You are on page 1of 19

Clinical Psychology Review 77 (2020) 101830

Contents lists available at ScienceDirect

Clinical Psychology Review


journal homepage: www.elsevier.com/locate/clinpsychrev

The applied implications of epigenetics in anxiety, affective and stress- T


related disorders - A review and synthesis on psychosocial stress,
psychotherapy and prevention
Miriam A. Schielea, Michael G. Gottschalka, Katharina Domschkea,b,⁎
a
Department of Psychiatry and Psychotherapy, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Hauptstrasse 5, D-79104 Freiburg,
Germany
b
Center for Basics in NeuroModulation, Faculty of Medicine, University of Freiburg, Breisacher Straße 64, D-79106 Freiburg, Germany

HIGHLIGHTS

• Epigenetic mechanisms are biochemical DNA modifications governing gene function.


• Epigenetic alterations arise following stressful or beneficial environmental input.
• Candidate genes of a diverse range of biological processes have been evaluated.
• Epigenetic alterations are modifiable/reversible by various forms of psychotherapy.
• Prospectively, epigenetic profiles may represent trait and therapy response markers.

ARTICLE INFO ABSTRACT

Keywords: Mental disorders are highly complex and multifactorial in origin, comprising an elaborate interplay of genetic
Epigenetics and environmental factors. Epigenetic mechanisms such as DNA modifications (e.g. CpG methylation), histone
Methylation modifications (e.g. acetylation) and microRNAs function as a translator between genes and the environment.
Histone modification Indeed, environmental influences such as exposure to stress shape epigenetic patterns, and lifetime experiences
Psychiatry
continue to alter the function of the genome throughout the lifespan. Here, we summarize the recently bur-
Psychotherapy
Resilience
geoning body of research regarding the involvement of aberrant epigenetic signatures in mediating an increased
vulnerability to a wide range of mental disorders. We review the current knowledge of epigenetic changes to
constitute useful markers predicting the clinical response to psychotherapeutic interventions, and of psy-
chotherapy to alter – and potentially reverse – epigenetic risk patterns. Given first evidence pointing to a
transgenerational transmission of epigenetic information, epigenetic alterations arising from successful psy-
chotherapy might be transferred to future generations and thus contribute to the prevention of mental disorders.
Findings are integrated into a multi-level framework highlighting challenges pertaining to the mechanisms of
action and clinical implications of epigenetic research. Promising future directions regarding the prediction,
prevention, and personalized treatment of mental disorders in line with a ‘precision medicine’ approach are
discussed.

“Psychological mechanisms […] parallel their epigenetic biological and been a conceptual dichotomy of biological risk factors and pharmaco-
non-biological determinants” Louis Kaywin (1960) logical treatments on one hand and psychological mechanisms as well
as psychotherapeutic interventions on the other hand. Recent neuro-
biological research, however, provides evidence for a convergence and
1. Introduction
integration of biological and psychological mechanisms on an epige-
netic level as foreshadowed by the psychoanalyst Louis Kaywin as early
Within the classical ”nature” and ”nurture”, i.e. biological vs. non-
as in 1960: “Psychological mechanisms […] parallel their epigenetic
biological vulnerability-stress-model of mental disorders, there has
biological and non-biological determinants” (Kaywin, 1960).


Corresponding author at: Department of Psychiatry and Psychotherapy, University of Freiburg, Hauptstrasse 5, D-79104 Freiburg, Germany.
E-mail address: katharina.domschke@uniklinik-freiburg.de (K. Domschke).

https://doi.org/10.1016/j.cpr.2020.101830
Received 11 September 2019; Received in revised form 16 January 2020; Accepted 20 January 2020
Available online 04 February 2020
0272-7358/ © 2020 The Author(s). Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/BY-NC-ND/4.0/).
M.A. Schiele, et al. Clinical Psychology Review 77 (2020) 101830

Family, twin and adoption studies point to a substantial contribu- resilience has been reviewed elsewhere (Jimenez et al., 2018; Miller,
tion of genetic factors to the pathogenesis of mental disorders. 2017; Szyf, 2015), here we will exclusively focus on human studies.
Heritability rates are estimated to be moderate to high for most mental
disorders, varying between ~40–80% (Gratten, Wray, Keller, & 2. Prologue: epigenetic mechanisms
Visscher, 2014), with the remaining variance being attributed to non-
genetic influences. Molecular genetic studies have identified chromo- Epigenetic mechanisms such as DNA modifications, histone mod-
somal loci and genetic variants to be associated with disorder risk. For ifications and microRNAs are defined as biochemical alterations at the
instance, linkage studies based on family and twin data have provided level of the DNA, its 3-D structure or of regulatory small non-coding
evidence for several putative chromosomal risk loci that co-segregate RNA sequences governing gene transcription and translation, without,
with mental disorders (e.g. schizophrenia: Riley & Kendler, 2006; de- however, entailing a change in the DNA sequence itself. A key feature of
pression: Levinson, 2006; anxiety disorders: Hamilton, 2009), yet, the epigenetic modifications is their capacity of dynamic reversibility.
loci in question often span large chromosomal regions containing many Compared to the epigenetic modifications expanded upon in this sec-
potential vulnerability genes, and replication findings regarding the tion, genetic alterations of the DNA strand (that is of the very sequence
susceptibility loci described so far are scarce. In hypothesis-driven of base pairs itself) as caused by mutation, transposition and re-
candidate gene and hypothesis-generating genome-wide association combination, are seldom if ever completely reversible within a given
studies (GWAS), a wide range of single nucleotide polymorphisms cellular system. In other words, genetic modifications can be viewed as
(SNPs) has been investigated in efforts to elucidate the genetic under- comparably slow, rare and permanent, while certain epigenetic mod-
pinnings of mental disorders. Several variants in putative risk genes ifications occur fast, commonly and dynamically in response to ex-
covering a plethora of biological processes in e.g. neurotransmitter, trinsic or intrinsic biological signals and have a capacity for complete
neuropeptide, neuroimmune or neuroendocrine signaling systems have reversal. Nevertheless, at this point it should be noted that epigenetics
been related, though not unequivocally, to different disease entities. and genetics share an inseparable interdependence, insomuch as epi-
These findings emphasize the complex, highly polygenic and partly genetic modifications influence the probability of regional mutations,
overlapping genetic architecture of mental disorders (see for example and mutations themselves can alter the entire cellular epigenetic sig-
Hoehe and Morris-Rosendahl, 2018 or Smoller, 2016). nature in return (Gibbs et al., 2010; Sweder & Hanawalt, 1993). For an
In addition to genetic influences, a number of environmental factors in-depth review of mental disorder epigenetics with a special focus on
such as childhood adversity or prolonged exposure to psychosocial pathophysiology and treatment options refer to Schuebel, Gitik,
stressors (Peedicayil, 2004; Rutten et al., 2013; Teicher & Samson, Domschke, & Goldman (2016).
2013) contribute to mental disorder risk. By contrast, positive en-
vironmental influences including functional coping strategies or social 2.1. DNA modifications
support can increase resilience against mental disorders (Rutten et al.,
2013). Yet, genetic and environmental factors – both detrimental and The classical set of epigenetic DNA modifications includes me-
beneficial – do not act in isolation but rather interactively with one thylation (addition of a CH3 group) and hydroxymethylation (addition
another in conferring mental disorder risk or resilience, respectively. A of a CH2OH group) primarily targeting carbon atoms of cytosine bases
number of gene-environment interactions (G × E) underlying mental of cytosine-guanine pairs often located in so-called CpG-islands
disorders have been reported, although not all G × E have been re- (chromosomal locations rich in cytosine-guanine pairs; the “p” de-
producible (Uher, 2014), which might be due to the influence of epi- notes the phosphate group between any two given DNA bases) (for
static effects, additional mitigating environmental factors and/or epi- examples see Fig. 1). Various other forms of DNA base modifications
genetic mechanisms (Schiele et al., 2016). have been described, yet they are far less well understood in their
Epigenetic mechanisms – etymologically derived from the Greek cellular function and occur considerably less often. CpG methylation is
prefix “epi”, meaning “over” or “above” genetics – possess a central conferred by DNA-methyltransferases (DNMTs). Demethylation is in-
function at the interface between genetics and environmental influ- duced indirectly by enzymes of the growth arrest and DNA damage 45
ences. They modulate gene function, are involved in cellular differ- (Gadd45) family, which replace the methylated by an unmethylated
entiation, respond to environmental triggers, are temporally dynamic, cytosine base via base excision repair (BER), or ten-eleven-transloca-
are modifiable – and potentially reversible – by (psycho-)pharmacolo- tion (TET) proteins, which induce hydroxymethylation followed by
gical interventions, and thus may carry great therapeutic potential BER (see Fig. 1). On a functional level, the transcriptomic effect of a
(Boks et al., 2012; Szyf & Bick, 2013). given epigenetic modification is crucially influenced by its location
Epigenetic alterations have been associated with various mental within the gene structure, which is composed of regulatory elements
disorders; however, an exhaustive review of epigenetic changes on a and the gene body itself, i.e. the protein-coding exons interspersed
categorical phenotype level is beyond the scope of this article. For with non-coding introns. For example, CpG methylation in promoters,
comprehensive reviews detailing epigenetic mental disorder associa- enhancers and transcription start sites generally results in a silencing
tions studies, the reader is referred elsewhere (Abdolmaleky, Zhou, & (reduction) of transcription of the respective gene, while reciprocally
Thiagalingam, 2015; Berkel & Pandey, 2017; Gottschalk & Domschke, a reduced methylation (i.e. hypomethylation) of the same region
2016; Klengel, Pape, Binder, & Mehta, 2014; Millan, 2014; Nestler, would result in a comparably increased transcription rate (Suzuki &
Pena, Kundakovic, Mitchell, & Akbarian, 2016; Park et al., 2019; Bird, 2008). This rule of thumb is reversed as soon as intragenic, e.g.
Schiele & Domschke, 2018). Here, we place a special focus on the role exonic and intronic regions are investigated; here, increased levels of
of epigenetics as a mediator or mechanism of psychosocial stress and methylation (i.e. hypermethylation) result in a proportionally in-
psychotherapeutic interventions, thus conferring risk or resilience to- creased transcription rate and vice versa hypomethylation leads to
wards anxiety, affective and stress-related disorders by maladaptively reduced transcription levels (Suzuki & Bird, 2008). The functional
or adaptively responding to environmental influences. An improved consequences of epigenetic DNA modifications other than methylation
understanding of the possibly heritable role of epigenetics in the com- are not as well understood. CpG hydroxymethylation for example has
plex pathogenesis of anxiety, affective and stress-related disorders and been hypothesized to functionally overwrite the effect of local CpG
the elucidation of epigenetic mechanisms of psychotherapy may prove methylation, and at the same time CpG-methylation-independent ef-
useful in the development of novel preventive measures and in the fects have been described. CpG methylation to date is the most well
optimization of psychotherapy in a field with a currently still high rate researched epigenetic modification and therefore constitutes a major
of non-remission and treatment resistance. While the exhaustive lit- focus of this review, whereas other epigenetic modifications remain a
erature state of rodent studies related to the (epi)genetics of stress and central topic for future investigations.

2
M.A. Schiele, et al. Clinical Psychology Review 77 (2020) 101830

Figure 1. Epigenetic mechanisms.


Ac=Acetylation; Ph=Phosphorylation; Me=Methylation; H2A=Histone H2A; H2B=Histone H2B; H3=Histone H3; H4=Histone H4; HDAC=Histone deacetylase;
HAT=Histone acetyltransferase; C=Cytosine; G=Guanine; A=Adenine; T=Thymine; C2,5,6=carbon atoms at positions 2, 5 and 6 in pyrimidine heterocycle;
N1,3,4=nitrogen atoms at positions 1, 3 and 4 in pyrimidine heterocycle; DNMT=DNA methyltransferase; CH3=methyl group; TET proteins=Ten-eleven-trans-
location proteins; Gadd45 proteins=Growth arrest and DNA damage 45 proteins; CH2OH=Hydroxymethyl group; BER=base excision repair

2.2. Histone modifications [HDAC]) (see Fig. 1).

The packaging density of DNA around histone protein (H2A, H2B, 2.3. MicroRNAs (miRNAs)
H3, H4) octamers (together referred to as nucleosomes) can vary: ac-
tively transcribed genes are more likely to be found in “euchromatin”, Further to covalent changes to DNA and histones, recent advances in
i.e. “open chromatin”, with genes being arranged like beads on a string our understanding of transcriptome regulation have suggested small
and thus accessible to the transcription machinery, while higher con- non-coding RNAs, so called microRNAs (miRNAs), as an additional
densed states of nucleosomes (i.e. “closed” or “heterochromatin”) ra- player influencing RNA stability and translation of entire gene net-
ther impede transcription and thus lead to silencing of the respective works. Spread throughout the genome, miRNA genes do not code for a
genes involved. The packaging density is crucially determined by the specific protein, but exert important effects on the post-translational
so-called “histone code” consisting of the sum of all covalent histone regulation of gene expression (Bartel, 2004). In brief, the primary
modifications including but not limited to methylation, acetylation, miRNA is processed by nuclear RNases (a protein class cleaving RNA
phosphorylation, ubiquitination, sumoylation and ADP-ribosylation molecules) into a pre-miRNA (Gregory et al., 2004), which is exported
(Jenuwein & Allis, 2001) of amino acid residues in the histones’ tail into the cytosol, where further RNase processing occurs, until it is
regions (see Fig. 1). This effect is mainly driven by affecting the elec- turned into a targeting sequence and integrated into a RNA-multi-
tronic charge of amino acid residues in the tail regions of histones, in protein-complex (Gregory, Chendrimada, Cooch, & Shiekhattar, 2005)
return altering the histone-DNA-binding affinity and the interaction of aimed at the 3’-untranslated region (3’-UTR) of messenger RNAs
histones with other proteins involved in the silencing and activation of (mRNAs) of specific genes, thereby downregulating their translation
genes, e.g. via promoting chromatin condensation or unfolding. Despite (Lim et al., 2005).
still constituting an extensive transcriptomic research topic, basic
principles of the histone code have been established. As outlined above 3. Epigenetic “scars” of psychosocial stress
for epigenetic DNA modifications, covalent histone alterations are
conferred by various protein classes building a tripartite system, one Considerable evidence for a lasting impact of stress-associated ex-
protein to induce the histone alteration (e.g. a histone acetyltransferase periences on gene expression and behavior via epigenetic programming
[HAT]), one protein to convey the transcriptional effect (e.g. a protein emerges from a plethora of studies in preclinical models of mental
with a bromodomain, targeted at binding to acetylated histones and disorders (Alyamani & Murgatroyd, 2018; Kundakovic & Champagne,
affecting DNA accessibility and/or transcription regulation), and finally 2015; Provencal & Binder, 2015). Here, we focus exclusively on human
a protein reversing the histone alteration (e.g. a histone deacetylase studies, exemplarily compiling results from investigations of various

3
Table 1
Primary findings of epigenetic studies related to psychosocial stress
Study Sample Psychosocial stressor Gene Primary findings
M.A. Schiele, et al.

