggest an epigenetic underpinning of anx-
ious behaviors. Further, it has been suggested that stress opens up “windows of
epigenetic plasticity” that are unique to the stressor and elicit dynamic effects based
on previous stress history [136, 137]. The recent work of the McEwen laboratory
has provided strong evidence for this nuanced view of the epigenetic effects of
stress. While chronic restraint stress resulted in reduced time spend in the light
component of the light/dark box, only a novel acute stressor led to persistent reduc-
tion in time spend in the light compartment. These differences corresponded to
changes in hippocampal long-term potentiation and NMDA receptor expression
[136, 137]. Acute restraint stress exposure has also been shown to convert DNA
methylation through the addition of a hydroxyl group of NR3C1 promoter in the
hippocampus [138]. More recently, hyper-hydroxymethylation has been observed
in regions associated with neuronal plasticity following acute restraint stress in the
hippocampus [139].
8.9 Prospects for an Epigenetic Pharmacology of Anxiety
Epigenetic interventions have proven effective in animal models of anxiety and
stress, and some psychiatric drugs, such as the mood stabilizer valproate, have
known epigenetic effects (valproate is an HDAC inhibitor). Thus, it would appear
that the prospects for epigenetic therapies for anxiety disorders are fairly high.
Most pharmacologic studies of drugs with epigenetic activities have focused on
histone acetylation, with the HDACs being the major targets. In fear extinction
models, which have substantial relevance to human AD, a variety of HDAC inhibi-
tors have been shown to be effective in enhancing extinction [140]. Similarly,
HDAC inhibition reversed the group differences in maternal behavior and adult
stress reactivity observed by Weaver in his landmark paper on epigenetic program-
ming of maternal behavior [33]. Similarly, the same phenotype and associated anx-
ious behavior could be reversed with central infusion of the methyl donor
S-adenosyl-methionine (SAMe) in adult animals [34, 35]. A number of studies have
found SAMe to be more effective than placebo in the treatment of depression,
though other well-designed trials have had negative results [141, 142]. A recent
Cochrane collaboration review concluded that there was not strong evidence for the
efficacy of SAMe in depression but that further research was warranted [143].
Studies of the efficacy of SAMe in the treatment of anxiety symptoms, however, are
very limited to date. DNA methyltransferase inhibitors such as zebularine,
N-phthaloyl-l-tryptophan and 5-aza-deoxycytidine have been shown to interfere
with fear memory formation in preclinical models [66, 118]. To date, little clinical
8 Anxiety and Epigenetics
158
work has been done with this class of drugs, likely due to concerns about side
effects, which are significant for some of these agents.
The study of epigenetic drug targets for anxiety remains in its infancy, and many
questions remain to be adequately researched. One such question is whether these
agents actually offer superior outcomes to existing treatments. Another is whether
they might be used in combination with both other drugs and behavioral interven-
tions to additive or even synergistic effect. Nonetheless, molecular epigenetics
offers a novel class of potential drug targets for disorders like AD which have his-
torically had relatively few molecular mechanisms with which to work.
Conclusions
Epigenetic mechanisms play a clear mechanistic role in animal models of anxi-
ety, and human epigenetic studies suggest that these observations are generaliz-
able to clinical populations. Indeed, some effort has already been made to
translate the preclinical findings in the field into the clinic. Nonetheless, signifi-
cant questions, particularly those relating to the time course and nature of epi-
genetic changes in humans, remain to be answered. Beyond the borders of what
might now be regarded as “classical” epigenetics, novel molecular mechanisms
of epigenomic, genomic, and epitranscriptomic plasticity are being revealed in
the brain in behavioral contexts relevant to anxiety disorders. Transposons,
which are mobile elements of the genome, have been shown to be regulated by
stress exposure in both humans and animal models [131, 144, 145]. The mito-
chondria, which contains its own genome, shows transcriptional regulation in
response to stress, and its function in the nucleus accumbens has been linked to
anxiety phenotypes and social subordination in mice [146148]. Even more
intriguingly, covalent modification of RNA in the prefrontal cortex, the methyla-
tion of adenosine, has been shown to associate with the development of fear
memory in mice [149]. This epitranscriptomic effect points to yet another layer
of molecular complexity that will need to be incorporated into our models of
anxiety, both normal and pathologic in model systems and in the clinic.
While neuroepigenetics is a relatively young science, it is already clear that it
has relevance to our understanding of AD. Indeed, it has begun to produce usable
translational findings for the treatment of disorders, like depression, which are
highly comorbid with numerous anxiety disorders. There is ample reason to
believe that neuroepigenetic mechanisms will continue to be a fruitful area of
research into the biology of anxiety and AD.
References
1. Kessler RC, Berglund P, Demler O, Jin R, Merikangas KR, Walters EE. Lifetime prevalence
and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey
Replication. Arch Gen Psychiatry. 2005;62:593–602.
2. Smoller JW. The genetics of stress-related disorders: PTSD, depression, and anxiety disorders.
Neuropsychopharmacology. 2016;41:297–319.
A.A. Bartlett et al.
159
3. Hunter RG, McEwen BS. Stress and anxiety across the lifespan: structural plasticity and
epigenetic regulation. Epigenomics. 2013;5:177–94.
4. Lai HMX, Cleary M, Sitharthan T, Hunt GE. Prevalence of comorbid substance use, anxiety
and mood disorders in epidemiological surveys, 1990–2014: a systematic review and meta-
analysis. Drug Alcohol Depend. 2015;154:1–13.
5. Coles ME, Schubert JR, Nota JA. Sleep, circadian rhythms, and anxious traits. Curr Psychiatry
Rep. 2015;17:73.
6. Mayer EA, Knight R, Mazmanian SK, Cryan JF, Tillisch K. Gut microbes and the brain:
paradigm shift in neuroscience. J Neurosci. 2014;34:15490–6.
7. DiCorcia JA, Tronick E. Quotidian resilience: exploring mechanisms that drive resilience
from a perspective of everyday stress and coping. Neurosci Biobehav Rev.
2011;35:1593–602.
8. Tronick E, Hunter RG. Waddington, dynamic systems, and epigenetics. Front Behav
Neurosci. 2016;10:107.
9. Hettema JM, Neale MC, Kendler KS. A review and meta-analysis of the genetic epidemiol-
ogy of anxiety disorders. Am J Psychiatry. 2001;158:1568–78.
10. Perez JA, Otowa T, Roberson-Nay R, Hettema JM. In: Charney DS, Nestler EJ, Sklar P,
Buxbaum JD, editors. Genetics of anxiety disorders in neurobiology of mental illness.
New York: OUP; 2013.
11. Binder EB, Bradley RG, Liu W, Epstein MP, Deveau TC, Mercer KB, et al. Association of
FKBP5 polymorphisms and childhood abuse with risk of posttraumatic stress disorder symp-
toms in adults. JAMA. 2008;299:1291–305.
12. Ressler KJ, Mercer KB, Bradley B, Jovanovic T, Mahan A, Kerley K, et al. Post-traumatic
stress disorder is associated with PACAP and the PAC1 receptor. Nature. 2011;470:492–7.
13. Domschke K, Deckert J, O’donovan MC, Glatt SJ. Meta-analysis of COMT val158met in
panic disorder: ethnic heterogeneity and gender specificity. Am J Med Genet B Neuropsychiatr
Genet. 2007;144B:667–73.
14. Kendler KS, Gardner CO, Annas P, Neale MC, Eaves LJ, Lichtenstein P. A longitudinal twin
study of fears from middle childhood to early adulthood: evidence for a developmentally
dynamic genome. Arch Gen Psychiatry. 2008;65:421–9.
15. Feder A, Nestler EJ, Charney DS. Psychobiology and molecular genetics of resilience. Nat
Rev Neurosci. 2009;10:446–57.
16. Griffiths BB, Hunter RG. Neuroepigenetics of stress. Neuroscience. 2014;275:420–35.
17. Feinstein JS, Adolphs R, Damasio A, Tranel D. The human amygdala and the induction and
experience of fear. Curr Biol. 2011;21:34–8.
18. Blanchard DC, Blanchard RJ. Innate and conditioned reactions to threat in rats with amygda-
loid lesions. J Comp Physiol Psychol. 1972;81:281–90.
19. Kim JJ, Rison RA, Fanselow MS. Effects of amygdala, hippocampus, and periaqueductal
gray lesions on short- and long-term contextual fear. Behav Neurosci. 1993;107:1093–8.
20. Weingarten H, White N. Exploration evoked by electrical stimulation of the amygdala of rats.
Physiol Psychol. 2013;6:229–35.
21. Maren S, Fanselow MS. Synaptic plasticity in the basolateral amygdala induced by hippo-
campal formation stimulation in vivo. J Neurosci. 1995;15:7548–64.
