Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Original article

A review of epigenetic contributions


to post-traumatic stress disorder
Hunter Howie, BS; Chuda M. Rijal, BS; Kerry J. Ressler, MD, PhD

Post-traumatic stress disorder (PTSD) is a syndrome which serves as a classic example of psychiatric disorders that result
from the intersection of nature and nurture, or gene and environment. By definition, PTSD requires the experience of a
traumatic exposure, and yet data suggest that the risk for PTSD in the aftermath of trauma also has a heritable (genetic)
component. Thus, PTSD appears to require both a biological (genetic) predisposition that differentially alters how the
individual responds to or recovers from trauma exposure. Epigenetics is defined as the study of changes in organisms caused
by modification of gene expression rather than alteration of the genetic code itself, and more recently it has come to refer to
direct alteration of DNA regulation, but without altering the primary sequence of DNA, or the genetic code. With regards
to PTSD, epigenetics provides one way for environmental exposure to be “written” upon the genome, as a direct result
of gene and environment (trauma) interactions. This review provides an overview of the main currently understood types
of epigenetic regulation, including DNA methylation, histone regulation of chromatin, and noncoding RNA regulation of
gene expression. Furthermore, we examine recent literature related to how these methods of epigenetic regulation may be
involved in differential risk and resilience for PTSD in the aftermath of trauma.
© 2019, AICH ‑ Servier Group Dialogues Clin Neurosci. 2019;21(4):417-428. doi:10.31887/DCNS.2019.21.4/kressler

Keywords: epigenetics; genetics; DNA methylation; histone acetylation; noncoding RNA; trauma; post-traumatic
stress disorder; childhood abuse

Introduction ations in cognition and mood; alterations in arousal and


reactivity associated with the traumatic event; and in some
Post-traumatic stress disorder (PTSD) is a Diagnostic cases, dissociative symptoms.1
and Statistical Manual of Mental Disorders 5th ed.

Copyright © 2019 AICH ‑ Servier Group. All rights reserved. www.dialogues-cns.org


(DSM-5) diagnosis marked by the development of stressor- While around 50% to 60% of the population will experi-
related symptoms following one or more traumatic events.1 ence traumatic stress over the course of their lifetime, the
Outlined in the DSM-5, a traumatic event is defined as expo- lifetime prevalence for PTSD using DSM-IV criteria has
sure to actual or threatened death, serious injury, or sexual been estimated at around 8.7%.2,3 The disparity between
violence that is experienced directly, witnessed, experienced trauma exposure and the development of trauma-related
vicariously through family or close friends, or experienced disorders has garnered much interest, and our understanding
repeatedly or with extreme exposure to aversive details of of what contributes to this susceptibility or resilience is still
the traumatic event. Diagnostic criteria and symptomology limited.4,5 The old debate of nature versus nurture sought
include the following: intrusive symptoms; avoidance of answers in a single domain; however, as our understanding
stimuli associated with the traumatic event; negative alter- continues to evolve, it has become clear that—like many

Author affiliations: Partners Healthcare, Boston, Massachusetts, US (Hunter Howie, Chuda M. Rijal, Kerry J. Ressler); McLean Hospital, Belmont, Massachusetts,
US (Hunter Howie, Chuda M. Rijal, Kerry J. Ressler); Harvard Medical School, Boston, Massachusetts, US (Kerry J. Ressler). Address for correspondence: Kerry J.
Ressler, MD, PhD, 115 Mill St, Belmont, MA 02478, US. (email: kressler@mclean.harvard.edu)

DIALOGUES IN CLINICAL NEUROSCIENCE • Vol 21 • No. 4 • 2019 • 417


Original article
Epigenetics of PTSD - Howie et al

other mental disorders–PTSD development is heavily In addition, PTSD has common comorbidity with other
influenced by an interplay between environmental factors mental disorders, namely major depression, substance
and genetic predisposition or heritability. The study of abuse, and other anxiety disorders.14 While the DSM-5 is
epigenetics bridges both sides of this debate and focuses limited by its definitions and diagnostic criteria, genetic
on the changes in gene expression that evidence suggests that these disorders
may be caused by our environment. may fall on a spectrum rather than
In our review on the epigenetics of Epigenetic approaches being entirely independent entities.
PTSD, we will discuss the heritability may afford novel
of this disorder and give an overview Epigenetic mechanisms
of epigenetic mechanisms, targets, targeted therapeutic
genome-wide association studies approaches to enhance Data from twin studies suggests that
(GWAS), and epigenome-wide asso- PTSD is at least partially heritable and,
ciation studies (EWAS) conducted to
treatment and by definition, requires influence from
date, and future directions for the field. prevention of PTSD environmental trauma.9 Epigenetics
is defined as the study of changes in
Heritability organisms caused by modification of
gene expression rather than alteration
Diagnosis of PTSD is reliant on environmental influence of the genetic code itself, and more recently it has come to
through a traumatic event. Given this fact, it may seem refer to direct alteration of DNA regulation, but without
backwards then to study the heritability of a disorder that altering the primary sequence of DNA, or the genetic code.
requires an outside event for its manifestation. Contrary to With regards to PTSD, epigenetics provides one way for
what may seem intuitive of a disorder with this diagnostic environmental exposure to be “written” upon the genome,
criterion, research suggests that genes do play a role, and as a direct result of gene and environment (trauma) inter-
perhaps a significant one, in the risk of developing PTSD. actions. The epigenome is influenced by both genetic and
environmental factors—the environment in effect is written
Twin studies serve as an invaluable tool to parse out genetic onto the genes themselves. While epigenetics does not
and environmental factors and contribute in concert with change the sequence of the DNA code, it does alter the
newer molecular and genetic methods to help piece together expression of genes and may contribute to long-lasting–in
a complete picture.6 In one twin study, heritable influences some cases intergenerational—phenotypic effects.15 There
accounted for 46% of the variance in PTSD, and for 71% of are several mechanisms that drive this process, three of
variance in females.7,8 Another study suggested that expo- which have been widely studied.
sure to assaultive trauma (robbery, sexual assault, and other
life-threatening events) may not be entirely random and is Histone modification
influenced by individual and familiar risk factors.9 Indeed, Histone proteins (H2A, H2B, H3, H4) help organize DNA
it is known that parental post-traumatic stress can cause into structured units called nucleosomes. Nucleosomes are
negative psychological outcomes and potential biological packaged units formed by spooling the DNA sequence of
alterations in their offspring, with several studies indicating 200 nucleotide base pairs around eight histones (octamer),
that severity of a parent’s PTSD symptoms may contribute which help to compact the DNA. The nucleosomes can be
to a child’s psychological difficulties—namely anxiety, thought of as “beads” and are connected by linker DNA
depression, and behavior problems.10,11 Furthermore, child- forming a collection of “beads on a string” called chromatin.
hood adversity has been strongly implicated in the devel- Chemical alteration or modification of histones–through
opment of many psychiatric disorders, and individuals who acetylation or deacetylation–influences the structure of
experience these early life adversities are at greater risk for chromatin, remodeling it to either coil or uncoil and altering
PTSD in adulthood.12,13 The psychological collateral of trau- the ability of RNA polymerase to transcribe genes. Thus,
ma-related distress can percolate through the family unit, histone acetylation or deacetylation regulates the extent to
potentially exacerbating risk factors that may lead to the which a gene is expressed by altering chromatin structures.
development of future psychological distress or disorder. Histone regulation has been implicated in a number of activ-