Hypothalamic–pituitary–adrenal (HPA) axis system


McGowan et al., 2009 Suicide victims (n=24) Childhood maltreatment NR3C1 ↑ NR3C1 methylation in suicide victims with maltreatment history
Healthy probands (n=12)
Tyrka et al., 2012 Healthy probands (n=99) Childhood maltreatment NR3C1 ↑ NR3C1 methylation associated with increased childhood maltreatment
Parent et al., 2017 Preschoolers (n=260) Childhood maltreatment over past NR3C1 ↑ NR3C1 methylation associated with increased childhood maltreatment over past six
six months months
↓ NR3C1 methylation change associated with increased childhood maltreatment at six-
month follow-up
Farrell et al., 2018 MDD patients (n=33) Childhood maltreatment NR3C1 ↑ NR3C1 methylation associated with increased childhood emotional abuse in MDD
Healthy probands (n=34) patients
Perroud et al., 2011 BPD patients (n=101) Childhood maltreatment NR3C1 ↑ NR3C1 methylation associated with increased childhood maltreatment in BPD patients
MDD patients (n=99) (high degree of childhood trauma) compared to MDD patients (low degree of childhood
trauma)
Radtke et al., 2015 Healthy probands (n=46) Childhood adversities NR3C1 ↑ NR3C1 methylation associated with increased number of childhood adversities
Oberlander et al., 2008 Newborns (n=85) Prenatal maternal depression and NR3C1 ↑ NR3C1 methylation associated with increased prenatal maternal depression and
anxiety symptoms anxiety symptoms
Klengel et al., 2013 Healthy probands (n=76) Childhood trauma FKBP5 ↓ FKBP5 methylation associated with increased childhood trauma
Monk et al., 2016 Healthy probands (n=61) Perceived stress during second FKBP5 ↑ FKBP5 methylation associated with increased perceived stress
trimester
Tozzi et al., 2018 MDD patients (n=56) Childhood trauma FKBP5 ↓ FKBP5 methylation associated with increased childhood trauma
Klinger-Konig et al., 2019 Community sample (n=3,965) Childhood maltreatment FKBP5 No significant interaction of FKBP5 methylation and childhood maltreatment on lifetime
MDD diagnoses

Monoaminergic and GABA-ergic systems


van IJzendoorn et al., 2010 Healthy probands (n=143) Unresolved loss and trauma SLC6A4 ↑ SLC6A4 methylation associated with lower levels of unresolved loss and trauma

4
Beach et al., 2011 Healthy probands (n=155) Sexual abuse SLC6A4 ↑ SLC6A4 methylation associated with degree of sexual abuse
Ouellet-Morin et al., 2013 Healthy monozygotic twin pairs (n=28) Bullying experiences SLC6A4 ↑ SLC6A4 methylation associated with degree of bullying
Domschke et al., 2012 PD patients (n=44) Life events during the past year MAOA ↓ MAOA methylation associated with increased negative life events
Healthy probands (n=44) ↑ MAOA methylation associated with increased positive life events
Domschke et al., 2013 PD patients (n=65) Life events during the past year GAD1 ↓ GAD1 methylation associated with increased negative life events
Healthy probands (n=65)

Neuropeptide and neurotrophic factor systems


Unternaehrer et al., 2016 Healthy infants (n=39) Stressful life events during the two OXTR ↓ OXTR methylation associated with increased stressful life events
years prior to birth
Smearman et al., 2016 Healthy probands (n=393) Childhood abuse OXTR ↑ OXTR methylation in interaction with increased childhood abuse predicted higher
ratings of anxiety and depression
Unternaehrer et al., 2015 Healthy probands (n=85) Maternal care during childhood OXTR ↑ OXTR methylation associated with lower maternal care
BDNF ↑ BDNF methylation associated with lower maternal care

MicroRNAs (miRNAs)
Wiegand et al., 2018 Healthy probands (n=24) Trier Social Stress Test - miR 21 correlated with experienced stress

Epigenome-wide association studies (EWAS)


Yang et al., 2013 Maltreated children (n=96) Childhood maltreatment EWAS 2,868 significantly differentially methylated CpGs, mainly in neoplasm-related genes
Non-maltreated children (n=96)
Mehta et al., 2013 PTSD patients with adverse childhood Adverse childhood events EWAS Significant differently methylated CpGs in genes related to development, adhesion and
events (n=32) migration of cells independent of adverse childhood events in PTSD, unique changes in
PTSD patients without adverse childhood CpGs related to nervous system development in PTSD with adverse childhood events
events (n=29)
Healthy probands with trauma exposure
outside of childhood (n=108)
Weder et al., 2014 Maltreated children (n=94) Childhood trauma EWAS ↓ Methylation in ID3, GRIN1 and TPPP related to increased depression scores,
Non-traumatized children (n=96) independent of childhood maltreatment
(continued on next page)
Clinical Psychology Review 77 (2020) 101830
M.A. Schiele, et al. Clinical Psychology Review 77 (2020) 101830

MDD=major depressive disorder; PD=panic disorder; PTSD=post-traumatic stress disorder; BPD=borderline personality disorder; NR3C1=glucocorticoid receptor gene; FKBP5=FK506 binding protein 51 gene;
SLC6A4=serotonin transporter gene; MAOA=monoamine oxidase A gene; GAD1=glutamate decarboxylase 67 gene; OXTR=oxytocin receptor gene; BDNF=brain derived neurotrophic factor gene; ID3=DNA-binding
protein inhibitor ID-3 gene; GRIN1=glutamate receptor NMDA type subunit 1 gene; TPPP=tubulin polymerization promoting protein gene; SPDEF=SAM pointed domain-containing Ets transcription factor gene;
representations of psychosocial stress associated with epigenetic al-

↓ Methylation at miR124-3 CpG site associated with increased childhood maltreatment

Post-hoc significant association of a SPDEF CpG site with the frequency of negative life
terations. In particular, we will summarize investigations of DNA me-
mainly in genes related to morphology and development and DNA and transcription

No significant epigenome-wide findings, yet top hits were primarily associated with
491 significantly differentially methylated CpGs predictive of depressive symptoms,
thylation involved in several biological systems such as the hypotha-
lamic-pituitary-adrenal (HPA) axis, a primary stress-related connection

Cumulative lifetime stress was associated with accelerated epigenetic aging

Degree of combat trauma was associated with accelerated epigenetic aging


between the CNS and the periphery, and within neurotransmitter-re-
lated signaling pathways including the monoaminergic and GABA-ergic
systems, often connected with the mechanisms of action of several
clinically available psychopharmacological compounds. Furthermore,
neuropeptide or neurotrophic factor systems, involved in inter-neuronal
and brain-body communication, as well as hypothesis-generating ap-
proaches on an epigenome-wide level will be expanded upon. Finally,
findings regarding miRNA expression and epigenetic aging associated
with psychosocial stress will be presented. For an overview of epige-
netic effects associated with psychosocial stressors, see Table 1.

3.1. Hypothalamic–pituitary–adrenal (HPA) axis system

The neuroendocrine integration provided by the HPA axis allows


methyltransferase activity

CpG=chromosomal location rich in cytosine-guanine pairs; EWAS=epigenome-wide association study; ↑=increased methylation; ↓=decreased methylation.
the body to react to various perceived forms of internal or external
stressors, regulating complex processes such as affectivity, immune
Primary findings

response and energy metabolism.


factor binding

The glucocorticoid receptor is a crucial effector of the HPA axis and


the central key component of the negative feedback loop regulating
events

cortisol secretion. The gene coding for the glucocorticoid receptor


(NR3C1; chromosomal location 5q31.3) consists of 11 exons, with the
untranslated exon 1 displaying nine identified variants, each accom-
DNA methylation-based predictor

DNA methylation-based predictor

panied by its own promoter region (1A, I, D, J, E, B, F, C, H). Most studies


focused on the promoter region of exon 1F, a particularly interesting
of age (“epigenetic clock”)

of age (“epigenetic clock”)

genomic region since it includes binding sites for the nerve growth
factor-inducible protein A (NGFI-A), which regulate NR3C1 gene ex-
pression (Weaver et al., 2007). In a seminal study, McGowan et al.
reported reduced hippocampal NR3C1 expression in an all-male post-
mortem investigation comparing 12 suicide victims with a history of
EWAS

EWAS

EWAS

EWAS

childhood abuse to 12 suicide victims without a history of childhood


Gene

abuse as well as to 12 healthy control subjects (McGowan et al., 2009).


Reduced expression was linked to increased hippocampal NR3C1 1F
Combat trauma during deployment
Stressful negative life events in the

promoter CpG methylation in suicide victims with maltreatment his-


Early life parental separation

tory. On a functional level, in vitro patch methylation mimicking this


epigenetic state resulted in decreased NGFI-A binding and corre-
Childhood maltreatment

Prenatal maternal stress

spondingly reduced NGFI-A-induced gene transcription (McGowan


Psychosocial stressor

et al., 2009). Accordingly, an investigation of the epigenetic modulation


Lifetime stressors

of NR3C1 1F promoter methylation in leukocytes of healthy probands


(n=99) revealed positive correlations between the degree of methyla-
past year

tion with relevance to NGFI-A binding and reported childhood mal-


treatment, parental loss and childhood adversity (Tyrka, Price, Marsit,
Walters, & Carpenter, 2012). Respectively, a negative correlation with
Individuals without early life separation

regard to the amount of total secreted cortisol following a dex-


EWAS meta-analysis (total n=1,740)
Individuals with early life separation

amethasone/corticotrophin-releasing hormone challenge has been de-


MDD patients with low childhood
BPD patients with high childhood

scribed (Tyrka et al., 2012). The complex nature of epigenetic mod-


Healthy adolescents (n=1,287)

ifications during early development was emphasized by a study


Healthy probands (n=392)
from their parents (n=83)

from their parents (n=83)

measuring NR3C1 promoter methylation in saliva of preschoolers


Military soldiers (n=96)
maltreatment (n=96)

maltreatment (n=93)

(n=260), who had experienced maltreatment in the past six months, at


two time points set six months apart (Parent et al., 2017). The mean
methylation of the promoter 1D region positively predicted childhood
maltreatment at baseline (also evaluated over a six month period), but
Sample

negatively predicted the change in methylation and maltreatment at


follow-up. Promoter 1F methylation predicted childhood adversity at
baseline, and methylation at CpG sites directly downstream of the
NGFI-A binding sites negatively predicted methylation change and
Rijlaarsdam et al., 2016

maltreatment at follow-up, suggesting that maltreated children display


Table 1 (continued)

increased NR3C1 promoter methylation after trauma and decreased


Khulan et al., 2014

Zannas et al., 2015


Prados et al., 2015

Epigenetic aging

Boks et al., 2015

promoter methylation at later follow-up stages (Parent et al., 2017).


Tay et al., 2019

Nevertheless, several studies have supported the notion that increased


NR3C1 methylation is associated with an increased risk of psycho-
Study

pathology, often in combination with G × E effects relating to negative


emotional experiences. DNA methylation of the NR3C1 1F promoter in

5
M.A. Schiele, et al. Clinical Psychology Review 77 (2020) 101830

whole blood samples was significantly higher in depressed individuals resulting in heightened systemic glucocorticoid resistance (Klengel
(n=33) compared to healthy probands (n=34), and mean exon 1F et al., 2013). Moreover, an evaluation of blood mRNA expression
methylation and NGFI-A binding site methylation positively correlated guided by GREs indicated that FKBP5 rs1360780 A allele carriers with
with increased levels of morning cortisol levels and average morning childhood abuse, but not without childhood abuse, displayed gluco-
cortisol output in depressed patients (Farrell et al., 2018). Furthermore, corticoid receptor resistance in transcripts representative of immune
increased NGFI-A binding site methylation was linked to the severity of and inflammatory signaling pathways, potentially reflecting a periph-
experienced emotional abuse, but not physical abuse and/or emo- eral phenotype promoting the vulnerability towards or development of
tional/physical neglect during childhood exclusively in depressed in- stress-related disorders (Klengel et al., 2013). Placental hypermethyla-
dividuals, but not in healthy probands (Farrell et al., 2018). Within a tion across NR3C1 and FKBP5 has been associated with higher second
mixed mental disorder cohort of 101 patients with borderline person- trimester perceived stress and increased anxious and depressive symp-
ality disorder (BPD) and a high degree of childhood trauma and of 99 toms, in case of FKBP5 in 61 women (Monk et al., 2016). Based on
patients with major depressive disorder (MDD) and a low degree of mediation analyses, perceived stress was associated with increased
childhood trauma, a significant link between higher peripheral blood FKBP5 methylation which in turn was associated with lower third tri-
NR3C1 exon 1F methylation and experiences of sexual abuse, and a mester fetal coupling (correlation of fetal movement and heart rate;
linear trend between the severity of sexual abuse and the extent of indicative of CNS maturation), denoting an epigenetic link between
methylation have been reported (Perroud et al., 2011). Similar asso- pregnancy stress and fetal development (Monk et al., 2016). Moreover,
ciations of higher NR3C1 exon 1F methylation levels with increased in 56 MDD patients, higher severity of childhood trauma has been
severity of childhood trauma were reported for physical abuse, physical linked to decreased whole blood FKBP5 methylation in the CpG island
neglect, emotional abuse and emotional neglect as well for the total covering the third GRE in intron 7, in carriers of the FKBP5 rs1360780
number of different types of childhood trauma (Perroud et al., 2011). A risk allele (Tozzi et al., 2018). Furthermore, lower FKBP5 intron 7
These results were further supported by an analysis of the effects of methylation was associated with reduced bilateral inferior orbitofrontal
methylation sites across NR3C1 exons 1 and 2 in 46 volunteers, high- gyrus (a cortical area implicated in response inhibition; particularly
lighting a CpG island in exon 1F as the only genomic region with a modulation and reappraisal of negative emotion) grey matter volume in
significant positive correlation between the degree of methylation and MDD patients (n=56) and controls (n=50), and with increased neu-
the number of childhood adversities on the one hand and with BPD and ronal activation during valence recognition of emotional faces in MDD
MDD symptoms on the other hand (Radtke et al., 2015). When eval- patients exclusively, linking FKBP5 demethylation with a neuronal
uated in a prediction model of future psychopathology, the number of network potentially involved in the expression of affective clinical
experienced childhood adversities, a combined principal component of symptoms (Tozzi et al., 2018). However, it should be noted that despite
NR3C1 exon 1 and 2 methylation and their interaction, all had sig- a recent replication of reduced whole blood FKBP5 intron 7 methylation
nificant predictive potential, yet interestingly, when evaluating the ef- in rs1360780 A/A genotype carriers and in comparably more depressed
fect of childhood adversity and exon 1F methylation alone on the de- individuals in a large community sample (n=3,965), main and inter-
velopment of BPD symptoms, only significant main effects, yet no action effects in combination with childhood maltreatment on lifetime
interaction effect, were found, emphasizing the complex nature of the MDD were not successfully re-established (Klinger-Konig et al., 2019),
underlying epigenetic mechanisms (Radtke et al., 2015). Notably, thus blurring the picture of FKBP5 methylation merging with early life
higher NR3C1 methylation in neonatal mononuclear cells extracted experiences as a reliable predictive biomarker of negative affective
from cord blood (n=85 newborns) at different exon 1F CpG sites was spectrum symptoms.
associated with increased prenatal maternal depression and anxiety Taken together, genes related to HPA axis regulation, especially
symptoms in the second, mid-third and third trimester (Oberlander NR3C1, are among the most promising epigenetic biomarkers, which
et al., 2008). Yet, only the heightened methylation of a CpG island co- are currently explored regarding their predictive potential at identi-
localizing with the NGFI-A binding site in newborns predicted infant fying individuals susceptible to maladaptive responses to psychological
HPA reactivity in form of an elevated cortisol release during a stress stressors, and could also serve as surrogate markers of HPA axis ac-
challenge at three months of age, hinting at the epigenetic impact of tivity, when comparing different treatment modalities (Zannas & West,
prenatal stress exposure (Oberlander et al., 2008). 2014).
Another important player of functional HPA axis regulation is coded
for by the peptidyl-prolyl cis-trans isomerase FKBP5 gene (chromo- 3.2. Monoaminergic and GABA-ergic systems
somal location 6p21.31). FKBP5 lowers the affinity of cortisol for its
receptor and hinders the translocation of the receptor-ligand-complex Amongst the most commonly investigated neurotransmitter sys-
into the nucleus, while at the same time partaking in an intracellular tems, especially due to its relation to affective and anxiety spectrum
ultra-short negative feedback loop downregulating NR3C1 activity disorders, the monoaminergic system has been a common point of focus
(Wochnik et al., 2005). Transcription of FKBP5 is regulated by mainly for epigenetic studies. The (transcriptionally) more active long allele (l)
intronic glucocorticoid-response elements (GRE) dependent on NR3C1 of the serotonin transporter linked polymorphic region (5-HTTLPR), a
binding, showcasing an intricate interplay affected by epigenetic part of the gene coding for the serotonin transporter (SLC6A4; chro-
modifications (Jaaskelainen, Makkonen, & Palvimo, 2011). A sig- mosomal location 17q11.2; regulates serotonergic signaling in the CNS
nificant interaction effect of FKBP5 rs1360780 A alleles (leading to an by sodium-dependent reuptake of serotonin from the synaptic cleft into
amplified transcription due to enabling a three-dimensional interaction the pre-synapse, therefore reducing neurotransmitter availability to
of distal GRE enhancer elements in FKBP5 intron 2 with the core pro- pre- and post-synaptic serotonin receptors), has been implicated as a
moter site, hypothesized to convey increased post-traumatic stress protective factor against MDD due to exposure to various lifetime
disorder [PTSD] risk) and early childhood trauma, independent of stressors. On an epigenetic level, higher degrees of promoter methyla-
adulthood trauma, has been reported for decreased peripheral blood tion have been related to increased experiences of unresolved loss or
methylation of GREs in intron 7 in 76 adults (Klengel et al., 2013). In trauma in l/l genotype carriers (van IJzendoorn, Caspers, Bakermans-
turn, dexamethasone exposure of proliferating, but not of differ- Kranenburg, Beach, & Philibert, 2010). In homozygote carriers of the
entiated, human hippocampal progenitor cells resulted in a stable de- less active short allele (s) - usually, but not unequivocally, implicated in
methylation of CpG sites in FKPB5 intron 7. This suggests a distinct an increased susceptibility for affective and anxiety-related traits -
window of vulnerability for relevant epigenetic modifications of the higher SLC6A4 promoter methylation was associated with lower levels
functional GREs, resulting in increased FKBP5 transcription due to of unresolved loss and trauma, indicating different molecular forms of
NR3C1 binding, hence an overreaction of the ultra-short feedback loop adjustment in risk allele carriers in 143 participants (van IJzendoorn,