22. LeDoux JE, Iwata J, Cicchetti P, Reis DJ. Different projections of the central amygdaloid
nucleus mediate autonomic and behavioral correlates of conditioned fear. J Neurosci.
1988;8:2517–29.
23. Campese VD, Kim J, Lázaro-Muñoz G, Pena L, LeDoux JE, Cain CK. Lesions of lateral or
central amygdala abolish aversive Pavlovian-to-instrumental transfer in rats. Front Behav
Neurosci. 2014;8:161.
24. Herry C, Ciocchi S, Senn V, Demmou L, Müller C, Lüthi A. Switching on and off fear by
distinct neuronal circuits. Nature. 2008;454:600–6.
25. Ciocchi S, Herry C, Grenier F, Wolff SBE, Letzkus JJ, Vlachos I, et al. Encoding of condi-
tioned fear in central amygdala inhibitory circuits. Nature. 2010;468:277–82.
8 Anxiety and Epigenetics
160
26. Milad MR, Quirk GJ. Neurons in medial prefrontal cortex signal memory for fear extinction.
Nature. 2002;420:70–4.
27. Milad MR, Vidal-Gonzalez I, Quirk GJ. Electrical stimulation of medial prefrontal cortex
reduces conditioned fear in a temporally specific manner. Behav Neurosci.
2004;118:389–94.
28. Quirk GJ, Likhtik E, Pelletier JG, Paré D. Stimulation of medial prefrontal cortex decreases
the responsiveness of central amygdala output neurons. J Neurosci. 2003;23:8800–7.
29. Soeter M, Kindt M. An abrupt transformation of phobic behavior after a post-retrieval amne-
sic agent. Biol Psychiatry. 2015;78:880–6.
30. Mackintosh M-A, Gatz M, Wetherell JL, Pedersen NL. A twin study of lifetime generalized
anxiety disorder (GAD) in older adults: genetic and environmental influences shared by neu-
roticism and GAD. Twin Res Hum Genet. 2006;9:30–7.
31. Lüthi A, Lüscher C. Pathological circuit function underlying addiction and anxiety disorders.
Nat Neurosci. 2014;17:1635–43.
32. Elliott E, Ezra-Nevo G, Regev L, Neufeld-Cohen A, Chen A. Resilience to social stress coin-
cides with functional DNA methylation of the Crf gene in adult mice. Nat Neurosci.
2010;13:1351–3.
33. Weaver ICG, Cervoni N, Champagne FA, D’Alessio AC, Sharma S, Seckl JR, et al. Epigenetic
programming by maternal behavior. Nat Neurosci. 2004;7:847–54.
34. Weaver ICG, Champagne FA, Brown SE, Dymov S, Sharma S, Meaney MJ, et al. Reversal of
maternal programming of stress responses in adult offspring through methyl supplementa-
tion: altering epigenetic marking later in life. J Neurosci. 2005;25:11045–54.
35. Weaver ICG, Meaney MJ, Szyf M. Maternal care effects on the hippocampal transcriptome
and anxiety-mediated behaviors in the offspring that are reversible in adulthood. Proc Natl
Acad Sci U S A. 2006;103:3480–5.
36. Pan-Vazquez A, Rye N, Ameri M, McSparron B, Smallwood G, Bickerdyke J, et al. Impact
of voluntary exercise and housing conditions on hippocampal glucocorticoid receptor, miR-
124 and anxiety. Mol Brain. 2015;8:40.
37. Spadaro PA, Flavell CR, Widagdo J, Ratnu VS, Troup M, Ragan C, et al. Long noncoding
RNA-directed epigenetic regulation of gene expression is associated with anxiety-like behav-
ior in mice. Biol Psychiatry. 2015;78:848–59.
38. Shen EY, Jiang Y, Mao W, Futai K, Hock H, Akbarian S. Cognition and mood-related behav-
iors in L3mbtl1 null mutant mice. PLoS One. 2015;10:e0121252.
39. Montesinos J, Pascual M, Rodríguez-Arias M, Miñarro J, Guerri C. Involvement of TLR4 in
the long-term epigenetic changes, rewarding and anxiety effects induced by intermittent etha-
nol treatment in adolescence. Brain Behav Immun. 2016;53:159–71.
40. Teppen TL, Krishnan HR, Zhang H, Sakharkar AJ, Pandey SC. The potential role of amyg-
daloid microRNA-494 in alcohol-induced anxiolysis. Biol Psychiatry. 2016;80:711–9.
41. Morgan CP, Bale TL. Early prenatal stress epigenetically programs dysmasculinization in
second-generation offspring via the paternal lineage. J Neurosci. 2011;31:11748–55.
42. Lehrner A, Bierer LM, Passarelli V, Pratchett LC, Flory JD, Bader HN, et al. Maternal PTSD
associates with greater glucocorticoid sensitivity in offspring of Holocaust survivors.
Psychoneuroendocrinology. 2014;40:213–20.
43. Yehuda R, Bierer LM. Transgenerational transmission of cortisol and PTSD risk. Prog Brain
Res. 2008;167:121–35.
44. Yehuda R, Bell A, Bierer LM, Schmeidler J. Maternal, not paternal, PTSD is related to
increased risk for PTSD in offspring of Holocaust survivors. J Psychiatr Res.
2008;42:1104–11.
45. Francis D, Diorio J, Liu D, Meaney MJ. Nongenomic transmission across generations of
maternal behavior and stress responses in the rat. Science. 1999;286:1155–8.
46. Beery AK, McEwen LM, MacIsaac JL, Francis DD, Kobor MS. Natural variation in maternal
care and cross-tissue patterns of oxytocin receptor gene methylation in rats. Horm Behav.
2016;77:42–52.
A.A. Bartlett et al.
161
47. Chagnon YC, Potvin O, Hudon C, Préville M. DNA methylation and single nucleotide vari-
ants in the brain-derived neurotrophic factor (BDNF) and oxytocin receptor (OXTR) genes
are associated with anxiety/depression in older women. Front Genet. 2015;6:230.
48. Non AL, Binder AM, Kubzansky LD, Michels KB. Genome-wide DNA methylation in neo-
nates exposed to maternal depression, anxiety, or SSRI medication during pregnancy.
Epigenetics. 2014;9:964–72.
49. Sharp H, Hill J, Hellier J, Pickles A. Maternal antenatal anxiety, postnatal stroking and emo-
tional problems in children: outcomes predicted from pre- and postnatal programming
hypotheses. Psychol Med. 2015;45:269–83.
50. Moser DA, Paoloni-Giacobino A, Stenz L, Adouan W, Manini A, Suardi F, et al. BDNF meth-
ylation and maternal brain activity in a violence-related sample. PLoS One. 2015;10:e0143427.
51. Kaufman D, Smith ELP, Gohil BC, Banerji M, Coplan JD, Kral JG, et al. Early appearance
of the metabolic syndrome in socially reared bonnet macaques. J Clin Endocrinol Metab.
2005;90:404–8.
52. Kaufman D, Banerji MA, Shorman I, Smith ELP, Coplan JD, Rosenblum LA, et al. Early-life
stress and the development of obesity and insulin resistance in juvenile bonnet macaques.
Diabetes. 2007;56:1382–6.
53. Chaudhury S, Aurbach EL, Sharma V, Blandino P, Turner CA, Watson SJ, et al. FGF2 is a
target and a trigger of epigenetic mechanisms associated with differences in emotionality:
partnership with H3K9me3. Proc Natl Acad Sci U S A. 2014;111:11834–9.
54. McEwen BS, Stellar E. Stress and the individual. Mechanisms leading to disease. Arch Intern
Med. 1993;153:2093–101.
55. McEwen BS. Allostasis and allostatic load: implications for neuropsychopharmacology.
Neuropsychopharmacology. 2000;22:108–24.
56. Francis DD, Meaney MJ. Maternal care and the development of stress responses. Curr Opin
Neurobiol. 1999;9:128–34.
57. Wosiski-Kuhn M, Stranahan AM. Opposing effects of positive and negative stress on hippo-
campal plasticity over the lifespan. Ageing Res Rev. 2012;11:399–403.
58. Vallée M, MacCari S, Dellu F, Simon H, Le Moal M, Mayo W. Long-term effects of prenatal
stress and postnatal handling on age-related glucocorticoid secretion and cognitive perfor-
mance: a longitudinal study in the rat. Eur J Neurosci. 1999;11:2906–16.
59. Vallée M, Mayo W, Dellu F, Le Moal M, Simon H, Maccari S. Prenatal stress induces high
anxiety and postnatal handling induces low anxiety in adult offspring: correlation with stress-
induced corticosterone secretion. J Neurosci. 1997;17:2626–36.