418 • DIALOGUES IN CLINICAL NEUROSCIENCE • Vol 21 • No. 4 • 2019


Original article
Epigenetics of PTSD - Howie et al

ities in the brain related to emotion regulation, for example events. Thus, it is important to understand the mechanism
traumatic memory encoding and fear extinction,16 which is of both addition and removal of methyl groups. Meth-
an important process that is dysregulated in PTSD. ylation is mediated by DNA methyltransferase proteins,
DNMT3a and DNMT3b, to add a methyl group to an
Histone acetylation occurs when the enzyme histone acetyl- unmethylated cytosine C5 position. 25 The oxidation of
transferase (HAT) interacts with the histone protein, adding 5-methylcytosine to 5-hydroxymethylcytosine is medi-
an acetyl group to lysine residues in the N-terminal tail of ated by ten-eleven translocation proteins (TET1, TET2,
the histone protein. Acetylation of histones by HAT causes and TET3); hydroxymethylcytosine is an intermediate step
the uncoiling or loosening of DNA, creating decondensed in DNA demethylation. DNA-binding proteins have also
and “open” chromatin structure (euchromatin), which allows shown to be involved in active demethylation of DNA, and
access to the DNA by proteins involved in the transcrip- other proteins involved in dynamic transcriptional acti-
tional machinery, copying the DNA sequence into RNA. vation or repression can also “recruit” DNMT and TET
Conversely, histone deacetylation occurs when histone proteins, leading to a longer-lasting alteration in DNA
deacetylase (HDAC) removes the acetyl group added by methylation status.26,27
the HATs. Deacetylation of histones by HDAC causes the
coiling of DNA, creating condensed and “closed” chromatin One example of DNA methylation findings are the role of
structure (heterochromatin) making it densely packed and differential methylation at the FKBP5 gene, which is further
more difficult to transcribe—in effect inhibiting or repressing outlined below as a critical regulator of the HPA cortisol
gene expression. Histone methylation provides yet another response. DNA demethylation at glucocorticoid receptor
mechanism of histone regulation, mediated by histone meth- binding site (GREs) within the FKBP5 gene in peripheral
yltransferases. Generally, histone methylation is thought to blood cells and hippocampal progenitor cells was found
have an opposite effect to histone acetylation, generally to be associated with prior exposure to childhood abuse.23
consistent with condensing chromatin structure, though this is Recent studies also suggest that epigenetic marks might be
also dependent on which specific amino acid components of transmitted down to the next generation, influencing the
the histones are modified. In summary, without changing the risk of diseases in offspring,28,29 though these have typically
DNA sequence or even directly modifying the DNA chemical been small or underpowered studies that require expansion
structure, this process of histone modification allows genes and replication.
to be turned on and off by making regions of DNA either
accessible or inaccessible to the transcriptional machinery. Noncoding RNA
Noncoding RNAs (ncRNAs) are transcripts from DNA,
DNA methylation but unlike other RNAs, ncRNAs are not translated into a
DNA modification through direct methylation is one polypeptide or protein sequence. ncRNAs are functional
epigenetic process that has been widely studied in PTSD. and are involved in the processing and regulation of other
Although there are more than 20 identified DNA modifi- RNAs such as messenger RNA (mRNA), transfer RNA
cations,17 5-methylcytosine (5-mc) and 5-hydroxymeth- (tRNA), and ribosomal RNA (rRNA). 30,31 Recent more
ylcytosine (5-hmc)18 are two types of methylation-related detailed reviews highlight the role of different types of
modifications that are highly prevalent in neurons related ncRNAs, such as micro RNA (miRNA), long noncoding
to known processes involved in PTSD, such as learning RNA (lncRNA), and retrotransposons, that may become
and extinction of conditioned fear.19‑21 DNA methylation useful biomarkers for trauma-related brain disorders such
changes within a gene can occur at any stage during the as PTSD.32
life cycle of a cell,22 and they have been characterized to
make a long-term impact on transcriptional response due miRNAs are characterized by 21 to 24 nucleotides in length.
to different stress-related environmental factors, including They are thought to generally bind to the 3’ untranslated
early adverse life events.23,24 region or other untranslated regions of their target mRNAs
to regulate gene expression.33 Studies have reported the
There have been many reports of increased and decreased miRNA expression level changes in experimental stress
DNA methylation in response to exposure of stressful life models.34‑36 One of those studies linked to hypothalamus-

DIALOGUES IN CLINICAL NEUROSCIENCE • Vol 21 • No. 4 • 2019 • 419


Original article
Epigenetics of PTSD - Howie et al

pituitary-adrenal (HPA) axis pathways identified a specific (FKBP5), discussing several of the more robust studies
miRNA, miR-34c, to be upregulated in a stress-dependent examining this target.
manner in mouse amygdala tissue.37 In rodents, miRNA also
seem to be regulating glucocorticoid receptor (GR) func- Repeated exposure to trauma alters endocrine mecha-
tion through post-transcriptional effects that are sensitive nisms involved in the stress response. The hypothalam-
to stress exposure, thus influencing the regulation of GR- ic-pituitary-adrenal (HPA) stress axis—in addition to
regulated downstream genes to alter the behavioral response filling many other roles such as immune and metabolic
to stress.34 function—guides the endocrine response to stress.45 HPA
axis dysregulation has been observed in both depression
lncRNAs are characterized by longer than 200 nucleotides and PTSD, though each disorder appears to have unique
in length and are also involved in regulation of gene expres- manifestations.45 The effects of this dysregulation change
sion in PTSD.38 Although there are not many studies on the HPA axis function, altering its response to cortisol
the role of lncRNAs in PTSD, Guffanti et al previously feedback. This is thought to be associated with physi-
identified a single nucleotide variant lncRNA- lncRN- ological and psychological emotion dysregulation and
ALINC01090 (previously called AC068718.1) that reached stress hyper-responsiveness, both of which are implicated
genome-wide significance in a GWAS of PTSD.39 in PTSD.41 Evidence also suggests that early life adver-
sity may contribute to epigenetic changes in the HPA axis
Candidate gene studies which may impact the development of PTSD.45