6
M.A. Schiele, et al. Clinical Psychology Review 77 (2020) 101830

et al., 2010). Contrarily, an all-female study (n=155) found greater significantly negatively correlated with the proportion of exposure to
levels of SLC6A4 promoter methylation in individuals who suffered negative life events during the past year in patients and controls, pri-
from sexual abuse and significant positive correlations between 5- marily in females, and was hypothesized to convey a compensatory
HTTLPR genotypes and hypermethylation with regard to antisocial mechanism in the face of adverse environmental stimuli by raising the
personality disorder symptoms, with higher degrees of correlation with availability of the inhibitory neurotransmitter GABA following stressful
an increasing number of s alleles (Beach, Brody, Todorov, Gunter, & experiences (Domschke et al., 2013).
Philibert, 2011). A longitudinal study of 28 monozygotic twin pairs
showed that bullied twins had higher SLC6A4 promoter methylation 3.3. Neuropeptide and neurotrophic factor systems
levels at age ten than non-bullied twins, while notably prior to any
bullying experience at age five methylation levels were indistinguish- The nonapeptide oxytocin, which is released by the neurohypo-
able, as were child-specific family environments and individual risk physis, regulates smooth muscle tonus during parturition and lactation,
factors prior to or concomitant to bullying (Ouellet-Morin et al., 2013). and has been shown to influence complex social cognition and affilia-
Overall methylation changes between the ages of five and ten were tive behavior with a high relevance to anxiety-related phenotypes (for
exclusively based on a single CpG island, at which methylation levels review see Gottschalk & Domschke, 2018).
also displayed a negative correlation with the cortisol response to a Methylation of the transcription-regulating exon 3 of the oxytocin
psychological stress challenge, supporting the hypothesis that the ser- receptor gene (OXTR, chromosomal location 3p25.3) in cord blood of
otonergic and HPA systems display a functional epigenetic connection 39 infants was negatively associated with the occurrence of stressful life
(Ouellet-Morin et al., 2013). events during the two years prior to birth, postpartum depression in-
However, attention has not solely been focused on the SLC6A4; the tensity, and maternal cortisol awaking response and diurnal cortisol
monoamine oxidase A gene (MAOA, chromosomal location Xp11.3), profile during the second trimester of pregnancy, independent of po-
whose protein product catalyzes the oxidative deamination of amines tentially confounding sociodemographic and birth-related factors
(e.g. 5-hydroxytryptamine, norepinephrine and epinephrine) and is (Unternaehrer et al., 2016). This suggests that prenatal adversity might
therefore important in the metabolism of neuro- and vaso-active trigger an epigenetic adaption in a gene with known relevance for
amines, has also been evaluated epigenetically. The MAOA gene is childbirth and mother-child bonding via increased expression of OXTR
commonly investigated for a functional genetic variation in its pro- (Unternaehrer et al., 2016). Moreover, it has been described that ex-
moter region, i.e. the upstream variable number of tandem repeats periences of childhood abuse lead to increased methylation down-
(uVNTR) consisting of “protective” short (transcriptionally less active) stream of exon 3, supporting the notion of differential effects of pre-
and “risk” long (transcriptionally more active) alleles predisposing for and postnatal stressors, while only an interaction of environmental
instance to panic-related phenotypes (Howe et al., 2016; Reif et al., trauma and methylation downstream of exon 3 and within exon 1
2012). The epigenetic signature of MAOA, in particular of CpG islands predicted adult psychopathological symptoms in 393 adult participants
flanking the uVNTR and covering the first exon, has also been in- (Smearman et al., 2016). Hypomethylation within exon 1 and hy-
vestigated with respect to stress-associated experiences. Comparing 44 permethylation downstream of exon 3 in combination with severe
female patients with panic disorder with 44 mentally healthy control childhood abuse was associated with higher adult ratings of anxiety and
probands showed a significantly lower average MAOA methylation in depression symptoms (Smearman et al., 2016). In line with these
panic disorder, principally in CpG islands downstream of the uVNTR findings on negative post-natal experiences, increased OXTR exon 3
and within the first exon (Domschke et al., 2012). While methylation methylation was furthermore linked to low reported maternal care
was not affected by age, smoking status or selective serotonin-reuptake during childhood, especially in males, as was hypermethylation of exon
inhibitor (SSRI) medication, the long uVNTR variants were associated 6 of the brain derived neurotrophic factor gene (BDNF, chromosomal
with increased overall methylation in healthy controls (Domschke location 11p14.1) (Unternaehrer et al., 2015). BDNF is a member of the
et al., 2012). Most strikingly, across female panic disorder patients and nerve growth factor family that binds to membrane-bound tyrosine
controls the proportion of negative life events during the past year was receptor kinases and promotes differentiation and survival of peripheral
significantly negatively correlated with CpG island methylation down- and central neuronal cell populations, influences axonal and dendritic
stream of the uVNTR and within the first MAOA exon (Domschke et al., growth and morphology, and regulates synaptic transmission and
2012). The proportion of positive recent life events was significantly plasticity.
positively correlated with methylation at both genomic sites, pointing In synopsis, these findings point towards early life stress to be ac-
towards a role of MAOA hypomethylation as an epigenetic correlate of companied by widespread epigenetic modifications across different
susceptibility-increasing environmental experiences in the etiology of neuropeptide and neurotrophic factor systems. Interestingly, psycho-
panic disorder (Domschke et al., 2012). social stress has been demonstrated to significantly increase whole
Notably, the majority of findings relating to the epigenetics of blood methylation of OXTR exon 3 in an acute post-stress phase (15
psychosocial stressors and monoaminergic neurotransmission point minutespast challenge) and to significantly decrease methylation, even
towards a hypermethylation of SLC6A4 and a hypomethylation of below the point of pre-stress baseline methylation, in a follow-up phase
MAOA, mirroring the results observed in categorical analyses of MDD (90 minutespast challenge) in 76 adults (Unternaehrer et al., 2012). No
patients (Bakusic, Schaufeli, Claes, & Godderis, 2017; Schiele et al., such changes were detectable for BDNF exon 6 methylation, high-
2019), further stressing the need for longitudinal study designs evalu- lighting the importance to evaluate the short-term and long-term tem-
ating methylation states over the disease course of affective disorders. poral dynamics of epigenetic modifications in light of potentially ad-
Besides the accumulating evidence for epigenetic changes in ser- verse environmental influences (Unternaehrer et al., 2012).
otonin signaling and metabolism, other neurotransmitter systems have
been implicated in mediating the influence of external stressors on the 3.4. MicroRNAs (miRNAs)
underlying pathophysiology of mental disorders. For example, average
methylation across CpG islands in the promoter region and the second Moreover, a recent investigation of stress-associated miRNAs in
intron of the GAD1 gene (chromosomal location 2q31.1) coding for the saliva found significant changes in miR 20b, miR 21 and miR 26b be-
glutamate decarboxylase 67, which catalyzes the pyridoxal phosphate- fore, during and after a psychosocial stress challenge (Trier Social Stress
dependent decarboxylation of glutamate to GABA, was found to be Test) in 24 healthy participants (Wiegand et al., 2018).: Concentrations
lower in panic disorder patients compared to healthy controls (n=65 of miR 21 correlated positively with experienced stress over the course
each), with a particularly strong effect in the promoter region of the challenge, while miR 21 and miR 26b concentrations correlated
(Domschke et al., 2013). This GAD1 promoter hypomethylation was positively with the secreted amount of the salivary stress marker alpha-

7
M.A. Schiele, et al. Clinical Psychology Review 77 (2020) 101830

amylase, suggesting an integration of miRNA-related processes into inactivated protein 1 (LGI1, chromosomal location 10q23.33) and 2
sympathetic stress regulation and offering a non-invasive way to de- (LGI2, chromosomal location 4p15.2) in addition to other genes related
termine a dynamic acute stress indicator (Wiegand et al., 2018). to brain functioning. Pathway analysis revealed an overrepresentation
of hypomethylated genes representing biological processes related to
3.5. Epigenome-wide association studies (EWAS) morphology and development, as well as DNA and transcription factor
binding (Khulan et al., 2014).
Similar to the non-hypothesis driven approach followed in genome- A surprising result arose from an EWAS investigation comparing an
wide association studies (GWAS), epigenome-wide association studies all-female set of patients with BPD (n=96) and a history of high
(EWAS) utilize high-throughput methylation profiling techniques, al- childhood adversity to patients with MDD (n=93) reporting low rates
lowing for the evaluation of thousands of CpG sites simultaneously in of childhood maltreatment: comparison of BPD to MDD and low to high
discovery approaches with ever-growing sample sizes aimed at the childhood trauma severity in multi- and univariate analyses highlighted
identification of novel epigenetic hotspots in relation to various phe- a CpG site close to miR 124-3, where hypomethylation was linked to
notypes of interest. BPD and increased childhood trauma (Prados et al., 2015). This result
An EWAS comparing maltreated children (n=96) removed from was validated via pyrosequencing and hypothesized to reflect the cri-
their parents by protective services with demographically matched tical role of neuronal development and HPA axis regulation under
children (n=96) found 2,868 significantly differentially methylated epigenetic control, given that miR 124-3 targets NR3C1 and the mi-
saliva CpG sites mainly situated in promoter regions and gene bodies. neralocorticoid receptor (NR3C2) (Prados et al., 2015).
When comparing the results to a pathway database of disorder-asso- Strikingly, a meta-analysis of two large EWAS sample sets (total
ciated biomarkers, authors discerned a significant enrichment for epi- n=1,740) found no epigenome-wide significant CpG methylation hit in
genetic changes in genes related to a diverse range of neoplasms in- cord blood extracted DNA associated with prenatal maternal stress
cluding lung cancer, colorectal cancer and breast cancer (Yang et al., (Rijlaarsdam et al., 2016). A follow-up analysis of the top EWAS meta-
2013). A unique experimental design accessing gene expression and analysis hits (p<.0001) in pathway databases indicated methyl-
EWAS data from PTSD patients with (n=32) and without (n=29) ad- transferase activity as the single most overrepresented molecular
verse childhood events compared to controls with trauma exposure function. A clustering approach aimed at identifying differentially
outside of their childhood (n=108) found that gene expression profiles methylated regions found no Bonferroni-corrected cluster associated
differed widely for both disorder groups (2% overlap) indicating dis- with prenatal maternal stress, underscoring the clinical need for larger
tinct PTSD profiles in the presence or absence of childhood trauma sample sizes in order to reach the statistical power to detect robust
(Mehta et al., 2013). For 94% of the loci, the direction of the change EWAS results (Rijlaarsdam et al., 2016).
predicted by differences in methylation agreed with the change ob- A large EWAS in 1,287 adolescents found post-hoc significant po-
served in gene expression, i.e. methylated CpGs close to the transcrip- sitive correlation of a CpG island within the promoter of the SAM
tion start site predicted reduced transcription. Furthermore, about two pointed domain-containing Ets transcription factor (SPDEF, chromo-
thirds of transcripts representative of PTSD with childhood abuse dis- somal location 6p21.31) with the frequency of negative stressful life
played methylation changes, but only one third of transcripts was re- events in the past year (Tay et al., 2019). Moreover, a significant in-
presentative of PTSD without childhood abuse, indicating that changes teraction effect between the frequency of stressful life events in the past
in gene expression are more prominently linked to changes in corre- year and increased methylation was discerned to result in an higher
sponding DNA methylation in the presence rather than the absence of incidence of lifetime binge drinking behavior. A methylation-expression
childhood abuse in PTSD (Mehta et al., 2013). Remarkably, evaluation quantitative trait loci (eQTL) analysis suggested that methylation would
of the associated biological processes affected by methylation indicated lead to expression changes in multiple trans-genes enriched for genes
an overlap related to the development, adhesion and migration of cells, previously linked to opioid-related disorders, alcoholism and tobacco
whereas in PTSD with childhood abuse unique nervous system devel- use, providing converging evidence for a novel epigenetic risk marker
opment processes were overrepresented. Conversely, PTSD without of adolescent substance abuse integrating the molecular effects of
abuse history distinctly displayed apoptosis processes, possibly re- psychosocial stress (Tay et al., 2019).
flecting differences in PTSD pathophysiology based on past experiences Due to small effect sizes more recent approaches with samples in the
of maltreatment (Mehta et al., 2013). thousands can be expected to return epigenome-wide significant find-
An EWAS in maltreated (n=94) vs. non-traumatized children ings related to psychosocial stressors which prove robust to replication
(n=96) analyzing DNA methylation as a predictor of dimensional de- attempts. Additionally, novel hits are expected to emerge with advances
pression ratings discovered three significant CpG sites within the genes in throughput and sequencing techniques. EWAS constitute a promising
coding for the DNA-binding protein inhibitor ID-3 (ID3, chromosomal field for future studies, especially given that they produce a quantitative
location 1p36.12), the inotropic glutamate receptor NMDA type subunit outcome measure, which makes them particularly attractive for statis-
1 (GRIN1, chromosomal location 9q34.3), and the tubulin poly- tical approaches in combination with dimensional phenotypes.
merization promoting protein (TPPP, chromosomal location 5p15.33),
with lower methylation correlating with increased depression scores 3.6. Epigenetic aging
(Weder et al., 2014). All three CpG sites were predictors independent of
interaction effects with childhood maltreatment, yet the ID3 CpG site Composite epigenomic markers such as a DNA methylation-based
showed an interaction effect with childhood maltreatment on cortisol predictor of age (“epigenetic clock”; Horvath, 2013) have attracted
secretion, with increased methylation predicting decreased diurnal great interest with regard to both stress and mental health. Accelerated
cortisol secretion in controls, yet increased cortisol secretion in mal- epigenetic aging has been shown to be conferred by cumulative, par-
treated children (Weder et al., 2014). ticularly personal lifetime stress in a cohort of highly traumatized
Comparing 83 males exposed to early life stress in form of separa- African-Americans (Zannas et al., 2015), and 6-month exposure of male
tion from their parents to 83 non-separated controls, no epigenome- soldiers to combat deployment in Afghanistan (Boks et al., 2015). In-
wide changes were detected. However, when evaluating the develop- creased epigenetic aging has also found to be associated with the ca-
ment of depressive symptoms over a 5-10 year period significant as- tegorical phenotype of major depression (Han et al., 2018). These
sociations for 491 CpG probes in 351 different genes were discovered findings suggest epigenetic aging as a possible mechanistic link be-
(Khulan et al., 2014). Among the most significant findings were hy- tween the experience of psychosocial stress and aging-related disease
pomethylations in the genes coding for the leucine-rich glioma- phenotypes (see Zannas, 2016).