60. Murmu MS, Salomon S, Biala Y, Weinstock M, Braun K, Bock J. Changes of spine density
and dendritic complexity in the prefrontal cortex in offspring of mothers exposed to stress
during pregnancy. Eur J Neurosci. 2006;24:1477–87.
61. Maccari S, Morley-Fletcher S. Effects of prenatal restraint stress on the hypothalamus-
pituitary- adrenal axis and related behavioural and neurobiological alterations.
Psychoneuroendocrinology. 2007;32(Suppl 1):S10–5.
62. Mueller BR, Bale TL. Sex-specific programming of offspring emotionality after stress early
in pregnancy. J Neurosci. 2008;28:9055–65.
63. Fujioka T, Sakata Y, Yamaguchi K, Shibasaki T, Kato H, Nakamura S. The effects of prenatal
stress on the development of hypothalamic paraventricular neurons in fetal rats. Neuroscience.
1999;92:1079–88.
64. Xu L, Sun Y, Gao L, Cai Y-Y, Shi S-X. Prenatal restraint stress is associated with demethyl-
ation of corticotrophin releasing hormone (CRH) promoter and enhances CRH transcrip-
tional responses to stress in adolescent rats. Neurochem Res. 2014;39:1193–8.
65. Palacios-García I, Lara-Vásquez A, Montiel JF, Díaz-Véliz GF, Sepúlveda H, Utreras E, et al.
Prenatal stress down-regulates Reelin expression by methylation of its promoter and induces
adult behavioral impairments in rats. PLoS One. 2015;10:e0117680.
66. Miller CA, Sweatt JD. Covalent modification of DNA regulates memory formation. Neuron.
2007;53:857–69.
8 Anxiety and Epigenetics
162
67. Oberlander TF, Weinberg J, Papsdorf M, Grunau R, Misri S, Devlin AM. Prenatal exposure
to maternal depression, neonatal methylation of human glucocorticoid receptor gene
(NR3C1) and infant cortisol stress responses. Epigenetics. 2008;3:97–106.
68. Hompes T, Izzi B, Gellens E, Morreels M, Fieuws S, Pexsters A, et al. Investigating the influ-
ence of maternal cortisol and emotional state during pregnancy on the DNA methylation
status of the glucocorticoid receptor gene (NR3C1) promoter region in cord blood. J Psychiatr
Res. 2013;47:880–91.
69. Mansell T, Novakovic B, Meyer B, Rzehak P, Vuillermin P, Ponsonby A-L, et al. The effects
of maternal anxiety during pregnancy on IGF2/H19 methylation in cord blood. Transl
Psychiatry. 2016;6:e765.
70. Monk C, Feng T, Lee S, Krupska I, Champagne FA, Tycko B. Distress during pregnancy:
epigenetic regulation of placenta glucocorticoid-related genes and fetal neurobehavior. Am
J Psychiatry. 2016;173:705–13.
71. Tyagi E, Zhuang Y, Agrawal R, Ying Z, Gomez-Pinilla F. Interactive actions of Bdnf meth-
ylation and cell metabolism for building neural resilience under the influence of diet.
Neurobiol Dis. 2015;73:307–18.
72. Vassoler FM, White SL, Schmidt HD, Sadri-Vakili G, Pierce RC. Epigenetic inheritance of a
cocaine-resistance phenotype. Nat Neurosci. 2013;16:42–7.
73. Liang F, Diao L, Liu J, Jiang N, Zhang J, Wang H, et al. Paternal ethanol exposure and behav-
ioral abnormities in offspring: associated alterations in imprinted gene methylation.
Neuropharmacology. 2014;81:126–33.
74. Lemaire V, Lamarque S, Le Moal M, Piazza P-V, Abrous DN. Postnatal stimulation of the
pups counteracts prenatal stress-induced deficits in hippocampal neurogenesis. Biol
Psychiatry. 2006;59:786–92.
75. Fujioka T, Fujioka A, Tan N, Chowdhury GM, Mouri H, Sakata Y, et al. Mild prenatal stress
enhances learning performance in the non-adopted rat offspring. Neuroscience.
2001;103:301–7.
76. Brand SR, Brennan PA. Impact of antenatal and postpartum maternal mental illness: how are
the children? Clin Obstet Gynecol. 2009;52:441–55.
77. Kember RL, Dempster EL, Lee THA, Schalkwyk LC, Mill J, Fernandes C. Maternal separa-
tion is associated with strain-specific responses to stress and epigenetic alterations to Nr3c1,
Avp, and Nr4a1 in mouse. Brain Behav. 2012;2:455–67.
78. Francis DD, Diorio J, Plotsky PM, Meaney MJ. Environmental enrichment reverses the
effects of maternal separation on stress reactivity. J Neurosci. 2002;22:7840–3.
79. Plotsky PM, Thrivikraman KV, Nemeroff CB, Caldji C, Sharma S, Meaney MJ. Long-term
consequences of neonatal rearing on central corticotropin-releasing factor systems in adult
male rat offspring. Neuropsychopharmacology. 2005;30:2192–204.
80. Caldji C, Diorio J, Meaney MJ. Variations in maternal care in infancy regulate the develop-
ment of stress reactivity. Biol Psychiatry. 2000;48:1164–74.
81. Tang AC, Reeb-Sutherland BC, Yang Z, Romeo RD, McEwen BS. Neonatal novelty-induced
persistent enhancement in offspring spatial memory and the modulatory role of maternal self-
stress regulation. J Neurosci. 2011;31:5348–52.
82. Akers KG, Yang Z, DelVecchio DP, Reeb BC, Romeo RD, McEwen BS, et al. Social com-
petitiveness and plasticity of neuroendocrine function in old age: influence of neonatal nov-
elty exposure and maternal care reliability. PLoS One. 2008;3:e2840.
83. Turner JD, Muller CP. Structure of the glucocorticoid receptor (NR3C1) gene 5 untranslated
region: identification, and tissue distribution of multiple new human exon 1. J Mol Endocrinol.
2005;35:283–92.
84. McGowan PO, Sasaki A, D’Alessio AC, Dymov S, Labonté B, Szyf M, et al. Epigenetic
regulation of the glucocorticoid receptor in human brain associates with childhood abuse. Nat
Neurosci. 2009;12:342–8.
85. Labonte B, Yerko V, Gross J, Mechawar N, Meaney MJ, Szyf M, et al. Differential glucocor-
ticoid receptor exon 1(B), 1(C), and 1(H) expression and methylation in suicide completers
with a history of childhood abuse. Biol Psychiatry. 2012;72:41–8.
A.A. Bartlett et al.
163
86. Alt SR, Turner JD, Klok MD, Meijer OC, Lakke EAJF, Derijk RH, et al. Differential expres-
sion of glucocorticoid receptor transcripts in major depressive disorder is not epigenetically
programmed. Psychoneuroendocrinology. 2010;35:544–56.
87. McGowan PO, Sasaki A, Huang TCT, Unterberger A, Suderman M, Ernst C, et al. Promoter-
wide hypermethylation of the ribosomal RNA gene promoter in the suicide brain. PLoS One.
2008;3:e2085.
88. Tyrka AR, Price LH, Marsit C, Walters OC, Carpenter LL. Childhood adversity and epigen-
etic modulation of the leukocyte glucocorticoid receptor: preliminary findings in healthy
adults. PLoS One. 2012;7:e30148.
89. Romens SE, McDonald J, Svaren J, Pollak SD. Associations between early life stress and
gene methylation in children. Child Dev. 2015;86:303–9.
90. Suderman M, Borghol N, Pappas JJ, Pinto Pereira SM, Pembrey M, Hertzman C, et al.
Childhood abuse is associated with methylation of multiple loci in adult DNA. BMC Med
Genet. 2014;7:13.
91. Eiland L, Ramroop J, Hill MN, Manley J, McEwen BS. Chronic juvenile stress produces
corticolimbic dendritic architectural remodeling and modulates emotional behavior in male
and female rats. Psychoneuroendocrinology. 2012;37:39–47.
92. Toda H, Boku S, Nakagawa S, Inoue T, Kato A, Takamura N, et al. Maternal separation
enhances conditioned fear and decreases the mRNA levels of the neurotensin receptor 1 gene
with hypermethylation of this gene in the rat amygdala. PLoS One. 2014;9:e97421.
93. Pesonen A-K, Räikkönen K. The lifespan consequences of early life stress. Physiol Behav.
2012;106:722–7.
94. Wu Y, Patchev AV, Daniel G, Almeida OFX, Spengler D. Early-life stress reduces DNA
methylation of the Pomc gene in male mice. Endocrinology. 2014;155:1751–62.