While twin studies suggest a genetic component to PTSD, FK506 binding protein 51 gene
candidate gene studies aid in identifying specific genes that The FK506 binding protein 51 (FKBP5) gene is perhaps
may be associated with the disorder. Candidate gene designs the most comprehensively studied candidate among
examine the main effect of specific genes on expression of genes related to the HPA axis. It is believed to be an
a disorder and typically focus on biological candidates that important regulator of stress response through altering
are selected using existing biological evidence.4,5 That said, GR sensitivity.5,46 Through its role as both an inhibitor of
there has been much pushback, even controversy, as to the GR translocation to the nucleus, but also an exquisitely
use of candidate gene studies in recent years, as large-scale stress- and GR-responsive gene, it is thought to act as a
GWAS studies have been possible and have expanded mark- rapid, intracellular feedback regulator of GR sensitivity
edly. Few if any prior candidate genes have been replicated within the cell. FKBP5 has shown strong association with
in a well-powered GWAS, though the Million Veteran’s PTSD in conjunction with a history of childhood trauma/
Program study appears to have found an HPA-related gene, abuse when examined in gene-environment interaction
corticotrophin releasing factor receptor (CRFR1), associ- (GxE) studies. 23,47,48 One such study identified allele-
ated with hyperarousal in PTSD.40 Additionally, without specific, early trauma exposure-dependent demethyla-
well-defined pathophysiology of a disease—as is the case tion of CpGs in FKBP5, which suggests a FKBP5 x child
for psychiatric disorders—it is not always straightforward to abuse interaction resulting in differential (?upregulated)
define candidate genes. Overall, prior candidate gene studies transcriptional activation of FKBP5 in response to child-
need to be regarded with skepticism until larger-scale repli- hood abuse.23 Wang et al systematically reviewed interac-
cations and extensions have more definitively demonstrated tions between FKBP5, early life stress, and risk for PTSD.
their involvement or lack thereof. Results from this meta-analysis revealed a significant
interaction between the T allele of rs1360780 and early
With those caveats in mind, we feel it is still worthwhile, life stress in those with PTSD. The C-allele of rs3800373
even if for historical purposes, to mention the work to date. and the T-allele of rs9470080 also interacted with early life
The current literature on the genetic markers for PTSD stress and predicted higher risk for PTSD.49 A second more
spans more than 100 studies published since 1991: for a recent meta-analysis reaffirmed these findings and adds
detailed overview of this literature, we direct readers to to the mounting evidence of an overall effect of FKBP5
several comprehensive reviews.4,5,41‑44 The present review interacting with trauma exposure on PTSD.50 Although the
focuses on the HPA axis and FK506 binding protein 51 precise mechanisms are still to be understood, a working

420 • DIALOGUES IN CLINICAL NEUROSCIENCE • Vol 21 • No. 4 • 2019


Original article
Epigenetics of PTSD - Howie et al

hypothesis is that exposure to prior trauma, particularly a 10-year period.44 A major challenge with this approach,
early life stress, interacts with stress-sensitive genotypes however, is amassing a large enough sample to achieve
through long-lasting DNA methylation changes in FKBP5, the required statistical power to detect these loci—with
leading to greater stress responsiveness later in life through a statistical P-value threshold of 5x10-8 required for the
altered GR sensitivity. multiple test correction after examining roughly a million
SNPs per individual.44 The Psychiatric Genomic Consor-
Epigenetics of intergenerational transmission tium (PGC) was organized in 2007 to centralize the GWAS
of stress from around the world and to adequately power analyses.42
Subsequently they are now the largest collaboration in the
Recent mechanistic studies using animal models have inves- history of psychiatry, with more than 250 000 subjects,
tigated the effects of stress on epigenetic machinery and and the inclusion of more than 500 scientists from 100
how future generations may be affected. Rodent studies countries.42
examining parental care influences revealed that differences
in maternal phenotypes, namely grooming behavior, had Already other mental disorders such as schizophrenia,
effects on their pup’s development of behavioral and HPA depression, and bipolar disorder have utilized GWAS
responses to stress as adults.51 Further studies examining successfully to identify genes and molecular pathways of
DNA methylation in high vs low maternal care parental interest, and only recently has focus turned to PTSD—
behaviors revealed elevated DNA methylation levels in the with the first GWAS recently published in 2013.53 We have
offspring after low maternal care, and lower methylation in identified 12 successful GWAS in PTSD to-date (Table 1)
those with high maternal care—potentially contributing to from individual studies, which discovered several genes
reduced transcriptional activation of the GR in the low-ma- of interest due to their prior associations with stress or
ternal care offspring.23,51 In humans, Yehuda et al examined epigenetic regulation of neuronal function, including
intergenerational effects of trauma in Holocaust survivors the following: LINC01090, BC036345, ZNRD1-ASI, and
and their offspring through measuring cytosine methylation RORA.40,42,46,54‑62 Most of these cohorts and many more
within the FKBP5 gene. Results revealed differential find- have been combined for the meta-analytic approaches of
ings for survivors and their offspring, with higher levels of the PGC, and are currently examining GWAS for PTSD
methylation in survivors compared with controls, and lower in >150 000 subjects. Thus, it is still relatively early in the
in offspring, further demonstrating the potential of trauma field and further research is required. GWAS are the first
influences on epigenetic mechanisms to have intergenera- step in identifying these genes, and further studies using
tional effects.52 While these studies have small sample sizes an array of molecular and clinical methods must still be
and need replication in much larger cohorts, as discussed employed to validate these findings.5 Given the multiple
below with genome-wide studies, they provide intriguing genes of small effect size that are found in large-scale
initial insight into gene regulation as a function of epigen- GWAS, it is not yet entirely clear how these findings will
etic alterations in trauma-related symptoms and syndromes. lead to actionable interventions. There are several thoughts
about this, which remain to be determined: (i) while any
Genome-wide association studies given SNP or gene may have small effects, “hub”genes or
combined pathways may be triangulated and together have
The current most powerful and robust method to study much larger effect size and serve as important targets repre-
the interplay between genetics and PTSD uses genome- senting additive risk from multiple genes and SNPs; (ii)
wide association studies (GWAS) to provide an unbiased while the common variant findings indeed have very small
approach to identify loci in the genome that have associ- effects when the entire syndrome of PTSD is considered,
ation with PTSD. Large-scale GWAS compare hundreds there may be yet-to-be-determined biological subtypes of
of thousands of single-nucleotide polymorphisms (SNP) PTSD, each of which is determined by a smaller number of
across the entire genome to identify variants that may have larger effect size variants with less biological heterogeneity;
a causal effect on the disorder. Only in recent years has or iii) while the common variants and genes themselves in
the fiscal feasibility of using this method become possible a causal fashion are of limited effect, their identification
with a drastic 2000-fold reduction in cost per genotype in will lead to novel understanding of the biology of PTSD,