8
M.A. Schiele, et al. Clinical Psychology Review 77 (2020) 101830

4. Transgenerational transmisson of stress via epigenetic responsiveness to environmental input as outlined above, recent re-
mechanisms? search has begun to place a particular focus on epigenetic changes in
relation to the effects of psychotherapy by examining the potential of
Preclinical studies suggest a potential intergenerational transmis- epigenetic patterns in predicting the therapeutic outcome, as well as
sion of epigenetic information as an engram of environmental experi- temporal modifications as possible mechanistic correlates of clinical
ences via multiple mechanisms conferred by paternal sperm and ma- changes occurring over the course of psychotherapeutic or – to a lesser
ternal germline to shape endocrine programming, brain development extent – also preventive interventions. For an overview of epigenetic
and the behavioral phenotype in the offspring (Babenko, Kovalchuk, & effects accompanying treatment response, see Table 2.
Metz, 2015; Bale, 2014; Dias & Ressler, 2014; Franklin et al., 2010;
Klengel, Dias, & Ressler, 2016; Nagy & Turecki, 2015; Rodgers, Morgan, 5.1. Hypothalamic–pituitary–adrenal (HPA) axis system
Bronson, Revello, & Bale, 2013; Stenz, Schechter, Serpa, & Paoloni-
Giacobino, 2018; Yeshurun & Hannan, 2019). It has to be noted, In 16 patients with PTSD, higher NR3C1 exon 1F methylation levels
however, that to date evidence from human studies is merely correla- prior to a 12-week prolonged exposure intervention were found to be
tional as detailed below. predictive of improved treatment response (greater changes in clin-
In a study examining 25 women exposed to the Tutsi genocide in ician-rated and self-reported symptom severity from pre- to post-
Rwanda during pregnancy and their children in comparison with 25 treatment) and lower post-treatment symptoms as well as of lower self-
non-exposed Rwandan control mothers and their offspring, Perroud reported (but not clinician-rated) symptom severity at follow-up
et al. (2014) observed higher rates of PTSD and depression symptoms in (Yehuda et al., 2013).
mothers exposed to the genocide as well as in their children. In addi- In a total of 16 combat veterans with PTSD, FKBP5 exon 1 promoter
tion, methylation of the NR3C1 exon 1F promoter region was increased methylation – although not predictive of treatment outcome per se –
in exposed compared to non-exposed mothers. Likewise, higher NR3C1 was found to decrease in responders to a 12-week prolonged exposure
methylation was also observed in children of genocide-exposed mothers psychotherapy (n=8) during the recovery period from post-treatment
relative to control children. In a retrospective study in 25 mothers ex- to follow-up (Yehuda et al., 2013). In another study, following mind-
posed to intimate partner violence during pregnancy and their children fulness-based stress reduction in 22 patients with PTSD, treatment re-
(n=25), NR3C1 promoter methylation in children as assessed during sponders (n=11) were characterized by a decrease in FKBP5 intron 7
adolescence was positively associated with maternal exposure to vio- methylation, whereas methylation increased in non-responders. Fur-
lence during pregnancy (Radtke et al., 2011). In 32 Holocaust survivors thermore, changes in PTSD symptoms from pre- to post-therapy were
and their 22 adult offspring compared to eight non-exposed control inversely correlated with methylation change in the investigated region
parents and their nine children, parental Holocaust exposure was found (Bishop et al., 2018).
to predict FKBP5 intron 7 methylation in offspring. Of note, increased In a sample of 111 adult patients diagnosed with agoraphobia with/
whole blood CpG methylation was observed in the third (of a total of without panic disorder or specific phobia treated with disorder-specific
three) GRE of FKBP5 intron 7 in Holocaust survivors compared to cognitive behavioral therapy (CBT), pre- to post-treatment percentage
contemporary Jewish controls, while children of Holocaust survivors changes in FKBP5 methylation at a single CpG site in intron 7 was in-
were characterized by decreased methylation compared to control versely related to changes in clinical severity observed at follow-up. The
children, which may possibly reflect a biological adaptation in response pre- to post-methylation change at one CpG site in the promoter region
to altered glucocorticoid levels (Yehuda et al., 2016). Furthermore, was furthermore nominally significantly associated with post-treat-
offspring carrying the FKBP5 rs1360780 A risk allele showed a sig- ment, but not follow-up, symptom severity. Moreover, a trend for an
nificant negative correlation between the degree of physical and sexual allele-specific methylation change from pre- to post-therapy at one CpG
childhood abuse and CpG methylation across the first GRE in intron 7, site located in the promoter region to be positively related to outcome
while G/G genotype carriers displayed a positive correlation, sug- at follow-up emerged in FKBP5 rs1360780 A risk allele carriers (Roberts
gesting a potential site-specific epigenetic response based on either et al., 2019). Enhanced treatment response has also been associated
personal childhood or parental trauma (Yehuda et al., 2016). Methy- with nominal increases in FKBP5 exon 1 methylation changes from pre-
lation of the third GRE in intron 7 was further positively correlated with to post-CBT at one CpG site in 98 children and adolescents with mixed
methylation at the same site in their offspring and overall average anxiety disorders, but again only in FKBP5 rs1360780 A risk allele
methylation of intron 7 was negatively correlated with wake-up cor- carriers (Roberts et al., 2015).
tisol, indicating a potential functional relevance to HPA axis regulation
(Yehuda et al., 2016). 5.2. Monoaminergic and GABA-ergic systems
In 21 families with school-age children, parental psychological
distress severity was found to be predictive of methylation status at In 116 children with mixed anxiety disorders following CBT, treat-
three CpG sites located in the dopamine transporter (DAT; chromo- ment non-responders were characterized by a decrease in methylation
somal location 5p15.33) promotor region in their children. Paternal – as driven by a single CpG site in the promoter region of the SLC6A4
but not maternal – methylation at three CpG sites were associated with gene post-treatment, while in turn, treatment responders – although not
methylation at three, albeit different, CpG sites in children (Cimino statistically significant – displayed a small increase in methylation at
et al., 2017). the same site on a descriptive level. Of note, patients who showed
While the familiality of mental disorders has been well established continued symptom improvement six months after psychotherapy ces-
and associations between the exposure to stressful experiences and sation were characterized by a significant increase in average SLC6A4
trauma with epigenetic alterations have been described extensively in promoter methylation relative to those who did not improve or wor-
the literature (see above), it has yet to be conclusively established if – sened at follow-up (Roberts et al., 2014).
and how – stress-induced epigenetic alterations can be passed on to A recent study on the effect of a mindfulness-based preventive in-
subsequent generations (Jawaid, Roszkowski, & Mansuy, 2018; tervention in medical students preparing for a stressful exam, however,
Youssef, Lockwood, Su, Hao, & Rutten, 2018). the effectiveness of the preventive measure, i.e. stress reduction, went
along with a decrease in SLC6A4 methylation (Stoffel et al., 2019).
5. Epigenetics: markers and mechanisms of psychotherapy and In 28 female patients with panic disorder in comparison to matched
prevention? healthy controls, DNA methylation in a region spanning exon 1/intron
1 of the MAOA gene was shown to significantly increase in treatment
In light of the dynamic nature of epigenetic modifications and their responders following a six-week exposure-based CBT, whereas

9
M.A. Schiele, et al. Clinical Psychology Review 77 (2020) 101830

Table 2
Primary findings of psychotherapyepigenetic studies
Study Diagnosis Cases Therapy form Therapy duration Gene Primary findings

Yehuda et al., 2013 PTSD n=16 PE 12 weeks NR3C1 ↑ NR3C1 baseline methylation in responders (n=8) post-PE
3-month FU FKBP5 ↓ FKBP5 methylation in responders (n=8) at FU
↑ FKBP5 methylation in non-responders (n=8) at FU
Bishop et al., 2018 PTSD n=22 MBSR 9 weeks FKBP5 ↓ Methylation in responders (n=11) post-MBSR
↑ Methylation in non-responders (n=11) post-MBSR
Roberts et al., 2019 AG, AG+PD, n=111 CBT Variable/not FKBP5 ↓ Methylation associated with higher symptom improvement post-
SpP specified CBT
6-month FU
Roberts et al., 2015 Mixed AD n=98 CBT Variable/not FKBP5 ↓ Methylation associated with greater reduction in symptom severity
specified at FU in FKBP5 rs1360780 T allele carriers
6-month FU
Roberts et al., 2014 Mixed AD n=116 CBT Variable/not SLC6A4 ↓ Methylation in non-responders post-CBT
specified ↑ Methylation in responders at FU
6-month FU
Ziegler et al., 2016 PD n=28 CBT 6 weeks MAOA ↑ Methylation in responders (n=11) post-CBT
Schiele et al., 2018 SpP n=28 CBT (VR) 2 weeks MAOA ↑ Methylation post-CBT
Perroud et al., 2013 BPD n=167 DBT 4 weeks BDNF ↓ Methylation in responders (n=115) post-DBT
↑ Methylation in non-responders (n=52) post-DBT
Thomas et al., 2018 BPD n=26 DBT 12 weeks BDNF ↓ Methylation post-DBT
Knoblich et al., 2018 BPD n=24 DBT 12 weeks APBA3 No methylation change post-DBT
MCF2 ↑ Baseline methylation in responders (n=7)
Kahl et al., 2016 MDD n=37 CBT 6 weeks SLC2A1 ↓ Methylation in remitters (n=18)
Ziegler et al., 2019 PD n=47 CBT 6 weeks EWAS ↑ IL1R1 methylation post-CBT
Vinkers et al., 2019 PTSD n=44 tfCBT (partially with 6-8-month FU EWAS Changes in 12 DMRs
EMDR) ↑ ZFP57 methylation in responders (n=11) at FU

PTSD=post-traumatic stress disorder; AG=agoraphobia; PD=panic disorder; SpP=specific phobia; AD=anxiety disorder; BPD=borderline personality disorder;
MDD=major depressive disorder; PE=prolonged exposure therapy; CBT=cognitive behavioral therapy; VR=virtual reality; MBSR=mindfulness-based stress re-
duction; DBT=dialectic behavioral therapy; tfCBT=trauma-focused psychotherapeutic intervention; FU=follow-up; NR3C1=glucocorticoid receptor gene;
FKBP5=FK506 binding protein 51 gene; SLC6A4=serotonin transporter gene; MAOA=monoamine oxidase A gene; BDNF=brain derived neurotrophic factor gene;
APBA3=amyloid beta precursor protein binding family A member 3 gene; MCF2=MCF.2 cell line derived transforming sequence gene; SLC2A1=solute carrier
family 2 member 1 gene; IL1R1=interleukin 1 receptor type 1 gene; ZFP57=zinc-finger protein 57 gene; EWAS=epigenome-wide association study;
DMR=differentially methylated region; ↑=increased methylation; ↓=decreased methylation.

methylation levels remained unchanged or even decreased in non-re- – was significantly related to favorable response to DBT in 24 patients
sponders. Of note, following therapy DNA methylation levels in CBT with BPD (Knoblich et al., 2018).
responders no longer differed from those in healthy controls (Ziegler Hypermethylation of the solute carrier family 2 member 1
et al., 2016). Similarly, in 28 female patients with acrophobia, sig- (SLC2A1, chromosomal location 1p34.2) promoter region in 37 patients
nificant increases in average MAOA methylation as well as at four single with MDD was found to decrease in treatment remitters compared to
CpG sites - conferring decreased MAOA expression in luciferase-based non-remitters after six weeks of inpatient treatment with CBT (Kahl
reporter gene assays - were observed following a two-session exposure- et al., 2016).
based treatment in virtual reality, and to correlate with improvements
in clinical symptoms (Schiele et al., 2018). 5.5. Epigenome-wide association studies (EWAS)

5.3. Neuropeptide and neurotrophic factor systems A longitudinal epigenome-wide study assessed changes on the DNA
methylome level corresponding to clinical effects of manualized six-
In 115 patients with BPD, following a four-week course of intensive week CBT in panic disorder using the Illumina MethylationEPIC
dialectic behavior therapy (I-DBT) treatment non-responders were BeadChip. Suggestive evidence was discerned for an increase in me-
found to increase in methylation at BDNF exons 1 and 4, while, in turn, thylation directly after CBT at cg06943668 in the interleukin 1 receptor
treatment responders decreased in methylation. Alterations in methy- type 1 (IL1R1) gene in treatment responders suggesting a potentially
lation were furthermore found to be related to changes in depressive immunomodulatory effect of successful CBT (Ziegler et al., 2019). In
symptom severity, hopelessness and impulsivity (Perroud et al., 2013). PTSD patients, response to trauma-focused CBT partially supplemented
In 41 patients with BPD, BDNF exon 4 hypermethylation was discerned by Eye Movement Desensitization and Reprocessing (EMDR) has been
in saliva, but not in blood, as compared to healthy controls. BDNF observed to be accompanied by dynamic alterations in 12 differentially
methylation in saliva was found to decrease following a 12-week dia- methylated genomic regions (DMRs) six to eight months after treat-
lectic behavioral therapy (DBT) program in a subsample of 26 BPD ment, with most consistent evidence for increasing methylation of the
patients, although changes in methylation did not correlate with zinc-finger protein 57 (ZFP57) genomic region along with successful
symptom improvement (Thomas et al., 2018). treatment (Vinkers et al., 2019).

5.4. Candidate genes related to other biological systems 5.6. Epigenetic aging

Pre-treatment increased methylation of the amyloid beta precursor The effects of stress on the composite epigenetic marker of “epige-
protein binding family A member 3 (APBA3, chromosomal location netic aging” as reviewed above might be preventable by targeted be-
19p13.3) and the MCF.2 cell line derived transforming sequence havioral interventions or resilience-increasing environments: While
(MCF2, chromosomal location Xq27.1) genes – both previously asso- depressive symptoms of parents living in the rural southeastern United
ciated with BPD in an epigenome-wide approach (Teschler et al., 2013) States, scored when their children were 11 years old, were found to be

10
M.A. Schiele, et al. Clinical Psychology Review 77 (2020) 101830

predictive of accelerated epigenetic aging in the offspring at age 20, the majority of PTSD-associated CpG sites are shared by all primates,
participation in a family-centered prevention training (Strong African while PTSD-associated CpG sites proximal to immune system-related
American Families [SAAF] program) seems to have prevented such genes may have disproportionately evolved during the more recent
alterations in youths. This protective effect on epigenetic aging in the evolutionary history.
intervention group could be attributed to reductions in harsh parenting
from age 11 to age 16 (Brody, Yu, Chen, Beach, & Miller, 2016). Also, 6.2. Epigenetics as a switch between risk and resilience?
while exposure to higher levels of racial discrimination in African
American youth from rural Georgia, USA, was associated with increased Regarding the role of epigenetic mechanisms in contributing to a
epigenetic aging, supportive family environments prevented ac- risk-increasing status towards mental disorders, there is no doubt that
celerated epigenetic aging despite exposure to racial discrimination environmental influences such as early, even prenatal trauma shape
(Brody, Miller, Yu, Beach, & Chen, 2016). epigenetic patterns and that life experience continues to alter the
function of the genome throughout the lifespan (see Fig. 2). As re-
6. Discussion and future directions viewed above (see section 3, ’Epigenetic ”scars“ of psychosocial stress’),
epigenetic changes in candidate genes including NR3C1, FKBP5,
6.1. Epigenetics at the interface between genes and environment in mental SLC6A4 and BDNF have repeatedly been associated with stress experi-
disorders ences and may thus constitute the most promising dynamic markers of
stress to date.
Mental disorders are highly complex and multifactorial in origin, The biochemical mechanisms translating input from, broadly, the
comprising an elaborate interplay of genetic and environmental (G × environment into epigenetic changes are not yet completely under-
E) factors that act on a number of biological systems, and thus require a stood, however, one lead points to a process involving glucocorticoids
framework that includes multiple levels of analysis. Epigenetic me- as markers of HPA axis activation following stress exposure to bind to
chanisms function as a translator between genes and the environment glucocorticoid receptors (GRs) in the cell, which then function as
and add yet another level of intricacy in the discussion of differential transcription factors in the nucleus and initiate a host of transcriptomic
susceptibility rather than vulnerability. The “differential susceptibility modifications resulting in enduring change of the epigenetic machinery
hypothesis” (Belsky & Pluess, 2009) entails a conceptualization of (Zannas & Chrousos, 2017). However, given the complex-genetic nature
plasticity genes rather than risk genes insofar as a given genotype of mental disorders, the exact mechanisms brokering the modulation of
promotes increased sensitivity to environmental conditions in general epigenetic signatures and their downstream consequences in response
and, depending on the nature of the environment, can result in bene- to environmental influences towards an increased risk for mental dis-
ficial or detrimental outcomes. Accordingly, experiences of stress and orders are yet to be conclusively determined and likely involve the
adversity do not necessarily result in a heightened susceptibility for intricate interplay between different neurotransmitter (e.g. mono-
mental disorders and, eventually, manifest pathology, but can convey aminergic and GABA-ergic, see above), neuropeptide, neurotrophic and
resilience. This phenomenon might be conferred by epigenetic me- other systems.
chanisms governing the functionality of putative “risk” genes de- Although research has mostly focused on the negative consequences
pending on environmental constellations. of stress exposure, changes in epigenetic signatures are suggested to
In this context, it is important to note that “environmental influ- equally reflect the biological impact of not only detrimental but also
ences” should be understood as an umbrella term encompassing any beneficial environmental influences, thus possibly contributing to an
and all input present in and arising from the environment and are not increased resilience towards mental disorders (see Fig. 2). For instance,
limited to the repercussions of explicitly adverse environmental insults. augmented maternal care has been shown to exert a preventive effect
Rather, environmental input equally entails the presence of beneficial by increasing resilience via epigenetic changes in rodents, resulting in
factors, for example related to interpersonal aspects (e.g. attachment, reduced depression- and anxiety-related behaviors in response to stress
social support, friendship/relationship quality and stability). Moreover, (Singh-Taylor et al., 2018) (for a review of animal models of epigenetic
psychosocial factors (e.g. financial status) and lifestyle aspects such as mechanisms conferring resilience towards mental disorders see Dudley,
bodily activity and exercise, diet, smoking behavior, or substance abuse Li, Kobor, Kippin, & Bredy, 2011). Accordingly, secure attachment as
have been linked to mental disorders one the one hand, and epigenetic compared to disorganized attachment in human infants accounted for
changes on the other. Finally, various characteristics of the surrounding 11.9% of the variation in genome-wide DNA methylation, which might
physical environment such as exposure to radiation, toxins, climate and contribute to differential resilience against mental disorders (Garg
seasonal change are capable of inducing a wide range of epigenetic et al., 2018). In particular, lower DNA methylation levels of the gene
modifications (see Kanherkar, Bhatia-Dey, & Csoka, 2014). coding for the oxytocin prepropeptide (OXT) have been linked to more
From an evolutionary perspective, adaptability to psychosocial secure attachment styles along with a superior ability to correctly re-
stressors and the environment as a whole might – apart from random cognize emotional facial expressions and thus potentially mediate
mutations, chromosomal and DNA sequence alterations generating higher resilience against mental disorders characterized by various
phenotypic variation and thus contributing to natural selection as as- forms of decreased social functioning (Haas et al., 2016). This notion
sumed in a neo-Darwinian model – also be conferred by epigenetic was further supported by a study reporting decreased OXTR methyla-
mechanisms. By promoting genetic mutations and by transitioning into tion to be associated with lower ratings of self-reported attachment
the germline during development, environmentally induced epigenetic anxiety in young probands (Ebner et al., 2019). As mentioned above,
marks are suggested to contribute to an ‘inheritance of acquired traits’ human MAOA methylation has been found to be positively correlated
and thus promote natural selection in a neo-Lamarckian way potentially with the percentage of recently experienced positive life events, while
facilitating neo-Darwinian evolution (Guerrero-Bosagna, 2019; Skinner, relative hypomethylation has been associated with the experience of
2015). Along these lines, epigenetic adaptive plasticity is considered to negative life events (Domschke et al., 2012) and a number of mental
affect speciation (for review see Hernando-Herraez, Garcia-Perez, disorders including panic disorder, depression and substance abuse (for
Sharp, & Marques-Bonet, 2015). However, the general conservation and review, see Ziegler & Domschke, 2018). The classical vulnerability-
divergence of DNA methylation patterns across species, with particular stress model of mental disorders, arguably one of the most influential
regard to traits related to mental disorders, remain to be elucidated in a models of disorder development, as well as conceptually similar ap-
systematic comparative epigenomics fashion. In a first pilot study, proaches such as the “allostatic load”, “double-hit” or “three-hit” hy-
Sipahi et al. (2014) have traced and characterized the evolution of potheses predominantly focus on risk-increasing factors that, assuming
genomic sites associated with the development of PTSD and found that a cumulative effect, will lead to manifestation of disease (Bayer, Falkai,

11
M.A. Schiele, et al. Clinical Psychology Review 77 (2020) 101830

Figure 2. Model of epigenetic mechanisms in mental disorder risk and resilience.