95. Perroud N, Zewdie S, Stenz L, Adouan W, Bavamian S, Prada P, et al. Methylation of sero-
tonin receptor 3a in ADHD, borderline personality, and bipolar disorders: link with severity
of the disorders and childhood maltreatment. Depress Anxiety. 2016;33:45–55.
96. McGorry PD, Purcell R, Goldstone S, Amminger GP. Age of onset and timing of treatment
for mental and substance use disorders: implications for preventive intervention strategies
and models of care. Curr Opin Psychiatry. 2011;24:301–6.
97. Blakemore S-J. Development of the social brain during adolescence. Q J Exp Psychol (Hove).
2008;61:40–9.
98. Blakemore S-J. Development of the social brain in adolescence. J R Soc Med.
2012;105:111–6.
99. Gunnar MR, Wewerka S, Frenn K, Long JD, Griggs C. Developmental changes in hypothala-
mus–pituitary–adrenal activity over the transition to adolescence: normative changes and
associations with puberty. Dev Psychopathol. 2009;21:69–85.
100. Romeo RD, Bellani R, Karatsoreos IN, Chhua N, Vernov M, Conrad CD, et al. Stress history
and pubertal development interact to shape hypothalamic-pituitary-adrenal axis plasticity.
Endocrinology. 2006;147:1664–74.
101. Isgor C, Kabbaj M, Akil H, Watson SJ. Delayed effects of chronic variable stress during
peripubertal-juvenile period on hippocampal morphology and on cognitive and stress axis
functions in rats. Hippocampus. 2004;14:636–48.
102. Araki R, Nishida S, Hiraki Y, Matsumoto K, Yabe T. DNA methylation of the GC box in the
promoter region mediates isolation rearing-induced suppression of srd5a1 transcription in the
prefrontal cortex. Neurosci Lett. 2015;606:135–9.
103. Reddy DS, Kulkarni SK. Differential anxiolytic effects of neurosteroids in the mirrored
chamber behavior test in mice. Brain Res. 1997;752:61–71.
104. Alisch RS, Chopra P, Fox AS, Chen K, White ATJ, Roseboom PH, et al. Differentially meth-
ylated plasticity genes in the amygdala of young primates are linked to anxious temperament,
an at risk phenotype for anxiety and depressive disorders. J Neurosci. 2014;34:15548–56.
105. Dadds MR, Moul C, Hawes DJ, Mendoza Diaz A, Brennan J. Individual differences in child-
hood behavior disorders associated with epigenetic modulation of the cortisol receptor gene.
Child Dev. 2015;86:1311–20.
8 Anxiety and Epigenetics
164
106. Pandey SC, Sakharkar AJ, Tang L, Zhang H. Potential role of adolescent alcohol exposure-
induced amygdaloid histone modifications in anxiety and alcohol intake during adulthood.
Neurobiol Dis. 2015;82:607–19.
107. Sakharkar AJ, Zhang H, Tang L, Baxstrom K, Shi G, Moonat S, et al. Effects of histone
deacetylase inhibitors on amygdaloid histone acetylation and neuropeptide Y expression: a
role in anxiety-like and alcohol-drinking behaviours. Int J Neuropsychopharmacol.
2014;17:1207–20.
108. Marschner A, Kalisch R, Vervliet B, Vansteenwegen D, Büchel C. Dissociable roles for the
hippocampus and the amygdala in human cued versus context fear conditioning. J Neurosci.
2008;28:9030–6.
109. Zovkic IB, Sweatt JD. Epigenetic mechanisms in learned fear: implications for
PTSD. Neuropsychopharmacology. 2013;38:77–93.
110. Sultan FA, Day JJ. Epigenetic mechanisms in memory and synaptic function. Epigenomics.
2011;3:157–81.
111. Griffith JS, Mahler HR. DNA ticketing theory of memory. Nature. 1969;223:580–2.
112. Crick F. Memory and molecular turnover. Nature. 1984;312:101.
113. Holliday R. Is there an epigenetic component in long-term memory? J Theor Biol.
1999;200:339–41.
114. Levenson JM, O’Riordan KJ, Brown KD, Trinh MA, Molfese DL, Sweatt JD. Regulation of
histone acetylation during memory formation in the hippocampus. J Biol Chem.
2004;279:40545–59.
115. Lubin FD, Roth TL, Sweatt JD. Epigenetic regulation of BDNF gene transcription in the
consolidation of fear memory. J Neurosci. 2008;28:10576–86.
116. Koshibu K, Gräff J, Beullens M, Heitz FD, Berchtold D, Russig H, et al. Protein phosphatase
1 regulates the histone code for long-term memory. J Neurosci. 2009;29:13079–89.
117. Koshibu K, Gräff J, Mansuy IM. Nuclear protein phosphatase-1: an epigenetic regulator of
fear memory and amygdala long-term potentiation. Neuroscience. 2011;173:30–6.
118. Maddox SA, Schafe GE. Epigenetic alterations in the lateral amygdala are required for recon-
solidation of a Pavlovian fear memory. Learn Mem. 2011;18:579–93.
119. Fischer A, Sananbenesi F, Wang X, Dobbin M, Tsai L-H. Recovery of learning and memory
is associated with chromatin remodelling. Nature. 2007;447:178–82.
120. Guan J-S, Haggarty SJ, Giacometti E, Dannenberg J-H, Joseph N, Gao J, et al. HDAC2 nega-
tively regulates memory formation and synaptic plasticity. Nature. 2009;459:55–60.
121. Gräff J, Joseph NF, Horn ME, Samiei A, Meng J, Seo J, et al. Epigenetic priming of memory
updating during reconsolidation to attenuate remote fear memories. Cell. 2014;156:261–76.
122. Nestler EJ, Hyman SE. Animal models of neuropsychiatric disorders. Nat Neurosci.
2010;13:1161–9.
123. Tsankova NM, Berton O, Renthal W, Kumar A, Neve RL, Nestler EJ. Sustained hippocampal
chromatin regulation in a mouse model of depression and antidepressant action. Nat Neurosci.
2006;9:519–25.
124. Tsankova NM, Kumar A, Nestler EJ. Histone modifications at gene promoter regions in rat
hippocampus after acute and chronic electroconvulsive seizures. J Neurosci.
2004;24:5603–10.
125. Renthal W, Maze I, Krishnan V, Covington HE, Xiao G, Kumar A, et al. Histone deacetylase
5 epigenetically controls behavioral adaptations to chronic emotional stimuli. Neuron.
2007;56:517–29.
126. LaPlant Q, Vialou V, Covington HE, Dumitriu D, Feng J, Warren BL, et al. Dnmt3a regulates
emotional behavior and spine plasticity in the nucleus accumbens. Nat Neurosci.
2010;13:1137–43.
127. Wilkinson MB, Xiao G, Kumar A, LaPlant Q, Renthal W, Sikder D, et al. Imipramine treat-
ment and resiliency exhibit similar chromatin regulation in the mouse nucleus accumbens in
depression models. J Neurosci. 2009;29:7820–32.
A.A. Bartlett et al.
165
128. Covington HE, Maze I, Sun H, Bomze HM, DeMaio KD, Wu EY, et al. A role for repressive
histone methylation in cocaine-induced vulnerability to stress. Neuron. 2011;71:656–70.
129. Maze I, Covington HE, Dietz DM, LaPlant Q, Renthal W, Russo SJ, et al. Essential role of
the histone methyltransferase G9a in cocaine-induced plasticity. Science. 2010;
327:213–6.
130. Hunter RG, McCarthy KJ, Milne TA, Pfaff DW, McEwen BS. Regulation of hippocampal H3
histone methylation by acute and chronic stress. Proc Natl Acad Sci U S A.
2009;106:20912–7.
131. Hunter RG, Murakami G, Dewell S, Seligsohn M, Baker ME, Datson NA, et al. Acute stress
and hippocampal histone H3 lysine 9 trimethylation, a retrotransposon silencing response.
Proc Natl Acad Sci U S A. 2012;109:17657–62.
132. Lapp HE, Hunter RG. The dynamic genome: transposons and environmental adaptation in
the nervous system. Epigenomics. 2016;8:237–49.
133. Rusiecki JA, Chen L, Srikantan V, Zhang L, Yan L, Polin ML, et al. DNA methylation in
repetitive elements and post-traumatic stress disorder: a case-control study of US military
service members. Epigenomics. 2012;4:29–40.
134. Patki G, Solanki N, Atrooz F, Ansari A, Allam F, Jannise B, et al. Novel mechanistic insights
into treadmill exercise based rescue of social defeat-induced anxiety-like behavior and mem-
ory impairment in rats. Physiol Behav. 2014;130:135–44.
135. Aguiar AS, Stragier E, da Luz SD, Remor AP, Oliveira PA, Prediger RD, et al. Effects of
exercise on mitochondrial function, neuroplasticity and anxio-depressive behavior of mice.