DIALOGUES IN CLINICAL NEUROSCIENCE • Vol 21 • No. 4 • 2019 • 421


Original article
Epigenetics of PTSD - Howie et al

STUDY SAMPLE SIZE COHORT SNP(S) GENE P VALUE HIGHLIGHTS

Logue et al, Discovery: Discovery: white rs8042149 Retinoid-related 2.50E-08


2013 N= 491 European American orphan receptor
military veterans alpha (RORA)

Replication: Replication: African


N=600 American military
veterans

Xie et al, Discovery: European American rs6812849 Tolloid-Like 1 3.10E-09


2013 N= 4344 and African American (TLL 1)

Replication: European American


N=2643 and African American

Guffanti et al, Discovery: Discovery: DNHS rs10170218 LINC01090 5.09E-08


2013 N= 413 women (long noncoding
RNA)

Replication: Replication: NHSII


N=2541 women

Wolf et al, Discovery: European American rs263232 Adenylyl cyclase 8 6.12E-07


2014 N= 484 military veterans (ADCY8)

Nievergelt Discovery: MRS military rs6482463 Phosphoribosyl 2.04E-09


et al, 2015 N=3494 veterans transferase
domain contai-
ning 1 (PRTFDC1)

Replication:
N= 491

Almli et al, Discovery: Discovery: Military rs717947 BC036345 1.28E-08


2015 N=147 veterans (long noncoding
RNA)

Replication: Replication: GTP


N=2006 Large urban
community cohort

422 • DIALOGUES IN CLINICAL NEUROSCIENCE • Vol 21 • No. 4 • 2019


Original article
Epigenetics of PTSD - Howie et al

STUDY SAMPLE SIZE COHORT SNP(S) GENE P VALUE HIGHLIGHTS

Ashley-Koch Discovery: Non-Hispanic black rs7866350 TBC1domain 1.10E-06


et al, 2015 N=1708 (NHB) (NHW Co- family member 2
Non-Hispanic white hort) (TBC1D2)
(NHW)

Stein et al, Discovery: American military rs159572 Ankyrin repeat 2.34E-08


2016 N=7774 veterans domain 55
(ANKRD55)

Replication: rs11085374 Zinc finger prot. 4.59E-08


N=5916 626 (ZNF626)

Kilaru et al, Discovery: GTP Large urban N/A Neuroligin 1 minSNP:


2016 N=3678 community cohort (NLGN1) 1.00E-06

Replication: DCHS pregnant south N/A ZNRD1-AS1 VEGAS:


N=205 African women (long noncoding 1.00E-06
RNA)

Melroy-Greif Discovery: Mexican Americans rs6681483 Olfactory recep- 1.83E-06


et al, 2017 N=512 and American Indians rs6667389 tor family 11
rs10888255 subfamily L
rs10888257 Member 1
(OR11L1)

Duncan et al, Discovery: Trauma exposed rs139558732 Kelch-like protein 3.33E-08 PGC-PTSD
2017 N=20730 adults from 11 African 1 (KLHL1)
contributing studies American

Morey et al, Discovery: European American rs6906714 LINC02571 5.99E-08


2018 N=157 and African American rs17012755 6.05E-08
military veterans rs76832471 6.51E-08
rs9499406 8.19E-08

Replication: GTP African American


N=133 women

Table I. Genome-wide association studies in PTSD. GTP- Grady Trauma Project; DCHS-Drakenstein Child Health Study;
NHSII- Nurses’ Health Study II.

DIALOGUES IN CLINICAL NEUROSCIENCE • Vol 21 • No. 4 • 2019 • 423


Original article
Epigenetics of PTSD - Howie et al

SAMPLE SIGNIFICANT
STUDY COHORT GENE P VALUE HIGHLIGHTS
SIZE CPG SITE(S)

Uddin et al, N=100 DNHS cg17709873 Retinoid-related orphan 3.00E-3


2010 cg25831111 receptor alpha (RORA) (unad-
Coenzyme A synthase justed)
(COASY) 1.00E-3
(unad-
justed)

Smith et al, N=110 African Ameri- cg24577137 Translocated promoter 1.90E-06


2011 cans cg08081036 region, nuclear basket 9.30E-06
cg20098659 protein (TPR) 4.30E-06
cg07967308 Annexin A2 (ANXA2) 8.00E-06
cg07759587 C-type lectin domain 1.10E-05
family 9 member a
(CLEC9A)
Acid phosphatase 5,
Tartrate resistant (ACP5)
TLR8 toll like receptor 8
(TLR8)

Uddin et al, N=100 DNHS 118 CpG 1.00E-2


2013 sites/ (unad-
116 genes justed)

Mehta et al, N=168 GTP 458 CpG <5.00E-2


2013 sites/
164 genes

Mehta et al, Discovery: Australian male cg26499155 Intergenic (43 kb from 7.94E-07
2017 N=211 Vi-etnam war cg02357741 leucine-rich repeat 2.24E-06
veterans cg09325682 contain-ing 3B [LRRC3B]) 3.28E-06
cg17750109 BR Serine/threonine 3.06E-06
Replication: GTP males cg16277944 kinase 1 (BRSK1) 4.95E-06
N=115 Lipocalin 8 (LCN8)
Nerve growth factor
(NGF)
Dedicator of cytokinesis
2 (DOCK2)