CH3=methyl group

& Maier, 1999; Daskalakis, Bagot, Parker, Vinkers, & de Kloet, 2013; 6.3. Psychotherapy-epigenetics: mechanisms of action and clinical
McEwen, 2003; Zubin & Spring, 1977). In turn, the concept of resilience implications
does not merely entail positive environmental constellations or the
absence of adversity, but rather successful adaptation in the face of As summarized above, there is a growing body of evidence that
adversity (Rutten et al., 2013). In light of the fact that experience of psychological interventions may exert their impact in part via epige-
negative life events or chronic stress does not necessarily lead to psy- netic regulation of gene expression in line with Eric Kandel’s early
chopathology, alternative concepts have gained traction, including the dictum “insofar as psychotherapy is successful in bringing about sub-
“stress inoculation” model, which postulates that early experiences of stantive changes in behaviour, it does so by producing alterations in
moderate stress can actually protect against the effects of environ- gene expression” (Kandel, 1998). Psychotherapy has been suggested to
mental insults later in life (Parker, Buckmaster, Schatzberg, & Lyons, function as an “epigenetic drug’” (Stahl, 2012) capable of inducing
2004), the “match-mismatch” hypothesis entailing that mental disorder epigenetic modifications by exercising its effects on neurobiological
susceptibility can result from a “mismatch” between early and adult circuits which are ultimately reflected on a clinical symptom level (see
environments and, consequently, an impaired ability to cope with Fig. 2). The precise molecular mechanisms of action in mediating
challenge (Nederhof & Schmidt, 2012), and – in a quite similar vein - psychotherapeutic outcome are yet to be determined, and several open
the “predictive adaptive response” hypothesis, which proposes that questions remain. Animal models have provided evidence for epigenetic
cues received in early life program the development of a phenotype that mechanisms to shape cognitive processes and memory formation (Day
is normally adapted to later environmental conditions and that negative & Sweatt, 2011; Halder et al., 2016; Levenson & Sweatt, 2005; Rudenko
health outcomes arise from a mismatch between the predicted and & Tsai, 2014) and to be involved in fear acquisition, consolidation and
actual environment (Bateson, Gluckman, & Hanson, 2014). Therefore, extinction as a preclinical model of anxiety-related phenotypes (Kwapis
while research into the deleterious consequences of stress experiences – & Wood, 2014). Promising animal studies applying psychoenviron-
i.e., increased vulnerability for psychopathology – has led us towards an mental interventions have shown environmental enrichment to exert an
increased understanding of the pathomechanisms of mental disorders, effect on a behavioral (e.g. reductions in anxiety-related behavior,
spotlighting positive mental health outcomes despite exposure to ad- improved task learning or facilitated memory retrieval) (Laviola,
versity can promote essential insights into the mechanisms facilitating Hannan, Macri, Solinas, & Jaber, 2008; Reynolds, Lane, & Richards,
resilience and, importantly, the epigenetic correlates operating at the 2010) as well as on a neurobiological level (van Praag, Kempermann, &
intersection between vulnerability and resilience. For instance, a study Gage, 2000): for instance, environmental enrichment in animal models
of 24 nurses working in a high-stress environment observed lower of mental and neurodegenerative disorders has been linked to mor-
SLC6A4 promoter methylation relative to a low work stress control phological changes such as dendritic and synaptic growth and neuro-
group (Alasaari et al., 2012), possibly indicating a compensatory, i.e. genesis in the hippocampus as well as molecular alterations including
adaptive effect promoting resilience given evidence from clinical sam- neurotransmitter levels, neurotransmitter receptor expression and
ples on increased SLC6A4 methylation to be associated with mental neurotrophin expression (for review, see Alwis & Rajan, 2014;
disorder susceptibility (for review, see Palma-Gudiel & Fananas, 2017). Baroncelli et al., 2010). With particular focus on epigenetic changes,
Accordingly, the eventual consequences of stress experience are likely environmental enrichment was observed to induce histone acetylation
mediated by epigenetic mechanisms functioning as a switch between, and methylation and to re-constitute learning behavior and access to
ultimately, risk and resilience. long-term memories after significant brain atrophy and neuronal loss

12
M.A. Schiele, et al. Clinical Psychology Review 77 (2020) 101830

had already occurred (Fischer, Sananbenesi, Wang, Dobbin, & Tsai, 2015), systematic human studies are warranted to further explore the
2007). Assuming learning processes to be at the core of psychother- potential of lifestyle interventions to modulate mental disorder risk on
apeutic change (Kandel, 1998), it might be speculated that similar an epigenetic level.
epigenetic remodeling and reprogramming may underlie therapeutic In sum, advances in our understanding of how psychotherapy and
mechanisms of action. However, extensive research is needed in order lifestyle interventions translate on a molecular level could subsequently
to address the underlying molecular signature and consequences of inform clinical practice, paving the way for personalized treatment
environmental influences as a whole and of psychotherapeutic inter- options by selecting modes of treatment established on multi-level in-
ventions in particular. A further challenge in therapyepigenetic re- formation and might aid in reducing treatment resistance rates. Finally,
search pertains to the conceptual characteristics of psychotherapy. the stability of symptomatic as well as epigenetic effects remains to be
Psychotherapeutic interventions differ greatly in theoretical back- elucidated. Gaining an improved understanding of the mechanisms
ground, approach, methodology, treatment plan, setting, duration, mediating treatment effects would give rise to devising strategies to
specificity, and techniques. So far, the psychotherapeutic interventions ensure that effects are preserved in a long-term manner, which, from a
studied comprise CBT, exposure therapy, mindfulness-based stress re- clinical point of view, could be absolutely crucial in relapse prevention.
duction or disorder specific approaches like DBT in BPD; notwith-
standing, epigenetic changes occurring over the course of other psy- 6.4. Targetable markers for preventive interventions increasing resilience
chotherapies including, but not limited to, psychoanalysis and
psychodynamic therapies, systemic therapy, schema therapy, or newer In light of the recently burgeoning body of research into the po-
approaches such as acceptance and commitment therapy (Hayes, Levin, tential of epigenetic modifications to predict mental disorder risk and
Plumb-Vilardaga, Villatte, & Pistorello, 2013), metacognitive therapy treatment outcome as well as of psychotherapeutic interventions to
(Wells, 2002) or interpersonal therapy (Cuijpers, Donker, Weissman, target – and potentially reverse - epigenetic risk patterns as reviewed
Ravitz, & Cristea, 2016) have yet to be addressed. Psychotherapy is above, this direction carries great promise for preventive interventions
highly effective in the treatment of mental disorders, however, the to potentially compensate for putative vulnerability factors pre-
mechanisms of action conveying psychotherapeutic change are not yet cipitating mental disorder onset – or to strengthen factors and resources
fully understood, and several challenges pertaining to the assessment of associated with resilient functioning (see Fig. 2). First promising evi-
treatment mediators and mechanisms remain (for review see Kazdin, dence in this regard emerges from a study showing response to a
2007). Yet, it would clearly address a growing clinical need to de- mindfulness-based preventive intervention to go along with decreasing
termine the impact of the putative specific and non-specific, i.e. more SLC6A4 methylation (Stoffel et al., 2019) as well as studies reporting
general and shared across different schools of psychotherapy, me- reduced harsh parenting conferred by a family-centered prevention
chanisms of action in in relation to neurobiological changes occurring training (Brody, Yu, et al., 2016) or the presence of supportive family
concurrently and, thus, to revisit the “Dodo bird conjecture” – i.e. the environments despite racial discrimination (Brody, Miller, et al., 2016)
proposition that common factors shared across different psychother- to protect against accelerated epigenetic aging. Future studies are
apeutic approaches account for a large proportion of treatment effec- warranted to investigate the malleability of epigenetic risk patterns by
tiveness stemming from Saul Rosenzweig’s (1936) seminal work (see preventive interventions more thoroughly in analogy to therapy-epi-
also e.g. Hofmann & Barlow, 2014; Luborsky, Diguer, Luborsky, & genetic research. Further trials probing the long-term stability of pre-
Schmidt, 1999; Marcus, O'Connell, Norris, & Sawaqdeh, 2014; vention-mediated epigenetic effects in longitudinal approaches would
Wampold et al., 1997) – from a molecular perspective. Systematically be of great clinical relevance, informing preventive-therapeutic proce-
comparing different psychotherapeutic approaches would allow to dures (e.g. application of booster sessions, augmentation strategies) in
differentiate whether epigenetic change is possibly specific, or whether increasing – and maintaining – resilience. Moreover, human studies
therapy outcome in general is facilitated through, or effectively based might in particular aim to validate the emerging evidence from pre-
on, common biological mechanisms. Along these lines, it would be clinical studies of resilience-increasing epigenetic patterns, such as
worthwhile to additionally look into epigenetic effects of lifestyle in- transgenerational effects of maternal care via epigenetic mechanisms
terventions such as meditation, yoga or other relaxation techniques, (Champagne, 2008). Thus, although currently still in its infancy, epi-
particularly since practices eliciting relaxation responses as the coun- genetic research holds great promise as targets for preventive inter-
terpart of the stress response have been demonstrated to evoke ex- ventions in at-risk individuals, that could be used to effectively antici-
pression rate changes in genes associated with pro-inflammatory re- pate and preclude disorder onset in the future.
sponses, stress-related cellular signaling, energy metabolism,
mitochondrial functioning, insulin secretion and telomere maintenance 6.5. Transgenerational relevance of epigenetics
(Bhasin et al., 2013). On an epigenetic level, a pilot study analyzing the
DNA methylome in blood cells of long-term meditators and meditation- First evidence points to the transgenerational transmission of epi-
naïve controls identified a protective effect of meditation against an genetic marks of acquired information across generations to shape de-
estimator of intrinsic epigenetic age acceleration (Chaix et al., 2017), velopmental and phenotypic variation in offspring (see Fig. 2). It re-
which might confer an increased resilience towards chronic diseases mains to be clarified, however, whether epigenetic transport of trauma
with comparably late onset. Another study investigating the impact of effects into subsequent generations necessarily increases vulnerability
an 8-week yoga intervention in distressed women discerned a potential to negative mental health outcomes or may on a molecular level also
immuno-regulatory effect of meditation, with reduced tumor necrosis serve to facilitate the ability to adapt to a changing environment, i.e.
factor gene (TNF) methylation in the yoga group as compared to the promote resilience, in the offspring (Yehuda, Lehrner, & Bierer, 2018).
control group (Harkess, Ryan, Delfabbro, & Cohen-Woods, 2016). Accordingly, these epigenetic processes may have evolved in order to
Likewise, other lifestyle interventions such as physical activity and program the phenotype to effectively and rapidly adapt to new en-
exercise have been proposed to exert a preventive effect on mental vironmental demands (Fischer, 2014; Gapp et al., 2016; Yeshurun &
disorder onset and to be involved in mediating resilient functioning Hannan, 2019). Along those lines, it might be feasible that epigenetic
(Ströhle, 2009). A first epigenetic study in this field suggests differential changes arising from successful psychotherapy could be transferred to
BDNF methylation to be associated with exercise in combat-exposed future generations and thus promote the ”transgenerational preven-
veterans with and without current PTSD (Voisey et al., 2019). Given tion” of mental disorders by pre-emptively equipping future genera-
some evidence from animal studies for physical exercise as an epige- tions with adaptive coping abilities on both a phenotypic and genomic
netic modulator of neural plasticity and cognitive function (see level and eventually decrease the risk for disorder manifestation in
Fernandes, Arida, & Gomez-Pinilla, 2017; Voisin, Eynon, Yan, & Bishop, subsequent generations. Nonetheless, at present, caution is warranted