Neuroscience. 2014;271:56–63.
136. Gray JD, Rubin TG, Hunter RG, McEwen BS. Hippocampal gene expression changes under-
lying stress sensitization and recovery. Mol Psychiatry. 2014;19:1171–8.
137. Nasca C, Zelli D, Bigio B, Piccinin S, Scaccianoce S, Nisticò R, et al. Stress dynamically
regulates behavior and glutamatergic gene expression in hippocampus by opening a window
of epigenetic plasticity. Proc Natl Acad Sci U S A. 2015;112:14960–5.
138. Li S, Papale LA, Kintner DB, Sabat G, Barrett-Wilt GA, Cengiz P, et al. Hippocampal
increase of 5-hmC in the glucocorticoid receptor gene following acute stress. Behav Brain
Res. 2015;286:236–40.
139. Li S, Papale LA, Zhang Q, Madrid A, Chen L, Chopra P, et al. Genome-wide alterations in
hippocampal 5-hydroxymethylcytosine links plasticity genes to acute stress. Neurobiol Dis.
2016;86:99–108.
140. Singewald N, Schmuckermair C, Whittle N, Holmes A, Ressler KJ. Pharmacology of cogni-
tive enhancers for exposure-based therapy of fear, anxiety and trauma-related disorders.
Pharmacol Ther. 2015;149:150–90.
141. De Berardis D, Orsolini L, Serroni N, Girinelli G, Iasevoli F, Tomasetti C, et al. A compre-
hensive review on the efficacy of S-Adenosyl-L-methionine in major depressive disorder.
CNS Neurol Disord Drug Targets. 2016;15:35–44.
142. Mischoulon D, Price LH, Carpenter LL, Tyrka AR, Papakostas GI, Baer L, et al. A double-
blind, randomized, placebo-controlled clinical trial of S-adenosyl-L-methionine (SAMe) ver-
sus escitalopram in major depressive disorder. J Clin Psychiatry. 2014;75:370–6.
143. Galizia I, Oldani L, Macritchie K, Amari E, Dougall D, Jones TN, et al. S-adenosyl methio-
nine (SAMe) for depression in adults. Cochrane Database Syst Rev. 2016;10:CD011286.
144. Hunter RG, Gagnidze K, McEwen BS, Pfaff DW. Stress and the dynamic genome: steroids,
epigenetics, and the transposome. Proc Natl Acad Sci U S A. 2015;112:6828–33.
145. Ponomarev I, Wang S, Zhang L, Harris RA, Mayfield RD. Gene coexpression networks in
human brain identify epigenetic modifications in alcohol dependence. J Neurosci.
2012;32:1884–97.
146. Hunter RG, Seligsohn M, Rubin TG, Griffiths BB, Ozdemir Y, Pfaff DW, et al. Stress and
corticosteroids regulate rat hippocampal mitochondrial DNA gene expression via the gluco-
corticoid receptor. Proc Natl Acad Sci U S A. 2016;113:9099–104.
8 Anxiety and Epigenetics
166
147. Du J, Wang Y, Hunter R, Wei Y, Blumenthal R, Falke C, et al. Dynamic regulation of mito-
chondrial function by glucocorticoids. Proc Natl Acad Sci U S A. 2009;106:3543–8.
148. Hollis F, van der Kooij MA, Zanoletti O, Lozano L, Cantó C, Sandi C. Mitochondrial func-
tion in the brain links anxiety with social subordination. Proc Natl Acad Sci U S A.
2015;112:15486–91.
149. Widagdo J, Zhao Q-Y, Kempen M-J, Tan MC, Ratnu VS, Wei W, et al. Experience-dependent
accumulation of N6-methyladenosine in the prefrontal cortex is associated with memory pro-
cesses in mice. J Neurosci. 2016;36:6771–7.
A.A. Bartlett et al.
... 4 Therefore, it is imperative to implement new therapeutic approaches and innovate the development of anxiolytic medications. Despite many proposed theories explaining the pathogenesis of anxiety, such as neurotransmitter deficits and neuroimmune dysregulation, 15 none have been universally accepted. Another hypothesis suggests that the gut microbiota may be closely associated with anxiety disorders. ...
Article
Full-text available
Purpose The Baihe Dihuang decoction (BDD) is a representative traditional Chinese medicinal formula that has been used to treat anxiety disorders for thousands of years. This study aimed to reveal mechanisms of anxiolytic effects of BDD with multidimensional omics. Methods First, 28-day chronic restraint stress (CRS) was used to create a rat model of anxiety, and the open field test and elevated plus maze were used to assess anxiety-like behavior. Enzyme-linked immunosorbent assay (ELISA), hematoxylin–eosin staining, and immunofluorescence staining were used to evaluate inflammatory response. Besides, 16S rRNA gene sequencing assessed fecal microbiota composition and differential microbiota. Non-targeted metabolomics analysis of feces was performed to determine fecal biomarkers, and targeted metabolomics was used to observe the levels of hippocampus neurotransmitters. Finally, Pearson correlation analysis was used to examine relationships among gut microbiota, fecal metabolites, and neurotransmitters. Results BDD significantly improved anxiety-like behaviors in CRS-induced rats and effectively ameliorated hippocampal neuronal damage and abnormal activation of hippocampal microglia. It also had a profound effect on the diversity of microbiota, as evidenced by significant changes in the abundance of 10 potential microbial biomarkers at the genus level. Additionally, BDD led to significant alterations in 18 fecal metabolites and 12 hippocampal neurotransmitters, with the majority of the metabolites implicated in amino acid metabolism pathways such as D-glutamine and D-glutamate, alanine, arginine and proline, and tryptophan metabolism. Furthermore, Pearson analysis showed a strong link among gut microbiota, metabolites, and neurotransmitters during anxiety and BDD treatment. Conclusion BDD can effectively improve anxiety-like behaviors by regulating the gut–brain axis, including gut microbiota and metabolite modification, suppression of hippocampal neuronal inflammation, and regulation of neurotransmitters.
... Nepovoljni međuljudski odnosi, stil života zasnovan na štetnim navikama, dugotrajna teška psihička stanja, napetost, anksioznost, zabrinutost, stres mogu dovesti do epigenetičkih modifikacija koje će aktivirati ili suprimirati gene i dovesti do pojave oboljenja. Osobe koje su depresivne i koje izvrše suicid kao i osobe koje su zanemarivane, zlostavljane i bile pod psihosocijalnom deprivacijom u detinjstvu imaju u mozgu povišenu metilaciju određenih regija genoma u odnosu na osobe koje su zdrave (18). ...
Article
Epigenetika je oblast medicine koja se intenzivno razvija i saznanja iz ove oblasti se primenjuju u preventivnoj pedijatriji i drugim oblastima medicine. Primena epigenetičkih saznanja u preventivnoj pedijatriji je sve zastupljenija i interesantna sa više aspekata. U određenim okolnostima, s jedne strane uticaj epigenetičkih faktora može da izmeni ekspresiju gena i da dovede do ispoljavanja bolesti, a može svojim uticajem da deluje i preventivno, i da odloži ispoljavanje bolesti, poboljša kvalitet života i utiče na produženje života. Izmenom epigenetičkih faktora može se uticati na to da se pojava oboljenja odloži ili ne ispolji. Kako su epigenetičke promene reverzibilne, i nema primarnog oštećenja DNK sekvence niti promena na hromozomima, postoje velike mogućnosti za osmišljavanje strategije intervencije promenama životnog stila i samim tim primenama tzv. epigenetičke preventivne terapije.
... When suffering from insomnia, tossing and turning in bed are often accompanied by heightened levels of stress and anxiety, which can activate emotion-regulating areas located in the cerebral cortex, such as the amygdala (55). These areas can release neurotransmitters like glutamate, which in turn can activate the hypothalamic-pituitary-adrenal axis and release stress hormones like corticosteroids that affect vagal and sympathetic nerve activity (56). Moreover, the hypothalamus can directly act on the vagal nucleus through the hypothalamic-vagal nucleus pathway, thus increasing its excitability (57). ...