Rutten et al, Discovery: Male Dutch 17 DMPs and Dual specificity phospha- <5.00E-2
2017 N=93 military vete- 12 DMRs tase 22 (DUSP22)
rans Histone cluster 1 H2A
pseudogene 2 (HIS-
Replication: Male American T1H2APS2)
N=98 mili-tary Hook microtubule tethe-
veterans ring Protein 2 (HOOK2)
Ninjurin 2 (NINJ2)
Paired box 8 (PAX8)
Ring finger protein 39
(RNF39)
Zinc finger protein 57
(ZFP57)

424 • DIALOGUES IN CLINICAL NEUROSCIENCE • Vol 21 • No. 4 • 2019


Original article
Epigenetics of PTSD - Howie et al

SAMPLE SIGNIFICANT
STUDY COHORT GENE P VALUE HIGHLIGHTS
SIZE CPG SITE(S)

Hammamieh et Training set: OEF/OIF Male 5578 diffe- 3662 DMGs <5.00E-2
al, 2017 N=99 Amer-ican rential-ly 3339 DMGs (unad-
Test set: military methylated justed)
N=60 veterans CpG islands
Merged:
N=159

Kuan et al, N= 473 WTC cg05693864 Zinc finger DHHC-type 1.73E-06


2017 cg06182923 containing 11 (ZDHHC11) 4.73E-05
cg08696494 CUB and sushi multiple 5.39E-05
cg25664402 domains 2 (CSMD2) 5.80E-05
cg05569176 Collagen type IX alpha 3 7.82E-05
cg09370982 chain (COL9A3) 8.97E-05
cg07654569 Intergenic 9.91E-05
Programed cell death
6 Interacting protein
(PDCD6IP)
TBC1 domain family
member 24 (TBC1D24)
Family with sequence
similarity 164, member
A (FAM164A)

Ratanathara- N=147 Four milita- Proposed – Consortium Planned


thorn et al, ry cohorts Study Description meta-
2017 (MRS, PRISMO, analysis
VA-M, and VA-
NCPTSD)
Three civilian
cohorts (DNHS,
GTP, and WTC)

Uddin et al, N=545 Civilian trauma cg23637605 Neuregulin 1 (NRG1) 4.66E-08 Meta-
2018 ex-posed cg19577098 Hepatocyte growth 1.47E-07 analysis
cohorts factor-regulated tyrosine
ki-nase substrate (HGS)

Table II. Epigenome-wide association studies in post-traumatic stress disorder. OEF/OIF-Operation Enduring/Iraqi Freedom;
MRS- Marine Resiliency Study; PRISMO- Prospective Research in Stress-Related Military Operations; VA-M- Veterans Affairs’
Mental Illness Research, Education and Clinical Centers; VA-NCPTSD- National Center for PTSD; WTC- World Trade Center 9/11
First Responders study.

DIALOGUES IN CLINICAL NEUROSCIENCE • Vol 21 • No. 4 • 2019 • 425


Original article
Epigenetics of PTSD - Howie et al

leading to novel more powerful interventions. In summary, Conclusions and future directions
the field is hopeful that many robust GWAS gene candidates
will be identified with these tools, potentially transforming Epigenetics provides potentially the best approach for under-
our approach to the biology of PTSD. standing the interaction of genetics with environmental expo-
sure to trauma in PTSD. Here we have reviewed some recent
Epigenome-wide association studies progress in understanding DNA methylation, histone regula-
tion, and noncoding RNA approaches to epigenetic regulation
Following a similar unbiased approach, epigenome-wide in PTSD. The most profound recent progress in the biology
association studies (EWAS) offer a novel approach to of PTSD has been the onset of large-scale collaborations to
finding candidate gene pathways through examining epigen- support enormous studies in the genetic architecture of PTSD
etic mechanisms at a genome-wide level. EWAS studies through the Psychiatric Genomics Consortium and collabo-
to-date have focused primarily on DNA methylation, rative GWAS studies, allowing compilation of hundreds of
which has been the most cost-effective to examine in large thousands of samples. These consortia are also beginning to
data sets.30 We have identified 10 EWAS studies to date allow combined datasets examining DNA methylation arrays
that examined DNA methylation (Table II).24,63‑71 Recent and RNA sequencing studies, which soon may also be very
EWAS focusing on combat veterans identified evidence for well-powered. Together such approaches offer great promise
a relationship between combat trauma and PTSD symptoms, for determining the true genetic and epigenetic architecture of
which may be mediated by longitudinal changes in DNA risk vs resilience in PTSD. Such progress may afford novel
methylation.66‑68 Analysis revealed a number of gene asso- targeted therapeutic approaches to enhance treatment and
ciations to PTSD symptom severity including the following: prevention of PTSD in the aftermath of trauma. n
BRSK1, LCN8, NFG, DOCK2, ZFP57, and RNF39.66‑68 The
PGC-PTSD has also commissioned a work group to focus Disclosure/Acknowledgements: KJR supported by NIH
on building a data set to adequately power large-scale PTSD (R21MH112956, P50MH115874, R01MH094757 and
research examining DNA methylation and other epigenetic R01MH106595) the Frazier Foundation Grant for Mood and
mechanisms.70 A recent publication by the PGC-PTSD Anxiety Research and Partners Healthcare Biobank. KJR
outlines a framework for using meta-analysis with modest has received consulting income from Alkermes, and is on
sample sizes to create well-powered epigenetic associa- scientific advisory boards for Janssen, Verily, and Resil-
tion.70 Further, Uddin et al conducted a meta-analysis using ience Therapeutics. He has also received sponsored research
three civilian cohorts and identified NRG1 (cg23637605) and support from Takeda and Brainsway. None of these sources
HGS (cg19577098) as biomarkers for PTSD.71 of industry support are related to the present review.