13
M.A. Schiele, et al. Clinical Psychology Review 77 (2020) 101830

in the interpretation of transgenerational evidence. The proposed cau- and variation in DNA methylation appears to be higher in neuronal cells
sative links between molecular adaptations and intergenerational phe- in response to the environment (Iwamoto et al., 2011). Therefore, cell
notypic manifestations are not yet conclusively proven, the im- type composition should be taken into account in the analysis of epi-
plementation of which is not trivial. Future studies need to address the genetic data (Montaño et al., 2013), and single cell approaches (“single-
co-segregation of epigenetic information with specific phenotypic traits cell epigenomics”) constitute a most promising future direction towards
while taking into account effects attributable to genetic, ecological and a better understanding of epigenetic mechanisms (Kelsey, Stegle, &
cultural inheritance (Horsthemke, 2018; Nagy & Turecki, 2015). Reik, 2017).
Next to biological confounders as reviewed above, technical aspects
6.6. Limitations such as sample storage at the appropriate temperature can constitute
another source of confounding effects. Along these lines, DNA methy-
Although advances in epigenetic research are of keen interest to lation arrays are susceptible to batch effects arising from random, un-
psychology and psychiatry, they are not without challenges and pitfalls. systematic fluctuations in laboratory conditions and handling. Several
The high sensitivity to external input and the highly dynamic nature of methods have been suggested to adjust for batch effects, including re-
epigenetic mechanisms that on the one hand allow for rapid and, cru- commendations for experimental designs and statistical approaches
cially, inducible change, can on the other hand simultaneously pose a (see for example Leek et al., 2010).
source of confounding effects. Biological confounders known to affect Finally, in a chicken-and-egg conundrum with regard to epigenetic
epigenetic patterns such as age (Xiao, Wang, & Kong, 2019), sex change occurring concomitantly with clinical change, it remains to be
(Yousefi et al., 2015), ethnicity (Fraser, Lam, Neumann, & Kobor, elucidated whether epigenetic change evokes psychotherapeutic effects
2012), body mass index (Sayols-Baixeras et al., 2017), smoking beha- or occurs as a function of therapy response. Longitudinal designs would
vior (Li et al., 2018) or nutritional aspects (Gadecka & Bielak- be most informative in this regard and allow for a better delineation of
Zmijewska, 2019) may introduce critical biases limiting the inter- cause from effect.
pretation of epigenetic data. Another caveat in epigenetic research
pertains to the ”tissue issue” (Bakulski, Halladay, Hu, Mill, & Fallin, 6.7. Future research
2016) owing to the fact that epigenetic studies so far have mostly (with
the exception of investigations in post-mortem samples) relied on per- Despite recent advances in epigenetic investigations into mental
ipheral tissues, which precludes definite conclusions from being drawn disorder onset, maintenance and treatment, several open questions re-
regarding CNS processes. However, given the difficulty to obtain re- main that are of great promise for future research directions. One major
levant brain tissue specimen in humans in vivo, the identification of challenge entails disentangling the molecular mechanisms and con-
epigenetic patterns in surrogate peripheral biomaterial such as whole sequences of psychotherapy whilst also taking into account different
blood, saliva or buccal cells as a proxy for brain epigenetic markers is schools, forms, and approaches. Large randomized controlled trials
necessary. In support of this endeavor, several studies have compared (RCTs) are needed to address this novel layer in the quest for de-
epigenetic patterns obtained in peripheral and central tissue: In rats, a termining the optimal therapeutic match for an individual patient,
direct correlation between catechol-O-methyltransferase (COMT) me- ideally by applying longitudinal intervention trials.
thylation in peripheral mononuclear blood cells and the prefrontal To date, efforts in identifying epigenetic correlates of mental dis-
cortex was observed (Ursini et al., 2011). In a comparison of epi- orders and their treatment have mostly focused on DNA methylation
genome-wide blood and brain DNA methylation patterns in differen- and less is known about other epigenetic processes introduced above,
tially reared rhesus macaques, a considerable functional overlap was such as hydroxymethylation, histone modifications, or miRNAs. Given
observed in DNA methylation profiles in T lymphocytes and prefrontal the considerable and intricate cross-talk of multiple epigenetic me-
tissue (Provencal et al., 2012). In a similar vein, in humans, some inter- chanisms (Molina-Serrano, Schiza, & Kirmizis, 2013), combinatory
individual variation in epigenome-wide DNA methylation as measured approaches would be most valuable in furthering our understanding of
in post-mortem brain samples and whole blood was reflected across how psychotherapy effects are transported.
tissues (Davies et al., 2012). In a positron emission tomography (PET) In addition to an intermediary role in translating between genes and
study measuring cerebral MAOA activity, peripheral MAOA methyla- the environment, epigenetic mechanisms may be determined by genetic
tion detected in leukocytes was found to inversely correlate with brain variation itself as evidenced by the fact that DNA methylation has been
MAOA levels in healthy men (Shumay, Logan, Volkow, & Fowler, shown to frequently occur in an allele-specific manner (He et al., 2015;
2012). However, for several other genes highly tissue-specific methy- Meaburn, Schalkwyk, & Mill, 2010; Shoemaker, Deng, Wang, & Zhang,
lation patterns emerge (cf. in silico analyses using databases such as 2010). Consequently, mapping of haplotype-dependent allele-specific
BECon (Edgar, Jones, Meaney, Turecki, & Kobor, 2017; available at DNA methylation (hap-ASM) and methylation-eQTLs may further
https://redgar598.shinyapps.io/BECon) or IMAGE-CpG (Braun et al., explain the interplay between genetic and environmental factors and
2019; available at https://han-lab.org/methylation/default/ aid in identifying disease-associated regulatory sequences (Do et al.,
imageCpG). Also, overlap in DNA methylation levels measured in 2017) in combined G × EpiG × E multi-level approaches.
saliva, buccal cells and blood is limited, and it has been suggested that Further research into the functional consequence and biological
DNA methylation patterns in saliva may be closer to that of brain tissue relevance of DNA methylation changes at particular CpG sites is war-
than DNA methylation in blood or buccal cells (Braun et al., 2019; Lowe ranted, preferably in “multi-omic” approaches by combining epige-
et al., 2013; Smith et al., 2015). Taken together, peripheral DNA me- netics with transcriptomics or in silico approaches (Ng et al., 2017).
thylation may partly function as a viable surrogate biomarker for cen- Such integrative studies on epigenetics and the ”omics” – tran-
tral processes, but is not unequivocally related to brain function. In scriptomics, proteomics, metabolomics and microbiomics – could an-
light of the fact that most research on the functional and mechanistic swer central questions, for instance following first hints that methio-
characteristics of epigenetic processes comes from preclinical studies, nine, biotin, and acetyl group donor metabolites produced by the gut
translation of animal findings to humans – and in turn back-transla- flora may affect epigenetic modifications (e.g. Househam, Peterson,
tion, i.e. validation of human findings in animal models – seems war- Mills, & Chopra, 2017; Strachan, Zhao, Roy-Byrne, Fowler, & Bacus,
ranted though challenging given the limited comparability of available 2018). Along these lines, interactions between the epigenome and the
(endo)phenotypes (see Lesch, 2011). immune system could be explored in elucidating the underlying pa-
Similarly, epigenetic processes are not only tissue specific, but can thophysiology of mental disorders and the potential effect of psy-
act in a cell type specific manner. For instance, distinctive DNA me- chotherapy in alleviating this maladaptive crosstalk. These multi-level
thylation patterns between neuronal and glial cells have been observed, analyses of multidimensional data entail the necessity of highly

14
M.A. Schiele, et al. Clinical Psychology Review 77 (2020) 101830

complex bioinformatic approaches in sufficiently powered samples. acid and vitamin B12 (see Peedicayil, 2012). Along those lines, epige-
Since mental disorders constitute complex genetic disorders, a netic modifiers such as S-adenosyl methionine (SAM-e) - a natural
multitude of interacting epigenetic variations is expected; therefore, ubiquitous methyl-group donor approved as nutritional supplement -
shifting the field from candidate gene approaches to EWAS, ideally folic acid or vitamin B12 (cobalamin) could potentially be used as add-
combined with GWAS, would prove fruitful in unraveling the complex on nutritional therapies in mental disorders and open up promising
biochemical nature of mental disorders in the future. paths for additional research, however, current evidence is still limited
While the role of epigenetic mechanisms in translating between (for review, see Dome, Tombor, Lazary, Gonda, & Rihmer, 2019)
environmental input and an individual's genetic background is un- Lastly, it is important to keep in mind the ethical, social and legal
disputed, several sources of variability remain that have yet to be implications of epigenetic research, necessitating the implementation of
identified. Investigations utilizing longitudinal family cohorts starting “epigenet(h)ics”, i.e. guidelines and policies for clinical and scientific
before conception and including well-characterized, extensively phe- use of epigenetic information (Dyke et al., 2019; Thomas, 2015).
notyped probands (Latendresse, Musci, & Maher, 2018; Provencal &
Binder, 2015) by collecting detailed documentation of environmental 7. Conclusion
events including frequency, sequence and individually perceived va-
lence to disentangle whether epigenetic patterns are cause, correlation Mental disorders are associated with epigenetic changes, however,
or consequence of the pathological state, and to furthermore identify it has still not been resolved whether these modifications are cause or
critical time periods during which the early environment can impact consequence of the elemental pathophysiology. Epigenetic alterations
brain and mental health development (Andersen, 2003). Additionally, arise in response to environmental signals and are modifiable by psy-
positive, counteracting and potentially resilience-increasing environ- chotherapy, though the underlying mechanisms of action are yet to be
mental factors should be taken into consideration. fully discerned. While epigenetic research holds great promise re-
Given the sex-specific impact of adverse environment on DNA me- garding the prediction, prevention, and personalized treatment of
thylation (e.g. Massart et al., 2016; Ostlund et al., 2016) and sex-spe- mental disorders, it is still in its infancy and limited in its clinical ap-
cific findings in studies on psychotherapeutic effects on methylation plication at the moment. Future investigations will have to explore the
(e.g. Schiele et al., 2018; Ziegler et al., 2016), upcoming studies may causality of how intervention effects are internalized on the molecular
want to systematically consider sex-specific effects in the susceptibility level and how to best translate novel insights relating to epigenetic
to psychopathology and the mediation of beneficial treatment effects in modifications into clinical practice. The field of epigenetics opens up
synopsis with epigenetic information (for review, see Tiwari & novel avenues, increasing our understanding of the intricate multi-level
Gonzalez, 2018). etiology of mental disorders which is hoped to strengthen the inter-
Intermediate (endo)phenotypes of mental disorders as well as the disciplinary dialogue between the fields of neurobiology/genetics with
different constructs advocated by the NIMH's Research Domain Criteria psychology/psychotherapy to their mutual benefit.
(RDoC) framework may constitute promising targets in epigenetic re-
search. For instance, future research may want to employ an “imaging Role of funding sources
epigenetic” approach in order to determine therapyepigenetic effects on
an intermediate (endo)phenotype level given a wealth of previous re- This work was partly supported by the Deutsche
search on psychotherapy effects on a neural networks level (for review, Forschungsgemeinschaft (DFG, German Research Foundation) – project
see Fournier & Price, 2014). Exemplarily, experiences of childhood number 44541416 – TRR 58 “Fear, Anxiety, Anxiety Disorders”, (pro-
trauma, male gender, and smaller hippocampal volume were in- jects C02 and Z02 to KD) and the German Ministry of Research and
dependently associated with greater peripheral SLC6A4 methylation Education (BMBF, 01EE1402F, PROTECT-AD, project P5 to KD). The
(Booij et al., 2015). SLC6A4 methylation was furthermore found to be funding sources had no further role in study design, in the collection,
related to insula activation during the processing of emotional stimuli analysis and interpretation of data, in the writing of the report, and in
in major depressive disorder (Frodl et al., 2015). Methylation of an the decision to submit the paper for publication.
AluJb element in the SLC6A4 promoter was also associated with
amygdala reactivity in depression (Schneider et al., 2018). In social Contributors
phobia, OXTR methylation was related to differential amygdala re-
activity to social phobia-related verbal stimuli (Ziegler et al., 2015). MAS, MGG and KD managed the literature search and wrote the
OXTR methylation was furthermore found to interact with callous-un- manuscript. All authors contributed to and have approved the final
emotional traits in predicting frontoparietal activation and amygdala- version of the manuscript.
frontoparietal disconnection, brain systems critical to decoding and
integrating socioaffective information, in youths with conduct disorder Declaration of Competing Interest
(Aghajani et al., 2018). Along these lines, future studies should address
psychotherapy effects in synopsis with epigenetic and neuroimaging KD is a member of the Janssen Pharmaceuticals, Inc. Steering
information in order to increase our understanding of epigenetic Committee Neurosciences. All other authors declare that they have no
changes with brain function, for instance by applying PET in order to conflicts of interest.
directly visualize epigenetic processes in the central nervous system
(Wang, Schroeder, & Hooker, 2014). Acknowledgements
Studies mostly in preclinical models suggest drugs modulating epi-
genetic mechanisms that may lead to the development of novel, in- MAS and KD are members of the Anxiety Disorders Research
novative treatment options such as HDAC or DNMT inhibitors (cf. Network (ADRN), European College of Neuropsychopharmacology
Whittle & Singewald, 2014). Such “epigenetic drugs” might serve to (ECNP). We gratefully acknowledge the support by S. Meixensberger
catalyze psychotherapeutic effects by enhancing learning and neuronal and C. Ziegler in the preparation of this manuscript.
plasticity and facilitating fear extinction (Gavin, Chase, & Sharma,
2011). Yet, clinical application is still a long way off, and several ca- References
veats such as the lack of selectivity and specificity of currently available
agents remain (see Szyf, 2015). Abdolmaleky, H. M., Zhou, J. R., & Thiagalingam, S. (2015). An update on the epigenetics
Moreover, nutrition can influence the epigenetic machinery, and of psychotic diseases and autism. Epigenomics, 7, 427–449.
Aghajani, M., Klapwijk, E. T., Colins, O. F., Ziegler, C., Domschke, K., Vermeiren, R., &
first studies have focused on the impact of haematopoietic drugs, folic