Article
Full-text available
Background The interactions and associations between obstructive sleep apnea (OSA), sleep-related phenotypes (SRPs), and gastroesophageal reflux disease (GERD) are complex, thus it is hard to explore the effect and direction of causalities. Study objectives A bidirectional Mendelian randomization (MR) study was performed to explore causal associations of GERD with OSA and SRPs (including insomnia, morningness, sleep duration, ease of getting up, daytime napping, daytime dozing, and snoring). Methods First, we gathered summary statistics from publicly available databases. Subsequently, we identified single-nucleotide polymorphisms without strong linkage (r² ≤ 0.001) by referencing relevant genome-wide association studies that met genome-wide significance criteria. Our primary analysis relied on inverse variance weighted to estimate the causal relationship. To ensure the validity of our findings, we also conducted several sensitivity analyses. These included MR Pleiotropy RESidual Sum and Outlier to detect and correct for potential pleiotropic effects, MR-Egger to assess directional pleiotropy, and weighted median analysis to further evaluate heterogeneity and pleiotropy. For the initial MR analysis, when causality was indicated by the results, instrumental variables that were significantly linked to the aforementioned confounding factors were removed. We will re-analyze the data after excluding outcome-related single nucleotide polymorphisms to confirm that the results are still consistent with the previous results. Results GERD was found to increase the risk of OSA (OR = 1.53, 95% CI = 1.37–1.70, p = 5.3 × 10⁻¹⁵), insomnia (OR = 1.14, 95% CI = 1.10–1.19, p = 1.3 × 10⁻¹⁰), snoring (OR = 1.09, 95% CI = 1.04–1.13, p = 6.3 × 10⁻⁵) and less sleep duration (OR = 0.94, 95% CI = 0.91–0.97, p = 3.7 × 10⁻⁴). According to the reverse-direction analysis, there is an elevated risk of GERD associated with OSA (OR = 1.07, 95% CI = 1.02–1.12, p = 0.005), insomnia (OR = 1.95, 95% CI = 1.60–2.37, p = 1.92 × 10⁻¹¹) and snoring (OR = 1.74, 95% CI = 1.37–2.21, p = 4.4 × 10⁻⁶). Conclusion Genetic susceptibility to GERD can elevate the likelihood of experiencing insomnia, snoring, and OSA, in addition to diminishing sleep duration. Conversely, a reverse MR analysis indicates that ameliorating any one of insomnia, snoring, or OSA can mitigate the risk of developing GERD.
... The etiology of anxiety disorders is influenced by genetic as well as environmental factors, e.g., stressful live events (Hettema et al., 2001;Faravelli et al., 2012) and interactions between them (G × E, Nugent et al., 2011). One mediator of those G x E interactions is the epigenetic regulation of gene expression (Bartlett et al., 2017). The best studied epigenetic mechanism is DNA methylation (DNAm), the covalent modification of cytosine in a cytosine-guanine dimer (CpG site). ...
Article
Full-text available
A number of case studies describing hypnotherapy in the treatment of anxiety disorder patients have already been published. Only a few randomized controlled trials (RCTs) investigated the efficacy of hypnotherapy but focused mainly on symptoms rather than specific mental disorders. The goal of this study was to investigate whether hypnotherapy (HT) was superior to a waitlist control group (WL) in the reduction of agoraphobia-related symptoms. Further goals were to report the feasibility of hypnotherapy as well as attrition and completion rates and detect (epi-)genetic variables, which might play a role in treatment outcome. This pilot study was based on a monocentric two-armed randomized controlled rater-blind clinical trial that was conducted between 2018 and 2020 with a waitlist control group. A total of 36 patients diagnosed with agoraphobia were randomized to either HT or WL. Patients in HT received individual outpatient treatment with hypnotherapy with 8 to 12 sessions for a period of 3 months. Patients in WL received HT after 3 months. Agoraphobia-related symptoms were assessed at baseline, after the treatment, and 3 months later in both groups with a clinician rating. The primary hypothesis concerning the difference between groups in the individual percentage symptom reduction could be confirmed in the intention-to-treat, not the per-protocol sample. Additionally, we applied repeated-measures analyses of variance and found a higher symptom decrease in HT compared with WL patients in three of the five imputed datasets. The dropout rate was low, and satisfaction with the treatment was high. HT patients experienced a strong symptom reduction after receiving hypnotherapy. WL patients improved slightly during the waiting period. The COMT Val108/158Met genotype had an effect on the agoraphobia-related symptoms as well as on COMT DNA methylation levels. This is the first study to indicate that hypnotherapy performed better than a waitlist control group regarding the reduction in anxiety symptoms in an RCT. Future studies should confirm the efficacy of hypnotherapy and compare the treatment with a standard treatment for anxiety disorders in a larger trial. Future studies should also investigate whether hypnotic susceptibility is associated with COMT Val108/158Met genotype and could predict treatment success for HT. Clinical trial registration https://classic.clinicaltrials.gov/ct2/show/NCT03684577, identifier: NCT03684577.
Article
Patients diagnosed with posttraumatic stress disorder (PTSD) present with a spectrum of debilitating anxiety symptoms resulting from exposure to trauma. Women are twice as likely to be diagnosed with anxiety and PTSD compared to men; however, the reason for this vulnerability remains unknown. We conducted four experiments where we first demonstrated a female vulnerability to stress‐enhanced fear learning (SEFL) with a moderate, acute early life stress (aELS) exposure (4 footshocks in a single session), compared to a more intense aELS exposure (15 footshocks in a single session) where males and females demonstrated comparable SEFL. Next, we demonstrated that this female vulnerability does not result from differences in footshock reactivity or contextual fear conditioning during the aELS exposure. Finally, using gonadectomy or sham surgeries in adult male and female rats, we showed that circulating levels of gonadal steroid hormones at the time of adult fear conditioning do not explain the female vulnerability to SEFL. Additional research is needed to determine whether this vulnerability can be explained by organizational effects of gonadal steroid hormones or differences in sex chromosome gene expression. Doing so is critical for a better understanding of increased female vulnerability to certain psychiatric diseases.
Chapter
Full-text available
The metamorphosis from larvae to adult butterflies has represented the “mystery” of life since the ancient Greeks. How could we explain the various steps of development from caterpillars to the most beautiful butterflies? A mystery prevalent in the twentieth century concerned the storage of the complete genetic information of an organism in the DNA of its every cell. How and why do so many different cell types develop throughout the lives of organisms at the right time and place? With the rise of epigenetics, the “fog” of mystery is starting to clear. We now know that the genetic storage medium in every living cell acquires an incredible plasticity through certain markings on the genetic text, linking the inheritable information with the contextuality of the real lifeworld of each organism. This means that the concrete living organism influences during development the various stages of gene expression, transcription, translation, immunity, and DNA repair actively and leads to various phenotypic outcomes without altering the DNA storage medium. More surprisingly, such variations of developmental phenotypes were found capable of successfully adapting to changing environmental circumstances. Better adapted organisms may lead to reprogramming in the epigenetic states which may reach an inheritable status for many generations or even become a fixed part of the genetic identity of the species. This is how evolution learns.
Article
Full-text available
Early life stress (ELS) in the form of trauma or caregiver abuse and neglect is often associated with psychopathology. However, not everyone exposed to ELS develops a pathology; others display resilience, or the ability to adapt and persevere despite ongoing adversity. Several molecular moderator variables between ELS and behavioral phenotypes have been proposed, including single nucleotide polymorphisms (SNPs) and epigenetic markers. Specifically, several SNPs and aberrant methylation or expression of genes associated with neurotransmitter systems and brain-derived neurotrophic factor have been associated with anxiety, depression or schizophrenia. The present review seeks to explore the relationship between SNPs, epigenomics and disease, and offer data to suggest several SNPs may also predict specific treatment efficacy and psychological resilience. Due to these different mental health outcomes as a function of ELS, it is critical that environmental moderators be equally considered in determining the ontology of resilient or pathological phenotypes; this includes the infant-caregiver relationship, and the degree of control, magnitude, and type of the stressor experienced. Finally, we will offer evidence to suggest that several intervention strategies, including drug treatment, environmental enrichment, or exercise can ameliorate many of the psychological, biological, and molecular consequences of ELS exposure, and help shift one toward a resilient phenotype.
Article
Inflammation stimulates the hypothalamic-pituitary adrenal (HPA) axis and triggers glial neuroinflammatory phenotypes, which reduces monoamine neurotransmitters by activating indoleamine 2,3-dioxygenase enzyme. These changes can induce psychiatric diseases, including anxiety. Corticotropin releasing hormone receptor 2 (CRHR2) in the HPA axis is involved in the etiology of anxiety. Omega(n)-3 polyunsaturated fatty acids (PUFAs) can attenuate anxiety through anti-inflammation and HPA axis modulation. However, the underlying molecular mechanism by CRHR2 modulates anxiety and its correlation with neuroinflammation remain unclear. Here, we first constructed a crhr2 zebrafish mutant line, and evaluated anxiety-like behaviors, gene expression associated with the HPA axis, neuroinflammatory response, neurotransmitters, and PUFAs profile in crhr2+/+ and crhr2-/- zebrafish. The crhr2 deficiency decreased cortisol levels and up-regulated crhr1 and down-regulated crhb, crhbp, ucn3l and proopiomelanocortin a (pomc a) in zebrafish. Interestingly, a significant increase in the neuroinflammatory markers, translocator protein (TSPO) and the activation of microglia M1 phenotype (CD11b) were found in crhr2-/- zebrafish. As a consequence, the expression of granulocyte-macrophage colony-stimulating factor, pro-inflammatory cytokines vascular endothelial growth factor, and astrocyte A1 phenotype c3 were up-regulated. While microglia anti-inflammatory phenotype (CD206), central anti-inflammatory cytokine interleukin-4, arginase-1, and transforming growth factor-β were downregulated. In parallel, crhr2-deficient zebrafish showed an upregulation of vdac1 protein, a TSPO ligand, and its downstream caspase-3. Furthermore, 5-HT/5-HIAA ratio was decreased and n-3 PUFAs deficiency was identified in crhr2-/- zebrafish. In conclusion, anxiety-like behavior displayed by crhr2-deficient zebrafish may be caused by the HPA axis dysfunction and enhanced neuroinflammation, which resulted in n-3 PUFAs and monoamine neurotransmitter reductions.