References
1. American Psychiatric Association. Diagnostic and 5. Smoller JW. The genetics of stress-related dis- 9. Afifi TO, Asmundson GJG, Taylor S, Jang KL.
Statistical Manual of Mental Disorders. 5th ed. Wash- orders: PTSD, depression, and anxiety disorders. The role of genes and environment on trauma expo-
ington DC: American Psychiatric Publishing; 2013. Neuropsychopharmacology. 2016;41(1):297-319. sure and posttraumatic stress disorder symptoms: A
2. Kessler RC, Berglund P, Demler O, Jin R, Mer- doi:10.1038/npp.2015.266. review of twin studies. Clin Psychol Rev. 2010;30(1):
ikangas KR, Walters EE. Lifetime prevalence and 6. Van Dongen J, Slagboom PE, Draisma HHM, 101-112. doi:10.1016/j.cpr.2009.10.002.
age-of-onset distributions of DSM-IV disorders in Martin NG, Boomsma DI. The continuing value 10. Leen-Feldner EW, Feldner MT, Knapp
the national comorbidity survey replication. Arch of twin studies in the omics era. Nat Rev Genet. A, Bunaciu L, Blumenthal H, Amstadter AB.
Gen Psychiatry. 2005;62:134-147. doi:10.7312/ 2012;13(9):640-653. doi:10.1038/nrg3243. Offspring psychological and biological cor-
atwo91826-016. 7. Sartor CE, McCutcheon VV, Pommer NE, et al. relates of parental posttraumatic stress: review
3. Ozer EJ, Best SR, Lipsey TL, Weiss DS. Pre- Common genetic and environmental contributions of the literature and research agenda. Clin Psy-
dictors of posttraumatic stress disorder and symp- to PTSD and alcohol dependence in young women. chol Rev. 2013;33(8):1106-1133. doi:10.1016/j.
toms in adults: a meta-analysis. Psychol Bull. Psychol Med. 2012;41(7):1497-1505. doi:10.1017/ cpr.2013.09.001.
2003;129(1):52-73. doi:10.1037/1942-9681.S.1.3. S0033291710002072. 11. Lambert JE, Holzer J, Hasbun A. Association
4. Sheerin CM, Lind MJ, Bountress K, Nugent NR, 8. Sartor CE, Grant JD, Lynskey MT, et al. Common between parents’ PTSD severity and children’s
Amstadter AB. The genetics and epigenetics of heritable contributions to low-risk trauma, high-risk psychological distress: a meta-analysis. J Trauma
PTSD: overview, recent advances, and future direc- trauma, posttraumatic stress disorder, and major Stress. 2014;27(1):9-17. doi:10.1002/jts.21891.
tions. Curr Opin Psychol. 2016;118(24):6072-6078. depression. Arch Gen Psychiatry. 2013;69(3):293- 12. Widom CS. Posttraumatic stress disorder in
doi:10.1002/cncr.27633. 299. doi:10.1001/archgenpsychiatry.2011.1385. abused and neglected children grown up. Am J