15
M.A. Schiele, et al. Clinical Psychology Review 77 (2020) 101830

van der Wee, N. J. A. (2018). Interactions between oxytocin receptor gene methy- early-life adversity outcome. Psychoneuroendocrinology, 38, 1858–1873.
lation and callous-unemotional traits impact socioaffective brain systems in conduct- Davies, M. N., Volta, M., Pidsley, R., Lunnon, K., Dixit, A., Lovestone, S., ... Mill, J. (2012).
disordered offenders. Biological Psychiatry: Cognitive Neuroscience and Neuroimaging, 3, Functional annotation of the human brain methylome identifies tissue-specific epi-
379–391. genetic variation across brain and blood. Genome Biology, 13, R43.
Alasaari, J. S., Lagus, M., Ollila, H. M., Toivola, A., Kivimaki, M., Vahtera, J., ... Paunio, T. Day, J. J., & Sweatt, J. D. (2011). Epigenetic mechanisms in cognition. Neuron, 70,
(2012). Environmental stress affects DNA methylation of a CpG rich promoter region 813–829.
of serotonin transporter gene in a nurse cohort. PLoS One, 7, e45813. Dias, B. G., & Ressler, K. J. (2014). Parental olfactory experience influences behavior and
Alwis, D. S., & Rajan, R. (2014). Environmental enrichment and the sensory brain: The neural structure in subsequent generations. Nature Neuroscience, 17, 89–96.
role of enrichment in remediating brain injury. Frontiers in Systems Neuroscience, 8, Do, C., Shearer, A., Suzuki, M., Terry, M. B., Gelernter, J., Greally, J. M., & Tycko, B.
156. (2017). Genetic-epigenetic interactions in cis: A major focus in the post-GWAS era.
Alyamani, R. A. S., & Murgatroyd, C. (2018). Epigenetic programming by early-life stress. Genome Biology, 18, 120.
Progress in Molecular Biology and Translational Science, 157, 133–150. Dome, P., Tombor, L., Lazary, J., Gonda, X., & Rihmer, Z. (2019). Natural health products,
Andersen, S. L. (2003). Trajectories of brain development: point of vulnerability or dietary minerals and over-the-counter medications as add-on therapies to anti-
window of opportunity? Neuroscience and Biobehavioral Reviews, 27, 3–18. depressants in the treatment of major depressive disorder: A review. Brain Research
Babenko, O., Kovalchuk, I., & Metz, G. A. (2015). Stress-induced perinatal and transge- Bulletin, 146, 51–78.
nerational epigenetic programming of brain development and mental health. Domschke, K., Tidow, N., Kuithan, H., Schwarte, K., Klauke, B., Ambree, O., ... Deckert, J.
Neuroscience and Biobehavioral Reviews, 48, 70–91. (2012). Monoamine oxidase A gene DNA hypomethylation - A risk factor for panic
Bakulski, K. M., Halladay, A., Hu, V. W., Mill, J., & Fallin, M. D. (2016). Epigenetic disorder? The International Journal of Neuropsychopharmacology, 15, 1217–1228.
research in neuropsychiatric disorders: the "tissue issue" Current Behavioral Domschke, K., Tidow, N., Schrempf, M., Schwarte, K., Klauke, B., Reif, A., ... Deckert, J.
Neuroscience Reports, 3, 264–274. (2013). Epigenetic signature of panic disorder: A role of glutamate decarboxylase 1
Bakusic, J., Schaufeli, W., Claes, S., & Godderis, L. (2017). Stress, burnout and depression: (GAD1) DNA hypomethylation? Progress in Neuro-Psychopharmacology & Biological
A systematic review on DNA methylation mechanisms. Journal of Psychosomatic Psychiatry, 46, 189–196.
Research, 92, 34–44. Dudley, K. J., Li, X., Kobor, M. S., Kippin, T. E., & Bredy, T. W. (2011). Epigenetic me-
Bale, T. L. (2014). Lifetime stress experience: Transgenerational epigenetics and germ cell chanisms mediating vulnerability and resilience to psychiatric disorders. Neuroscience
programming. Dialogues in Clinical Neuroscience, 16, 297–305. and Biobehavioral Reviews, 35, 1544–1551.
Baroncelli, L., Braschi, C., Spolidoro, M., Begenisic, T., Sale, A., & Maffei, L. (2010). Dyke, S. O. M., Saulnier, K. M., Dupras, C., Webster, A. P., Maschke, K., Rothstein, M., ...
Nurturing brain plasticity: Impact of environmental enrichment. Cell Death and Joly, Y. (2019). Points-to-consider on the return of results in epigenetic research.
Differentiation, 17, 1092–1103. Genome Medicine, 11, 31.
Bartel, D. P. (2004). MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell, Ebner, N. C., Lin, T., Muradoglu, M., Weir, D. H., Plasencia, G. M., Lillard, T. S., ...
116, 281–297. Connelly, J. J. (2019). Associations between oxytocin receptor gene (OXTR) me-
Bateson, P., Gluckman, P., & Hanson, M. (2014). The biology of developmental plasticity thylation, plasma oxytocin, and attachment across adulthood. International Journal of
and the predictive adaptive response hypothesis. The Journal of Physiology, 592, Psychophysiology, 136, 22–32.
2357–2368. Edgar, R. D., Jones, M. J., Meaney, M. J., Turecki, G., & Kobor, M. S. (2017). BECon: A
Bayer, T. A., Falkai, P., & Maier, W. (1999). Genetic and non-genetic vulnerability factors tool for interpreting DNA methylation findings from blood in the context of brain.
in schizophrenia: the basis of the "two hit hypothesis" Journal of Psychiatric Research, Translational Psychiatry, 7, e1187.
33, 543–548. Farrell, C., Doolin, K., N, O. L., Jairaj, C., Roddy, D., Tozzi, L., ... O'Keane, V. (2018). DNA
Beach, S. R., Brody, G. H., Todorov, A. A., Gunter, T. D., & Philibert, R. A. (2011). methylation differences at the glucocorticoid receptor gene in depression are related
Methylation at 5HTT mediates the impact of child sex abuse on women's antisocial to functional alterations in hypothalamic-pituitary-adrenal axis activity and to early
behavior: An examination of the Iowa adoptee sample. Psychosomatic Medicine, 73, life emotional abuse. Psychiatry Research, 265, 341–348.
83–87. Fernandes, J., Arida, R. M., & Gomez-Pinilla, F. (2017). Physical exercise as an epigenetic
Belsky, J., & Pluess, M. (2009). Beyond diathesis stress: Differential susceptibility to en- modulator of brain plasticity and cognition. Neuroscience and Biobehavioral Reviews,
vironmental influences. Psychological Bulletin, 135, 885–908. 80, 443–456.
Berkel, T. D., & Pandey, S. C. (2017). Emerging role of epigenetic mechanisms in alcohol Fischer, A. (2014). Epigenetic memory: The Lamarckian brain. The EMBO Journal, 33,
addiction. Alcoholism, Clinical and Experimental Research, 41, 666–680. 945–967.
Bhasin, M. K., Dusek, J. A., Chang, B. H., Joseph, M. G., Denninger, J. W., Fricchione, G. Fischer, A., Sananbenesi, F., Wang, X., Dobbin, M., & Tsai, L. H. (2007). Recovery of
L., ... Libermann, T. A. (2013). Relaxation response induces temporal transcriptome learning and memory is associated with chromatin remodelling. Nature, 447,
changes in energy metabolism, insulin secretion and inflammatory pathways. PLoS 178–182.
One, 8, e62817. Fournier, J. C., & Price, R. B. (2014). Psychotherapy and neuroimaging. Focus (American
Bishop, J. R., Lee, A. M., Mills, L. J., Thuras, P. D., Eum, S., Clancy, D., ... Lim, K. O. Psychiatric Publishing), 12, 290–298.
(2018). Methylation of FKBP5 and SLC6A4 in relation to treatment response to Franklin, T. B., Russig, H., Weiss, I. C., Graff, J., Linder, N., Michalon, A., ... Mansuy, I. M.
mindfulness based stress reduction for posttraumatic stress disorder. Frontiers in (2010). Epigenetic transmission of the impact of early stress across generations.
Psychiatry, 9, 418. Biological Psychiatry, 68, 408–415.
Boks, M. P., de Jong, N. M., Kas, M. J., Vinkers, C. H., Fernandes, C., Kahn, R. S., ... Fraser, H. B., Lam, L. L., Neumann, S. M., & Kobor, M. S. (2012). Population-specificity of
Ophoff, R. A. (2012). Current status and future prospects for epigenetic psycho- human DNA methylation. Genome Biology, 13, R8.
pharmacology. Epigenetics, 7, 20–28. Frodl, T., Szyf, M., Carballedo, A., Ly, V., Dymov, S., Vaisheva, F., ... Booij, L. (2015).
Boks, M. P., van Mierlo, H. C., Rutten, B. P., Radstake, T. R., De Witte, L., Geuze, E., ... DNA methylation of the serotonin transporter gene (SLC6A4) is associated with brain
Vermetten, E. (2015). Longitudinal changes of telomere length and epigenetic age function involved in processing emotional stimuli. Journal of Psychiatry &
related to traumatic stress and post-traumatic stress disorder. Neuroscience, 40, 296–305.
Psychoneuroendocrinology, 51, 506–512. Gadecka, A., & Bielak-Zmijewska, A. (2019). Slowing down ageing: The role of nutrients
Booij, L., Szyf, M., Carballedo, A., Frey, E. M., Morris, D., Dymov, S., ... Frodl, T. (2015). and microbiota in modulation of the epigenome. Nutrients, 11.
DNA methylation of the serotonin transporter gene in peripheral cells and stress- Gapp, K., Bohacek, J., Grossmann, J., Brunner, A. M., Manuella, F., Nanni, P., & Mansuy,
related changes in hippocampal volume: A study in depressed patients and healthy I. M. (2016). Potential of environmental enrichment to prevent transgenerational
controls. PLoS One, 10, e0119061. effects of paternal trauma. Neuropsychopharmacology, 41, 2749–2758.
Braun, P. R., Han, S., Hing, B., Nagahama, Y., Gaul, L. N., Heinzman, J. T., ... Shinozaki, Garg, E., Chen, L., Nguyen, T. T. T., Pokhvisneva, I., Chen, L. M., Unternaehrer, E., ...
G. (2019). Genome-wide DNA methylation comparison between live human brain Mavan Study, T. (2018). The early care environment and DNA methylome variation
and peripheral tissues within individuals. Translational Psychiatry, 9, 47. in childhood. Development and Psychopathology, 30, 891–903.
Brody, G. H., Miller, G. E., Yu, T., Beach, S. R., & Chen, E. (2016). Supportive family Gavin, D. P., Chase, K. A., & Sharma, R. P. (2011). Enhancement of psychotherapy using
environments ameliorate the link between racial discrimination and epigenetic aging: epigenetic modulating drugs. Medical Hypotheses, 77, 121–124.
A replication across two longitudinal cohorts. Psychological Science, 27, 530–541. Gibbs, J. R., van der Brug, M. P., Hernandez, D. G., Traynor, B. J., Nalls, M. A., Lai, S. L.,
Brody, G. H., Yu, T., Chen, E., Beach, S. R., & Miller, G. E. (2016). Family-centered ... Singleton, A. B. (2010). Abundant quantitative trait loci exist for DNA methylation
prevention ameliorates the longitudinal association between risky family processes and gene expression in human brain. PLoS Genetics, 6, e1000952.
and epigenetic aging. Journal of Child Psychology and Psychiatry, 57, 566–574. Gottschalk, M. G., & Domschke, K. (2016). Novel developments in genetic and epigenetic
Chaix, R., Alvarez-Lopez, M. J., Fagny, M., Lemee, L., Regnault, B., Davidson, R. J., ... mechanisms of anxiety. Current Opinion in Psychiatry, 29, 32–38.
Kaliman, P. (2017). Epigenetic clock analysis in long-term meditators. Gottschalk, M. G., & Domschke, K. (2018). Oxytocin and anxiety disorders. Current Topics
Psychoneuroendocrinology, 85, 210–214. in Behavioral Neurosciences, 35, 467–498.
Champagne, F. A. (2008). Epigenetic mechanisms and the transgenerational effects of Gratten, J., Wray, N. R., Keller, M. C., & Visscher, P. M. (2014). Large-scale genomics
maternal care. Frontiers in Neuroendocrinology, 29, 386–397. unveils the genetic architecture of psychiatric disorders. Nature Neuroscience, 17,
Cimino, S., Cerniglia, L., Ballarotto, G., Marzilli, E., Pascale, E., D'Addario, C., ... Tambelli, 782–790.
R. (2017). DNA methylation at the DAT promoter and risk for psychopathology: Gregory, R. I., Chendrimada, T. P., Cooch, N., & Shiekhattar, R. (2005). Human RISC
Intergenerational transmission between school-age youths and their parents in a couples microRNA biogenesis and posttranscriptional gene silencing. Cell, 123,
community sample. Frontiers in Psychiatry, 8, 303. 631–640.
Cuijpers, P., Donker, T., Weissman, M. M., Ravitz, P., & Cristea, I. A. (2016). Interpersonal Gregory, R. I., Yan, K. P., Amuthan, G., Chendrimada, T., Doratotaj, B., Cooch, N., &
psychotherapy for mental health problems: A comprehensive meta-analysis. The Shiekhattar, R. (2004). The microprocessor complex mediates the genesis of
American Journal of Psychiatry, 173, 680–687. microRNAs. Nature, 432, 235–240.
Daskalakis, N. P., Bagot, R. C., Parker, K. J., Vinkers, C. H., & de Kloet, E. R. (2013). The Guerrero-Bosagna, C. (2019). From epigenotype to new genotypes: Relevance of epige-
three-hit concept of vulnerability and resilience: Toward understanding adaptation to netic mechanisms in the emergence of genomic evolutionary novelty. Seminars in Cell

16
M.A. Schiele, et al. Clinical Psychology Review 77 (2020) 101830

& Developmental Biology, 97, 86–92. V. (2018). DNA methylation of APBA3 and MCF2 in borderline personality disorder:
Haas, B. W., Filkowski, M. M., Cochran, R. N., Denison, L., Ishak, A., Nishitani, S., & Potential biomarkers for response to psychotherapy. European
Smith, A. K. (2016). Epigenetic modification of OXT and human sociability. Neuropsychopharmacology, 28, 252–263.
Proceedings of the National Academy of Sciences of the United States of America, 113, Kundakovic, M., & Champagne, F. A. (2015). Early-life experience, epigenetics, and the
E3816–E3823. developing brain. Neuropsychopharmacology, 40, 141–153.
Halder, R., Hennion, M., Vidal, R. O., Shomroni, O., Rahman, R. U., Rajput, A., ... Bonn, S. Kwapis, J. L., & Wood, M. A. (2014). Epigenetic mechanisms in fear conditioning:
(2016). DNA methylation changes in plasticity genes accompany the formation and Implications for treating post-traumatic stress disorder. Trends in Neurosciences, 37,
maintenance of memory. Nature Neuroscience, 19, 102–110. 706–720.
Hamilton, S. P. (2009). Linkage and association studies of anxiety disorders. Depression Latendresse, S. J., Musci, R., & Maher, B. S. (2018). Critical issues in the inclusion of
and Anxiety, 26, 976–983. genetic and epigenetic information in prevention and intervention trials. Prevention
Han, L. K. M., Aghajani, M., Clark, S. L., Chan, R. F., Hattab, M. W., Shabalin, A. A., ... Science, 19, 58–67.
Penninx, B. (2018). Epigenetic aging in major depressive disorder. The American Laviola, G., Hannan, A. J., Macri, S., Solinas, M., & Jaber, M. (2008). Effects of enriched
Journal of Psychiatry, 175, 774–782. environment on animal models of neurodegenerative diseases and psychiatric dis-
Harkess, K. N., Ryan, J., Delfabbro, P. H., & Cohen-Woods, S. (2016). Preliminary in- orders. Neurobiology of Disease, 31, 159–168.
dications of the effect of a brief yoga intervention on markers of inflammation and Leek, J. T., Scharpf, R. B., Bravo, H. C., Simcha, D., Langmead, B., Johnson, W. E., ...
DNA methylation in chronically stressed women. Translational Psychiatry, 6, e965. Irizarry, R. A. (2010). Tackling the widespread and critical impact of batch effects in
Hayes, S. C., Levin, M. E., Plumb-Vilardaga, J., Villatte, J. L., & Pistorello, J. (2013). high-throughput data. Nature Reviews. Genetics, 11, 733–739.
Acceptance and commitment therapy and contextual behavioral science: Examining Lesch, K. P. (2011). When the serotonin transporter gene meets adversity: The con-
the progress of a distinctive model of behavioral and cognitive therapy. Behavior tribution of animal models to understanding epigenetic mechanisms in affective
Therapy, 44, 180–198. disorders and resilience. Current Topics in Behavioral Neurosciences, 7, 251–280.
He, J., Sun, M. A., Wang, Z., Wang, Q., Li, Q., & Xie, H. (2015). Characterization and Levenson, J. M., & Sweatt, J. D. (2005). Epigenetic mechanisms in memory formation.
machine learning prediction of allele-specific DNA methylation. Genomics, 106, Nature Reviews. Neuroscience, 6, 108–118.
331–339. Levinson, D. F. (2006). The genetics of depression: A review. Biological Psychiatry, 60,
Hernando-Herraez, I., Garcia-Perez, R., Sharp, A. J., & Marques-Bonet, T. (2015). DNA 84–92.
methylation: Insights into human evolution. PLoS Genetics, 11, e1005661. Li, S., Wong, E. M., Bui, M., Nguyen, T. L., Joo, J. E., Stone, J., ... Hopper, J. L. (2018).
Hoehe, M. R., & Morris-Rosendahl, D. J. (2018). The role of genetics and genomics in Causal effect of smoking on DNA methylation in peripheral blood: A twin and family
clinical psychiatry. Dialogues in Clinical Neuroscience, 20, 169–177. study. Clinical Epigenetics, 10, 18.
Hofmann, S. G., & Barlow, D. H. (2014). Evidence-based psychological interventions and Lim, L. P., Lau, N. C., Garrett-Engele, P., Grimson, A., Schelter, J. M., Castle, J., ...
the common factors approach: The beginnings of a rapprochement? Psychotherapy Johnson, J. M. (2005). Microarray analysis shows that some microRNAs down-
(Chicago, Ill.), 51, 510–513. regulate large numbers of target mRNAs. Nature, 433, 769–773.
Horsthemke, B. (2018). A critical view on transgenerational epigenetic inheritance in Lowe, R., Gemma, C., Beyan, H., Hawa, M. I., Bazeos, A., Leslie, R. D., ... Ramagopalan, S.
humans. Nature Communications, 9, 2973. V. (2013). Buccals are likely to be a more informative surrogate tissue than blood for
Horvath, S. (2013). DNA methylation age of human tissues and cell types. Genome Biology, epigenome-wide association studies. Epigenetics, 8, 445–454.
14, R115. Luborsky, L., Diguer, L., Luborsky, E., & Schmidt, K. A. (1999). The efficacy of dynamic
Househam, A. M., Peterson, C. T., Mills, P. J., & Chopra, D. (2017). The effects of stress versus other psychotherapies - Is it true that "everyone has won and all must have prizes"? -
and meditation on the immune system, human microbiota, and epigenetics. Advances An update. Washington: Amer Psychiatric Press, Inc.
in Mind-Body Medicine, 31, 10–25. Marcus, D. K., O'Connell, D., Norris, A. L., & Sawaqdeh, A. (2014). Is the Dodo bird
Howe, A. S., Buttenschon, H. N., Bani-Fatemi, A., Maron, E., Otowa, T., Erhardt, A., ... De endangered in the 21st century? A meta-analysis of treatment comparison studies.
Luca, V. (2016). Candidate genes in panic disorder: Meta-analyses of 23 common Clinical Psychology Review, 34, 519–530.
variants in major anxiogenic pathways. Molecular Psychiatry, 21, 665–679. Massart, R., Nemoda, Z., Suderman, M. J., Sutti, S., Ruggiero, A. M., Dettmer, A. M., ...
van IJzendoorn, M. H., Caspers, K., Bakermans-Kranenburg, M. J., Beach, S. R., & Szyf, M. (2016). Early life adversity alters normal sex-dependent developmental
Philibert, R. (2010). Methylation matters: Interaction between methylation density dynamics of DNA methylation. Development and Psychopathology, 28, 1259–1272.
and serotonin transporter genotype predicts unresolved loss or trauma. Biological McEwen, B. S. (2003). Mood disorders and allostatic load. Biological Psychiatry, 54,
Psychiatry, 68, 405–407. 200–207.
Iwamoto, K., Bundo, M., Ueda, J., Oldham, M. C., Ukai, W., Hashimoto, E., ... Kato, T. McGowan, P. O., Sasaki, A., D'Alessio, A. C., Dymov, S., Labonte, B., Szyf, M., ... Meaney,
(2011). Neurons show distinctive DNA methylation profile and higher interindividual M. J. (2009). Epigenetic regulation of the glucocorticoid receptor in human brain
variations compared with non-neurons. Genome Research, 21, 688–696. associates with childhood abuse. Nature Neuroscience, 12, 342–348.
Jaaskelainen, T., Makkonen, H., & Palvimo, J. J. (2011). Steroid up-regulation of FKBP51 Meaburn, E. L., Schalkwyk, L. C., & Mill, J. (2010). Allele-specific methylation in the
and its role in hormone signaling. Current Opinion in Pharmacology, 11, 326–331. human genome: Implications for genetic studies of complex disease. Epigenetics, 5,
Jawaid, A., Roszkowski, M., & Mansuy, I. M. (2018). Transgenerational epigenetics of 578–582.
traumatic stress. Progress in Molecular Biology and Translational Science, 158, 273–298. Mehta, D., Klengel, T., Conneely, K. N., Smith, A. K., Altmann, A., Pace, T. W., ... Binder,
Jenuwein, T., & Allis, C. D. (2001). Translating the histone code. Science, 293, 1074–1080. E. B. (2013). Childhood maltreatment is associated with distinct genomic and epi-
Jimenez, J. P., Botto, A., Herrera, L., Leighton, C., Rossi, J. L., Quevedo, Y., ... Luyten, P. genetic profiles in posttraumatic stress disorder. Proceedings of the National Academy
(2018). Psychotherapy and genetic neuroscience: An emerging dialog. Frontiers in of Sciences of the United States of America, 110, 8302–8307.
Genetics, 9, 257. Millan, M. J. (2014). The epigenetic dimension of Alzheimer's disease: Causal, con-
Kahl, K. G., Georgi, K., Bleich, S., Muschler, M., Hillemacher, T., Hilfiker-Kleinert, D., ... sequence, or curiosity? Dialogues in Clinical Neuroscience, 16, 373–393.
Frieling, H. (2016). Altered DNA methylation of glucose transporter 1 and glucose Miller, C. W. T. (2017). Epigenetic and neural circuitry landscape of psychotherapeutic
transporter 4 in patients with major depressive disorder. Journal of Psychiatric interventions. Psychiatry Journal, 2017, 5491812.
Research, 76, 66–73. Molina-Serrano, D., Schiza, V., & Kirmizis, A. (2013). Cross-talk among epigenetic
Kandel, E. R. (1998). A new intellectual framework for psychiatry. The American Journal modifications: Lessons from histone arginine methylation. Biochemical Society
of Psychiatry, 155, 457–469. Transactions, 41, 751–759.
Kanherkar, R. R., Bhatia-Dey, N., & Csoka, A. B. (2014). Epigenetics across the human Monk, C., Feng, T., Lee, S., Krupska, I., Champagne, F. A., & Tycko, B. (2016). Distress
lifespan. Frontiers in Cell and Development Biology, 2, 49. during pregnancy: Epigenetic regulation of placenta glucocorticoid-related genes and
Kaywin, L. (1960). An epigenetic approach to the psychoanalytic theory of instincts and fetal neurobehavior. The American Journal of Psychiatry, 173, 705–713.
affects. Journal of the American Psychoanalytic Association, 8, 613–658. Montaño, C. M., Irizarry, R. A., Kaufmann, W. E., Talbot, K., Gur, R. E., Feinberg, A. P., &
Kazdin, A. E. (2007). Mediators and mechanisms of change in psychotherapy research. Taub, M. A. (2013). Measuring cell-type specific differential methylation in human
Annual Review of Clinical Psychology, 3, 1–27. brain tissue. Genome Biology, 14, R94.
Kelsey, G., Stegle, O., & Reik, W. (2017). Single-cell epigenomics: Recording the past and Nagy, C., & Turecki, G. (2015). Transgenerational epigenetic inheritance: An open dis-
predicting the future. Science, 358, 69–75. cussion. Epigenomics, 7, 781–790.
Khulan, B., Manning, J. R., Dunbar, D. R., Seckl, J. R., Raikkonen, K., Eriksson, J. G., & Nederhof, E., & Schmidt, M. V. (2012). Mismatch or cumulative stress: Toward an in-
Drake, A. J. (2014). Epigenomic profiling of men exposed to early-life stress reveals tegrated hypothesis of programming effects. Physiology & Behavior, 106, 691–700.
DNA methylation differences in association with current mental state. Translational Nestler, E. J., Pena, C. J., Kundakovic, M., Mitchell, A., & Akbarian, S. (2016). Epigenetic
Psychiatry, 4, e448. basis of mental illness. Neuroscientist, 22, 447–463.
Klengel, T., Dias, B. G., & Ressler, K. J. (2016). Models of intergenerational and trans- Ng, B., White, C. C., Klein, H. U., Sieberts, S. K., McCabe, C., Patrick, E., ... De Jager, P. L.
generational transmission of risk for psychopathology in mice. (2017). An xQTL map integrates the genetic architecture of the human brain's tran-
Neuropsychopharmacology, 41, 219–231. scriptome and epigenome. Nature Neuroscience, 20, 1418–1426.
Klengel, T., Mehta, D., Anacker, C., Rex-Haffner, M., Pruessner, J. C., Pariante, C. M., ... Oberlander, T. F., Weinberg, J., Papsdorf, M., Grunau, R., Misri, S., & Devlin, A. M.
Binder, E. B. (2013). Allele-specific FKBP5 DNA demethylation mediates gene- (2008). Prenatal exposure to maternal depression, neonatal methylation of human
childhood trauma interactions. Nature Neuroscience, 16, 33–41. glucocorticoid receptor gene (NR3C1) and infant cortisol stress responses. Epigenetics,
Klengel, T., Pape, J., Binder, E. B., & Mehta, D. (2014). The role of DNA methylation in 3, 97–106.
stress-related psychiatric disorders. Neuropharmacology, 80, 115–132. Ostlund, B. D., Conradt, E., Crowell, S. E., Tyrka, A. R., Marsit, C. J., & Lester, B. M.
Klinger-Konig, J., Hertel, J., Van der Auwera, S., Frenzel, S., Pfeiffer, L., Waldenberger, (2016). Prenatal stress, fearfulness, and the epigenome: Exploratory analysis of sex
M., ... Grabe, H. J. (2019). Methylation of the FKBP5 gene in association with FKBP5 differences in DNA methylation of the glucocorticoid receptor gene. Frontiers in
genotypes, childhood maltreatment and depression. Neuropsychopharmacology, 44, Behavioral Neuroscience, 10, 147.
930–938. Ouellet-Morin, I., Wong, C. C., Danese, A., Pariante, C. M., Papadopoulos, A. S., Mill, J., &
Knoblich, N., Gundel, F., Bruckmann, C., Becker-Sadzio, J., Frischholz, C., & Nieratschker, Arseneault, L. (2013). Increased serotonin transporter gene (SERT) DNA methylation