Article
Full-text available
Both depression and obesity are health problems frequently encountered all around the world. The studies carried out in recent years indicate that there is a relationship between obesity and depression but a complete cause and effect relationship cannot be determined. The purpose of this research is to seek an answer to the question of does depression cause obesity o r does obesity prompt depression. Studies were found using PubMed, PsycINFO, and EMBASE databases and selected on several criteria. When the relationship between depression and obesity is examined, the most basic question is which one affects the other one, in other words what the direction of the relationship between them is. Consequently: studies indicate that there is a relationship between obesity and depression and focus on cause and effect relationship. It is argued that this relationship is bilateral, and it is stated that many factors can affect this relationship. It has been found that obesity increases the depression risk, and depression is also a predictor factor for the development of obesity. In conclusion, it can be said that there is not a single cause for the relationship between obesity and depression, and it has a multi-factorial structure.
Article
Full-text available
It is well known that the hypothalamo-pituitary-adrenal (HPA) axis is altered by early environmental experiences, particularly in the perinatal period. This may be one mechanism by which the environment changes the physiology of the animal such that individual differences in adult adaptative capabilities, such as behavioral reactivity and memory performance, are observable. To determine the origin of these behavioral individual differences, we have investigated whether the long-term influence of prenatal and postnatal experiences on emotional and cognitive behaviors in adult rats are correlated with changes in HPA activity. To this end, prenatal stress of rat dams during the last week of gestation and postnatal daily handling of rat pups during the first 3 weeks of life were used as two environmental manipulations. The behavioral reactivity of the adult offspring in response to novelty was evaluated using four different parameters: the number of visits to different arms in a Y-maze, the distance covered in an open field, the time spent in the corners of the open field, and the time spent in the open arms of an elevated plus-maze. Cognitive performance was assessed using a water maze and a two-trial memory test. Adult prenatally stressed rats showed high anxiety-like behavior, expressed as an escape behavior to novelty correlated with high secretion of corticosterone in response to stress, whereas adult handled rats exhibited low anxiety-like behavior, expressed as high exploratory behavior correlated with low secretion of corticosterone in response to stress. On the other hand, neither prenatal stress nor handling changed spatial learning or memory performance. Taken together, these results suggest that individual differences in adult emotional status may be governed by early environmental factors; however, perinatal experiences are not effective in influencing adult memory capacity.
Article
Full-text available
Significance Successful response to stress requires that an organism rapidly direct its energy toward an appropriate survival response. The brain is central to successful survival decisions, and therefore its ability to allocate energetic resources precisely in response to stress is paramount. Glucocorticoid stress hormones have long been known to assist in the liberation of energy during stress via their ability to regulate the activity of the nuclear genome. The cellular powerhouse, the mitochondria, also contains a genome; herein we show that glucocorticoids, acting through their receptors, regulate the expression of mitochondrial genes in the brain. These findings demonstrate a direct molecular linkage between stress and mitochondrial function.
Article
Full-text available
Unlabelled: The RNA modification N(6)-methyladenosine (m(6)A) influences mRNA stability and cell-type-specific developmental programming, and is highly abundant in the adult brain. However, it has not been determined whether m(6)A is dynamically regulated by experience. Based on transcriptome-wide profiling of m(6)A, we report that the level of m(6)A increases in the medial prefrontal cortex (mPFC) of mice in response to behavioral experience. The modulation was enriched near the stop codon of mRNAs, including genes related to neuronal plasticity. In primary cortical neurons, in vitro, modulation of m(6)A by the RNA demethylase FTO influenced the degradation profiles of a subset of transcripts with modulated sites. In vivo, the expression of Fto and the m(6)A methyltransferase, Mettl3 correlated with the observed increase in m(6)A levels post-training. Furthermore, targeted knockdown of FTO in the mPFC led to enhanced consolidation of cued fear memory. Thus, together with its role in early development, the dynamic regulation of m(6)A in the adult brain serves as an important epitranscriptomic mechanism associated with behavioral adaptation. Significance statement: N(6)-methyladenosine (m(6)A) is the most prevalent internal modification on RNA, however, its cellular dynamics in vivo remains elusive. Here we provide the first demonstration of m(6)A upregulation in the mouse medial prefrontal cortex (mPFC) following behavioral training. Knocking down the m(6)A demethylase FTO in the mPFC, which increases total m(6)A level, results in enhanced consolidation of fear memory. Our findings suggest that m(6)A is regulated in an activity-dependent manner in the adult brain, and may function to fine-tune mRNA turnover during memory-related processes.
Article
Full-text available
Waddington coined the term “epigenetic” to attempt to explain the complex, dynamic interactions between the developmental environment and the genome that led to the production of phenotype. Waddington’s thoughts on the importance of both adaptability and canalization of phenotypic development are worth recalling as well, as they emphasize the available range for epigenetic action and the importance of environmental feedback (or lack thereof) in the development of complex traits. We suggest that a dynamic systems view fits well with Waddington’s conception of epigenetics in the developmental context, as well as shedding light on the study of the molecular epigenetic effects of the environment on brain and behavior. Further, the dynamic systems view emphasizes the importance of the multi-directional interchange between the organism, the genome and various aspects of the environment to the ultimate phenotype.
Article
Full-text available
Compelling evidence suggests that maternal mental health in pregnancy can influence fetal development. The imprinted genes, insulin-like growth factor 2 (IGF2) and H19, are involved in fetal growth and each is regulated by DNA methylation. This study aimed to determine the association between maternal mental well-being during pregnancy and differentially methylated regions (DMRs) of IGF2 (DMR0) and the IGF2/H19 imprinting control region (ICR) in newborn offspring. Maternal depression, anxiety and perceived stress were assessed at 28 weeks of pregnancy in the Barwon Infant Study (n=576). DNA methylation was measured in purified cord blood mononuclear cells using the Sequenom MassArray Platform. Maternal anxiety was associated with a decrease in average ICR methylation (Δ=-2.23%; 95% CI=-3.68 to -0.77%), and across all six of the individual CpG units in anxious compared with non-anxious groups. Birth weight and sex modified the association between prenatal anxiety and infant methylation. When stratified into lower (⩽3530 g) and higher (>3530 g) birth weight groups using the median birth weight, there was a stronger association between anxiety and ICR methylation in the lower birth weight group (Δ=-3.89%; 95% CI=-6.06 to -1.72%), with no association in the higher birth weight group. When stratified by infant sex, there was a stronger association in female infants (Δ=-3.70%; 95% CI=-5.90 to -1.51%) and no association in males. All the linear regression models were adjusted for maternal age, smoking and folate intake. These findings show that maternal anxiety in pregnancy is associated with decreased IGF2/H19 ICR DNA methylation in progeny at birth, particularly in female, low birth weight neonates. ICR methylation may help link poor maternal mental health and adverse birth outcomes, but further investigation is needed.
Article
Full-text available
Studies in humans and experimental animals have demonstrated the vulnerability of the adolescent brain to actions of ethanol and the long-term consequences of binge drinking, including the behavioral and cognitive deficits that result from alcohol neurotoxicity, and increased risk to alcohol abuse and dependence. Although the mechanisms that participate in these effects are largely unknown, we have shown that ethanol by activating innate immune receptors, toll-like receptor 4 (TLR4), induces neuroinflammation, impairs myelin proteins and causes cognitive dysfunctions in adolescent mice. Since neuroimmune signaling is also involved in alcohol abuse, the aim of this study was to assess whether ethanol treatment in adolescence promotes the long-term synaptic and molecular events associated with alcohol abuse and addiction. Using wild-type (WT) and TLR4-deficient (TLR4-KO) adolescent mice treated intermittently with ethanol (3 g/kg) for 2 weeks, we showed that binge-like ethanol treatment in adolescent mice promotes short- and long-term alterations in synaptic plasticity and epigenetic changes in the promoter region of bdnf and fosb, which increased their expression in the mPFC of young adult animals. These molecular events were associated with long-term rewarding and anxiogenic-related behavioral effects, along with increased alcohol preference. Our results further showed the participation of neuroimmune system activation and the TLR4 signaling response since deficient mice in TLR4 (TLR4-KO) are protected against molecular and behavioral alterations of ethanol in the adolescent brain. Our results highlight a new role of the neuroimmune function and open up new avenues to develop pharmacological treatments that can normalize the immune signaling responsible for long-term effects in adolescence, including alcohol abuse and related disorders.