426 • DIALOGUES IN CLINICAL NEUROSCIENCE • Vol 21 • No. 4 • 2019


Original article
Epigenetics of PTSD - Howie et al

Psychiatry. 1999;156(8):1223-1229. doi:10.1176/ transcriptional regulation in pluripotency and early 40. Gelernter J, Sun N, Polimanti R, et al.
ajp.156.8.1223. differentiation. Development. 2019;146(19). pii: Department of Veterans Affairs Cooperative
13. Molnar BE, Buka SL, Kessler RC. Child Sex- dev164772. doi:10.1242/dev.164772. Studies Program (#575B) and Million Veteran
ual abuse and subsequent psychopathology: results 28. Yehuda R, Daskalakis NP, Lehrner A, et al. Program.Genome-wide association study of
from the National Comorbidity Survey. Am J Public Influences of maternal and paternal PTSD on epi- post-traumatic stress disorder reexperiencing
Health. 2001;91(5):753-760. genetic regulation of the glucocorticoid receptor symptoms in >165,000 US veterans. Nat Neuro-
14. Brady KT, Killeen TK, Brewerton T, Lucerini S. gene in Holocaust survivor offspring. Am J Psy- sci. 2019;22(9):1394-1401. doi:10.1038/s41593-
Comorbidity of personality disorders and posttrau- chiatry. 2014;171(8):872-880. doi:10.1176/appi. 019-0447-7. Epub 2019 Jul 29. PubMed PMID:
matic stress disorder. J Clin Psychiatry. 2000;61:22- ajp.2014.13121571. 31358989.
32. doi:10.1016/j.eurpsy.2007.01.069. 29. Bierer LM, Bader HN, Daskalakis NP, et al. Ele- 41. Almli, Lynn M, Fani, Negar, Smith, Alicia K,
15. Klengel T, Dias BG, Ressler KJ. Models of vation of 11beta-hydroxysteroid dehydrogenase type Ressler KJ. Genetic approaches to understanding
intergenerational and transgenerational transmission 2 activity in Holocaust survivor offspring: evidence post-traumatic stress disorder. Intern J Neurop-
of risk for psychopathology in mice. Neuropsycho- for an intergenerational effect of maternal trauma schopharmacol. 2015;17(2):355-370. doi:10.1017/
pharmacology. 2016;41(1):219-231. doi:10.1038/ exposure. Psychoneuroendocrinology. 2014;48:1- S1461145713001090.
npp.2015.249. 10. doi:10.1016/j.psyneuen.2014.06.001. 42. Logue MW, Amstadter AB, Baker DG, et al.
16. Andero R, Ressler KJ. Fear extinction and 30. Collins LJ, Schönfeld B, Chen XS. Chapter 4 The Psychiatric Genomics Consortium Posttrau-
BDNF: Translating animal models of PTSD to - The epigenetics of non-coding RNA. In: Tollefs- matic Stress Disorder Workgroup: posttraumatic
genes. Genes Brain Behav. 2013;31(9):1713-1723. bol T, ed. Handbook of Epigenetics. San Diego, stress disorder enters the age of large-scale
doi:10.1109/TMI.2012.2196707. CA: Academic Press; 2011:49-61. doi:https://doi. genomic collaboration. Neuropsychopharmacology.
17. Korlach J, Turner SW. Going beyond five org/10.1016/B978-0-12-375709-8.00004-6. 2015;40(10):2287-2297. doi:10.1038/npp.2015.118.
bases in DNA sequencing. Curr Opin Struct Biol. 31. Qureshi IA, Mehler MF. Emerging roles of 43. Cornelis MC, Nugent NR, Amstadter AB,
2012;22(3):251-261. doi:10.1016/j.sbi.2012.04.002. non-coding RNAs in brain evolution, develop- Koenen KC. Genetics of post-traumatic stress
18. Khare T, Pai S, Koncevicius K, et al. 5-hmC in ment, plasticity and disease. Nat Rev Neurosci. disorder: Review and recommendations for
the brain is abundant in synaptic genes and shows 2012;13(8):528-541. doi:10.1038/nrn3234. genome-wide association studies. Curr Psychiatry
differences at the exon-intron boundary. Nat Struct 32. Daskalakis NP, Rijal CM, King C, Huckins Rep. 2010;12(4):313-326. doi:10.1007/s11920-010-
Mol Biol. 2012;19(10):1037-1043. doi:10.1038/ LM, Ressler KJ. Recent genetics and epigenetics 0126-6.
nsmb.2372. approaches to PTSD. Curr Psychiatry Rep. 44. Psychiatric GWAS Consortium Coordinating
19. Guo JU, Su Y, Zhong C, Ming G, Song H. 2018;20(5). doi:10.1007/s11920-018-0898-7. Committee TPG, Cichon S, Craddock N, et al.
Hydroxylation of 5-methylcytosine by TET1 33. Pasquinelli AE. MicroRNAs and their targets: Genomewide association studies: history, ratio-
promotes active DNA demethylation in the adult recognition, regulation and an emerging reciprocal nale, and prospects for psychiatric disorders. Am J
brain. Cell. 2011;145(3):423-434. doi:10.1016/j. relationship. Nat Rev Genet. 2012;13(4):271-282. Psychiatry. 2009;166(5):540-556. doi:10.1176/appi.
cell.2011.03.022. doi:10.1038/nrg3162. ajp.2008.08091354.
20. Lister R, Mukamel EA, Nery JR, et al. Global 34. Jung SH, Wang Y, Kim T, et al. Molecular 45. McGowan PO. Epigenomic mechanisms of early
epigenomic reconfiguration during mammalian brain mechanisms of repeated social defeat-induced glu- adversity and HPA dysfunction: Considerations
development. Science. 2013;341(6146):1237905. cocorticoid resistance: Role of microRNA. Brain for PTSD research. Front Psychiatry. 2013;4:1-6.
doi:10.1126/science.1237905. Behav Immun. 2015;44:195-206. doi:10.1016/j. doi:10.3389/fpsyt.2013.00110.
21. Li X, Wei W, Zhao Q-Y, et al. Neocortical bbi.2014.09.015. 46. Almli LM, Stevens JS, Smith AK, et al. A
Tet3-mediated accumulation of 5-hydroxymeth- 35. Meerson A, Cacheaux L, Goosens KA, Sapolsky genome-wide identified risk variant for PTSD is
ylcytosine promotes rapid behavioral adaptation. RM, Soreq H, Kaufer D. Changes in brain MicroR- a methylation quantitative trait locus and confers
Proc Natl Acad Sci U S A. 2014;111(19):7120-7125. NAs contribute to cholinergic stress reactions. J decreased cortical activation to fearful faces.
doi:10.1073/pnas.1318906111. Mol Neurosci. 2010;40(1-2):47-55. doi:10.1007/ Front Mol Neurosci. 2015;8:68. doi:10.1002/
22. Sutherland JE, Costa M. Epigenetics and s12031-009-9252-1. ajmg.b.32315.
the environment. Ann N Y Acad Sci U S A. 36. Schouten M, Aschrafi A, Bielefeld P, Doxakis 47. Binder EB, Bradley RG, Liu W, et al. Asso-
2003;983:151-160. E, Fitzsimons CP. microRNAs and the regulation ciation of FKBP5 Polymorphisms and childhood
23. Klengel T, Mehta D, Anacker C, et al. of neuronal plasticity under stress conditions. abuse with risk of posttraumatic stress disorder
Allele-specific FKBP5 DNA demethylation medi- Neuroscience. 2013;241:188-205. doi:10.1016/j. symptoms in adults. JAMA. 2008;299(11):1291-
ates gene– childhood trauma interactions. Nat neuroscience.2013.02.065. 1305. doi:10.1016/j.bmcl.2009.08.098.
Neurosci. 2013;16(1):33-41. doi:10.1038/nn.3275. 37. Haramati S, Navon I, Issler O, et al. MicroRNA 48. Xie P, Kranzler HR, Poling J, et al. Interaction
24. Mehta D, Klengel T, Conneely KN, et al. as repressors of stress-induced anxiety: the case of of FKBP5 with childhood adversity on risk for
Childhood maltreatment is associated with dis- amygdalar miR-34. J Neurosci. 2011;31(40):14191- post-traumatic stress disorder. Neuropsychophar-
tinct genomic and epigenetic profiles in post- 14203. doi:10.1523/JNEUROSCI.1673-11.2011. macology. 2010;35(8):1684-1692. doi:10.1038/
traumatic stress disorder. Proc Natl Acad Sci 38. Qingzhen L, Jiehua M, Zhiyang Y, Hongjun npp.2010.37.
U S A. 2013;110(20):8302-8307. doi:10.1073/ L, Chunlong C, Weiyan L. Distinct hippocampal 49. Wang Q, Shelton RC, Dwivedi Y. Interaction
pnas.1217750110. expression profiles of lncRNAs in rats exhib- between early-life stress and FKBP5 gene variants
25. Novik KL, Nimmrich I, Genc B, et al. Epig- iting a PTSD-like syndrome. Mol Neurobiol. in major depressive disorder and post-traumatic
enomics: genome-wide study of methylation phe- 2016;53(4):2161-2168. doi:10.1007/s12035-015- stress disorder: A systematic review and meta-anal-
nomena. Curr Issues Mol Biol. 2002;4(4):111-128. 9180-8. ysis. J Affect Disord. 2018;118(24):6072-6078.
26. Sardina JL, Collombet S, Tian TV, et al. 39. Guffanti G, Galea S, Yan L, et al. Genome-wide doi:10.1002/cncr.27633.
Transcription factors drive Tet2-mediated association study implicates a novel RNA gene, the 50. Hawn SE, Sheerin CM, Lind MJ, et al. GxE
enhancer demethylation to reprogram cell fate. Cell lincRNA AC068718.1, as a risk factor for post-trau- effects of FKBP5 and traumatic life events on
Stem Cell. 2018;23(5):727-741.e9. doi:10.1016/j. matic stress disorder in women. Psychoneuroendo- PTSD: A meta-analysis. J Affect Disord. 2019:455-
stem.2018.08.016. crinology. 2013;38(12):3029-3038. doi:10.1016/j. 462. doi:10.1016/j.jad.2018.09.058.
27. Gökbuget D, Blelloch R. Epigenetic control of psyneuen.2013.08.014. 51. Zhang TY, Labonté B, Wen XL, Turecki