Article
Full-text available
It is known that increased circulating glucocorticoids in the wake of excessive, chronic, repetitive stress increases anxiety and impairs Brain-Derived Neurotrophic Factor (BDNF) signaling. Recent studies of BDNF gene methylation in relation to maternal care have linked high BDNF methylation levels in the blood of adults to lower quality of received maternal care measured via self-report. Yet the specific mechanisms by which these phenomena occur remain to be established. The present study examines the link between methylation of the BDNF gene promoter region and patterns of neural activity that are associated with maternal response to stressful versus non-stressful child stimuli within a sample that includes mothers with interpersonal violence-related PTSD (IPV-PTSD). 46 mothers underwent fMRI. The contrast of neural activity when watching children-including their own-was then correlated to BDNF methylation. Consistent with the existing literature, the present study found that maternal BDNF methylation was associated with higher levels of maternal anxiety and greater childhood exposure to domestic violence. fMRI results showed a positive correlation of BDNF methylation with maternal brain activity in the anterior cingulate (ACC), and ventromedial prefrontal cortex (vmPFC), regions generally credited with a regulatory function toward brain areas that are generating emotions. Furthermore we found a negative correlation of BDNF methylation with the activity of the right hippocampus. Since our stimuli focus on stressful parenting conditions, these data suggest that the correlation between vmPFC/ACC activity and BDNF methylation may be linked to mothers who are at a disadvantage with respect to emotion regulation when facing stressful parenting situations. Overall, this study provides evidence that epigenetic signatures of stress-related genes can be linked to functional brain regions regulating parenting stress, thus advancing our understanding of mothers at risk for stress-related psychopathology.
Article
Background: Depression is a recurrent illness with high rates of chronicity, treatment-resistance and significant economic impact. There is evidence in the literature that S-adenosyl methionine (SAMe), a naturally occurring compound in the human body, has antidepressant efficacy. This product may be an important addition to the armamentarium of antidepressant agents. Objectives: To assess the effects of SAMe in comparison with placebo or antidepressants for the treatment of depression in adults. Search methods: We searched the Cochrane Common Mental Disorders Group's Specialised Register (CCMDCTR Studies and Reference Register), MEDLINE, EMBASE, PsycINFO, international trial registers ClinicalTrials.gov and the World Health Organization trials portal (ICTRP). We checked reference lists, performed handsearching and contacted experts in the field. The CCMDCTR literature search was last updated on 5 February 2016. Selection criteria: Randomised controlled trials comparing SAMe with placebo or antidepressants in adults with a diagnosis of major depression. Data collection and analysis: Two authors independently performed extraction of data and assessment of risk of bias. We contacted trialists of included studies for additional information. Main results: This systematic review included eight trials comparing SAMe with either placebo, imipramine, desipramine or escitalopram. We accepted trials that used SAMe as monotherapy or as add-on therapy to selective serotonin reuptake inhibitors (SSRIs), and we accepted both oral and parenteral administration. The review involved 934 adults, of both sexes, from inpatient and outpatient settings.The trials were at low risk of reporting bias. We judged the risk of selection, performance, detection and attrition bias as unclear or low, and one study was at high risk of attrition bias.There was no strong evidence of a difference in terms of change in depressive symptoms from baseline to end of treatment between SAMe and placebo as monotherapy (standardised mean difference (SMD) -0.54, 95% confidence interval (CI) -1.54 to 0.46; P = 0.29; 142 participants; 2 studies; very low quality evidence). There was also no strong evidence of a difference in terms of drop-out rates due to any reason between SAMe and placebo, when used as monotherapy (risk ratio (RR) 0.88, 95% CI 0.61 to 1.29; P = 0.52; 142 participants; 2 studies; low quality evidence).Low quality evidence showed that the change in depressive symptoms from baseline to end of treatment was similar between SAMe and imipramine, both as monotherapy (SMD -0.04, 95% CI -0.34 to 0.27; P = 0.82; 619 participants; 4 studies). There was also no strong evidence of a difference between SAMe and a tricyclic antidepressant in terms of drop-outs due to any reason (RR 0.61, 95% CI 0.28 to 1.31; P = 0.2; 78 participants; 3 studies; very low quality evidence).There was little evidence of a difference in terms of change in depressive symptoms from baseline to end of treatment between SAMe and escitalopram, both as monotherapy (MD 0.12, 95% CI -2.75 to 2.99; P = 0.93; 129 participants; 1 study; low quality evidence). There was no strong evidence of a difference between SAMe and escitalopram in terms of drop-outs due to any reason (RR 0.81, 95% CI 0.57 to 1.16; P = 0.26; 129 participants; 1 study; low quality evidence).There was low quality evidence that SAMe is superior to placebo as add-on to SSRIs in terms of change in depressive symptoms from baseline to end of treatment (MD -3.90, 95% CI -6.93 to -0.87; P = 0.01; 73 participants; 1 study). There was no strong evidence of a difference between SAMe and placebo as adjunctive therapy to an SSRI in terms of drop-outs due to any reason (RR 0.70, 95% CI 0.31 to 1.56; P = 0.38; 73 participants; 1 study; very low quality evidence).For all comparisons, secondary outcome measures of response and remission rates were consistent with these primary outcome measures.With regard to all extractable measures of the acceptability of SAMe, the quality of the evidence was low to very low. SAMe was not different from placebo and established antidepressants. The exception was that compared to imipramine, fewer participants experienced troublesome adverse effects when treated with parenteral SAMe.The specific adverse effects were not detailed in most of the included studies. There were two reports of mania/hypomania recorded for 441 participants in the SAMe arm. Authors' conclusions: Given the absence of high quality evidence and the inability to draw firm conclusions based on that evidence, the use of SAMe for the treatment of depression in adults should be investigated further. Future trials should be in the form of large randomised controlled clinical trials of high methodological quality, with particular attention given to randomisation, allocation concealment, blinding and the handling of missing data. Comparator antidepressants from all classes should be used. Adverse events should be detailed for each participant, bearing in mind that induction of mania is of particular interest.
Article
Objective: Increased risk of psychopathology is observed in children exposed to maternal prenatal distress, and elevated maternal cortisol and epigenetic regulation of placental glucocorticoid-pathway genes are potential mechanisms. The authors examined maternal distress and salivary cortisol in relation to fetal movement and heart rate ("coupling") and DNA methylation of three glucocorticoid pathway genes-HSD11B2, NR3C1, and FKBP5-in term placentas. Method: Mood questionnaires and salivary cortisol were collected from 61 women between 24-27 gestational weeks, and fetal assessment was conducted at 34-37 weeks. Placental CpG methylation in the three genes was analyzed using 450K Beadchips and bisulfite sequencing; correlations between maternal and fetal variables and DNA methylation were tested; and maternal distress effects on fetal behavior via DNA methylation were investigated. Results: Perceived stress (Perceived Stress Scale), but not cortisol, was associated with altered CpG methylation in placentas. In the highest tertile of the Perceived Stress Scale, the Beadchip data revealed modestly elevated methylation of HSD11B2, associated with lower fetal coupling (β=-0.51), and modestly elevated methylation of FKBP5, also with lower fetal coupling (β=-0.47). These increases in methylation were validated by bisulfite sequencing, where they occurred in a minority of clones. Conclusions: This is the first study to link the effects of pregnant women's distress on the fetus and epigenetic changes in placental genes. Since increased DNA methylation in HSD11B2 and FKBP5 are seen in a minority of bisulfite sequencing clones, these epigenetic changes, and functional consequences, may affect subpopulations of placental cells.
Article
Classically thought as genomic clutter, the functional significance of transposable elements (TEs) has only recently become a focus of attention in neuroscience. Increasingly, studies have demonstrated that the brain seems to have more retrotransposition and TE transcription relative to other somatic tissues, suggesting a unique role for TEs in the central nervous system. TE expression and transposition also appear to vary by brain region and change in response to environmental stimuli such as stress. TEs appear to serve a number of adaptive roles in the nervous system. The regulation of TE expression by steroid, epigenetic and other mechanisms in interplay with the environment represents a significant and novel avenue to understanding both normal brain function and disease.