DIALOGUES IN CLINICAL NEUROSCIENCE • Vol 21 • No. 4 • 2019 • 427


Original article
Epigenetics of PTSD - Howie et al

G, Meaney MJ. Epigenetic mechanisms for the post-traumatic stress disorder in two high-risk pop- explain the relationship between socioeconomic
early environmental regulation of hippocam- ulations. Twin Res Hum Genet. 2016;118(24):6072- position and risk of mental illness: preliminary
pal glucocorticoid receptor gene expression in 6078. doi:10.1002/cncr.27633. findings from an urban community based sam-
rodents and humans. Neuropsychopharmacology. 59. Morey RA, Garrett ME, Stevens JS, et al. ple. Biodemography Soc Biol. 2013;59(1):68-84.
2013;38(1):111-123. doi:10.1038/npp.2012.149. Genome wide association study of hippocampal doi:10.1080/19485565.2013.774627.
52. Yehuda R, Daskalakis NP, Bierer LM, et subfield volume in PTSD cases and trauma-ex- 66. Mehta D, Bruenig D, Carillo-Roa T, et al.
al. Holocaust exposure induced intergener- posed controls. bioRxiv 456988; doi:https://doi. Genomewide DNA methylation analysis in com-
ational effects on FKBP5 methylation. Biol org/10.1101/456988. bat veterans reveals a novel locus for PTSD.
Psychiatry. 2016;80(5):372-380. doi:10.1016/j. 60. Nievergelt CM, Maihofer AX, Mustapic M, et Acta Psychiatr Scand. 2017;136(5). doi:10.1111/
biopsych.2015.08.005. al. Genomic predictors of combat stress vulnera- acps.12778.
53. Logue MW, Ph D, Baldwin C, et al. A genome- bility and resilience in U.S. Marines: A genome- 67. Rutten BPF, Vermetten E, Vinkers CH, et al.
wide association study of posttraumatic stress dis- wide association study across multiple ancestries Longitudinal analyses of the DNA methylome in
order identifies the retinoid-related orphan receptor implicates PRTFDC1 as a potential PTSD gene. deployed military servicemen identify suscepti-
alpha (RORA) gene as a significant risk locus. Psychoneuroendocrinology. 2015;51(2015):459- bility loci for post-traumatic stress disorder. Mol
Mol Psychiatry. 2013;18(8):937-942. doi:10.1038/ 471. doi:10.1016/j.psyneuen.2014.10.017. Psychiatry. 2017;23(5):1145-1156. doi:10.1038/
mp.2012.113.A. 61. Stein MB, Chen C, Ursano RJ, et al. Genome- mp.2017.120.
54. Xie P, Kranzler HR, Yang C, Zhao H, Farrer wide association studies of posttraumatic stress 68. Hammamieh R, Chakraborty N, Gautam A, et al.
LA, Gelernter J. Genome-wide association study disorder in two cohorts of US army soldiers. JAMA Whole-genome DNA methylation status associated
identifies new susceptibility loci for posttraumatic Psychiatry. 2017;73(7):695-704. doi:10.1001/jama- with clinical PTSD measures of OIF/OEF veterans.
stress disorder. Biol Psychiatry. 2013;74(9):656- psychiatry.2016.0350. Transl Psychiatry. 2017;7(7):e1169. doi:10.1038/
663. doi:10.1016/j.biopsych.2013.04.013. 62. Wolf EJ, Rasmusson AM, Mitchell KS, Logue tp.2017.129.
55. Duncan LE, Ratanatharathorn A, Aiello AE, et al. MW, Baldwin CT, Miller MW. A genome-wide 69. Kuan PF, Waszczuk MA, Kotov R, et al. An
Largest GWAS of PTSD (N=20 070) yields genetic association study of clinical symptoms of dissocia- epigenome-wide DNA methylation study of PTSD
overlap with schizophrenia and sex differences in tion in a trauma-exposed sample. Depress Anxiety. and depression in World Trade Center responders.
heritability. Mol Psychiatry. 2018;23(3):666-673. 2015;31(4):352-360. doi:10.1002/da.22260.A. Transl Psychiatry. 2017;7(6):e1158. doi:10.1038/
doi:10.1038/mp.2017.77. 63. Uddin M, Aiello AE, Wildman DE, et al. Epi- tp.2017.130.
56. Ashley-Koch AE, Garrett ME, Gibson J, et al. genetic and immune function profiles associated 70. Ratanatharathorn A, Boks MP, Maihofer AX,
Genome-wide association study of posttraumatic with posttraumatic stress disorder. Proc Natl Acad et al. Epigenome-wide association of PTSD from
stress disorder in a cohort of Iraq- Afghanistan Sci U S A. 2010;107(20):9470-9475. doi:10.1073/ heterogeneous cohorts with a common multi-site
era veterans. J Affect Disord. 2016;(184):225-234. pnas.0910794107. analysis pipeline. Am J Med Genet Part B Neuro-
doi:10.1016/j.jad.2015.03.049. 64. Smith AK, Conneely KN, Kilaru V, et al. psychiatr Genet. 2017;174(6):619-630. doi:10.1002/
57. Kilaru V, Iyer S V., Almli LM, et al. Genome- Differential immune system DNA methylation ajmg.b.32568.
wide gene-based analysis suggests an association and cytokine regulation in post-traumatic stress 71. Uddin M, Ratanatharathorn A, Armstrong
between Neuroligin 1 (NLGN1) and post-traumatic disorder. Am J Med Genet Part B Neuropsychi- D, et al. Epigenetic meta-analysis across three
stress disorder. Transl Psychiatry. 2016;6(5):e820. atr Genet. 2011;156(6):700-708. doi:10.1002/ civilian cohorts identifies NRG1 and HGS as
doi:10.1038/tp.2016.69. ajmg.b.31212. blood-based biomarkers for post-traumatic stress
58. Melroy-Greif WE, Wilhelmsen KC, Yehuda 65. Uddin M, Galea S, Chang SC, Koenen KC, disorder. Epigenomics. 2018;10(12). doi:10.2217/
R, Ehlers CL. Genome-wide association study of Wildman DE, Aiello AE. Epigenetic signatures may epi-2018-0049.

428 • DIALOGUES IN CLINICAL NEUROSCIENCE • Vol 21 • No. 4 • 2019

You might also like