17
M.A. Schiele, et al. Clinical Psychology Review 77 (2020) 101830

is associated with bullying victimization and blunted cortisol response to stress in wide association study. The REGICOR study. Epigenetics, 12, 909–916.
childhood: A longitudinal study of discordant monozygotic twins. Psychological Schiele, M. A., & Domschke, K. (2018). Epigenetics at the crossroads between genes,
Medicine, 43, 1813–1823. environment and resilience in anxiety disorders. Genes, Brain, and Behavior, 17,
Palma-Gudiel, H., & Fananas, L. (2017). An integrative review of methylation at the e12423.
serotonin transporter gene and its dialogue with environmental risk factors, psy- Schiele, M. A., Kollert, L., Lesch, K. P., Arolt, V., Zwanzger, P., Deckert, J., ... Domschke,
chopathology and 5-HTTLPR. Neuroscience and Biobehavioral Reviews, 72, 190–209. K. (2019). Hypermethylation of the serotonin transporter gene promoter in panic
Parent, J., Parade, S. H., Laumann, L. E., Ridout, K. K., Yang, B. Z., Marsit, C. J., ... Tyrka, disorder-Epigenetic imprint of comorbid depression? European
A. R. (2017). Dynamic stress-related epigenetic regulation of the glucocorticoid re- Neuropsychopharmacology, 29, 1161–1167.
ceptor gene promoter during early development: The role of child maltreatment. Schiele, M. A., Ziegler, C., Holitschke, K., Schartner, C., Schmidt, B., Weber, H., ...
Development and Psychopathology, 29, 1635–1648. Domschke, K. (2016). Influence of 5-HTT variation, childhood trauma and self-effi-
Park, C., Rosenblat, J. D., Brietzke, E., Pan, Z., Lee, Y., Cao, B., ... McIntyre, R. S. (2019). cacy on anxiety traits: a gene-environment-coping interaction study. Journal of Neural
Stress, epigenetics and depression: A systematic review. Neuroscience and Transmission (Vienna), 123, 895–904.
Biobehavioral Reviews, 102, 139–152. Schiele, M. A., Ziegler, C., Kollert, L., Katzorke, A., Schartner, C., Busch, Y., ... Domschke,
Parker, K. J., Buckmaster, C. L., Schatzberg, A. F., & Lyons, D. M. (2004). Prospective K. (2018). Plasticity of functional MAOA gene methylation in acrophobia. The
investigation of stress inoculation in young monkeys. Archives of General Psychiatry, International Journal of Neuropsychopharmacology, 21, 822–827.
61, 933–941. Schneider, I., Kugel, H., Redlich, R., Grotegerd, D., Burger, C., Burkner, P. C., ... Hohoff, C.
Peedicayil, J. (2004). Psychosocial factors in the pathogenesis of mental disorders. The (2018). Association of serotonin transporter gene AluJb methylation with major
British Journal of Psychiatry, 185, 520. depression, amygdala responsiveness, 5-HTTLPR/rs25531 polymorphism, and stress.
Peedicayil, J. (2012). Role of epigenetics in pharmacotherapy, psychotherapy and nu- Neuropsychopharmacology, 43, 1308–1316.
tritional management of mental disorders. Journal of Clinical Pharmacy and Schuebel, K., Gitik, M., Domschke, K., & Goldman, D. (2016). Making sense of epige-
Therapeutics, 37, 499–501. netics. The International Journal of Neuropsychopharmacology, 19.
Perroud, N., Paoloni-Giacobino, A., Prada, P., Olie, E., Salzmann, A., Nicastro, R., ... Shoemaker, R., Deng, J., Wang, W., & Zhang, K. (2010). Allele-specific methylation is
Malafosse, A. (2011). Increased methylation of glucocorticoid receptor gene (NR3C1) prevalent and is contributed by CpG-SNPs in the human genome. Genome Research,
in adults with a history of childhood maltreatment: A link with the severity and type 20, 883–889.
of trauma. Translational Psychiatry, 1, e59. Shumay, E., Logan, J., Volkow, N. D., & Fowler, J. S. (2012). Evidence that the methy-
Perroud, N., Rutembesa, E., Paoloni-Giacobino, A., Mutabaruka, J., Mutesa, L., Stenz, L., lation state of the monoamine oxidase A (MAOA) gene predicts brain activity of MAO
... Karege, F. (2014). The Tutsi genocide and transgenerational transmission of ma- A enzyme in healthy men. Epigenetics, 7, 1151–1160.
ternal stress: Epigenetics and biology of the HPA axis. The World Journal of Biological Singh-Taylor, A., Molet, J., Jiang, S., Korosi, A., Bolton, J. L., Noam, Y., ... Baram, T. Z.
Psychiatry, 15, 334–345. (2018). NRSF-dependent epigenetic mechanisms contribute to programming of
Perroud, N., Salzmann, A., Prada, P., Nicastro, R., Hoeppli, M. E., Furrer, S., ... Malafosse, stress-sensitive neurons by neonatal experience, promoting resilience. Molecular
A. (2013). Response to psychotherapy in borderline personality disorder and me- Psychiatry, 23, 648–657.
thylation status of the BDNF gene. Translational Psychiatry, 3, e207. Sipahi, L., Uddin, M., Hou, Z. C., Aiello, A. E., Koenen, K. C., Galea, S., & Wildman, D. E.
van Praag, H., Kempermann, G., & Gage, F. H. (2000). Neural consequences of environ- (2014). Ancient evolutionary origins of epigenetic regulation associated with post-
mental enrichment. Nature Reviews. Neuroscience, 1, 191–198. traumatic stress disorder. Frontiers in Human Neuroscience, 8, 284.
Prados, J., Stenz, L., Courtet, P., Prada, P., Nicastro, R., Adouan, W., ... Perroud, N. Skinner, M. K. (2015). Environmental epigenetics and a unified theory of the molecular
(2015). Borderline personality disorder and childhood maltreatment: A genome-wide aspects of evolution: A Neo-Lamarckian concept that facilitates Neo-Darwinian evo-
methylation analysis. Genes, Brain, and Behavior, 14, 177–188. lution. Genome Biology and Evolution, 7, 1296–1302.
Provencal, N., & Binder, E. B. (2015). The effects of early life stress on the epigenome: Smearman, E. L., Almli, L. M., Conneely, K. N., Brody, G. H., Sales, J. M., Bradley, B., ...
From the womb to adulthood and even before. Experimental Neurology, 268, 10–20. Smith, A. K. (2016). Oxytocin receptor genetic and epigenetic variations: Association
Provencal, N., Suderman, M. J., Guillemin, C., Massart, R., Ruggiero, A., Wang, D., ... with child abuse and adult psychiatric symptoms. Child Development, 87, 122–134.
Szyf, M. (2012). The signature of maternal rearing in the methylome in rhesus ma- Smith, A. K., Kilaru, V., Klengel, T., Mercer, K. B., Bradley, B., Conneely, K. N., ... Binder,
caque prefrontal cortex and T cells. The Journal of Neuroscience, 32, 15626–15642. E. B. (2015). DNA extracted from saliva for methylation studies of psychiatric traits:
Radtke, K. M., Ruf, M., Gunter, H. M., Dohrmann, K., Schauer, M., Meyer, A., & Elbert, T. evidence tissue specificity and relatedness to brain. American Journal of Medical
(2011). Transgenerational impact of intimate partner violence on methylation in the Genetics. Part B, Neuropsychiatric Genetics, 168b, 36–44.
promoter of the glucocorticoid receptor. Translational Psychiatry, 1, e21. Smoller, J. W. (2016). The genetics of stress-related disorders: PTSD, depression, and
Radtke, K. M., Schauer, M., Gunter, H. M., Ruf-Leuschner, M., Sill, J., Meyer, A., & Elbert, anxiety disorders. Neuropsychopharmacology, 41, 297–319.
T. (2015). Epigenetic modifications of the glucocorticoid receptor gene are associated Stahl, S. M. (2012). Psychotherapy as an epigenetic 'drug': Psychiatric therapeutics target
with the vulnerability to psychopathology in childhood maltreatment. Translational symptoms linked to malfunctioning brain circuits with psychotherapy as well as with
Psychiatry, 5, e571. drugs. Journal of Clinical Pharmacy and Therapeutics, 37, 249–253.
Reif, A., Weber, H., Domschke, K., Klauke, B., Baumann, C., Jacob, C. P., ... Deckert, J. Stenz, L., Schechter, D. S., Serpa, S. R., & Paoloni-Giacobino, A. (2018). Intergenerational
(2012). Meta-analysis argues for a female-specific role of MAOA-uVNTR in panic transmission of DNA methylation signatures associated with early life stress. Current
disorder in four European populations. American Journal of Medical Genetics. Part B, Genomics, 19, 665–675.
Neuropsychiatric Genetics, 159B, 786–793. Stoffel, M., Aguilar-Raab, C., Rahn, S., Steinhilber, B., Witt, S. H., Alexander, N., & Ditzen,
Reynolds, S., Lane, S. J., & Richards, L. (2010). Using animal models of enriched en- B. (2019). Effects of mindfulness-based stress prevention on serotonin transporter
vironments to inform research on sensory integration intervention for the re- gene methylation. Psychotherapy and Psychosomatics, 88, 317–319.
habilitation of neurodevelopmental disorders. Journal of Neurodevelopmental Strachan, E., Zhao, J., Roy-Byrne, P. P., Fowler, E., & Bacus, T. (2018). Study design and
Disorders, 2, 120–132. rationale for the mood and methylation study: A platform for multi-omics in-
Rijlaarsdam, J., Pappa, I., Walton, E., Bakermans-Kranenburg, M. J., Mileva-Seitz, V. R., vestigation of depression in twins. Twin Research and Human Genetics, 21, 507–513.
Rippe, R. C., ... van, I. M. H. (2016). An epigenome-wide association meta-analysis of Ströhle, A. (2009). Physical activity, exercise, depression and anxiety disorders. Journal of
prenatal maternal stress in neonates: A model approach for replication. Epigenetics, Neural Transmission (Vienna), 116, 777–784.
11, 140–149. Suzuki, M. M., & Bird, A. (2008). DNA methylation landscapes: Provocative insights from
Riley, B., & Kendler, K. S. (2006). Molecular genetic studies of schizophrenia. European epigenomics. Nature Reviews. Genetics, 9, 465–476.
Journal of Human Genetics, 14, 669–680. Sweder, K. S., & Hanawalt, P. C. (1993). Transcription-coupled DNA repair. Science, 262,
Roberts, S., Keers, R., Breen, G., Coleman, J. R. I., Johren, P., Kepa, A., ... Wong, C. C. Y. 439–440.
(2019). DNA methylation of FKBP5 and response to exposure-based psychological Szyf, M. (2015). Epigenetics, a key for unlocking complex CNS disorders? Therapeutic
therapy. American Journal of Medical Genetics. Part B, Neuropsychiatric Genetics, 180, implications. European Neuropsychopharmacology, 25, 682–702.
150–158. Szyf, M., & Bick, J. (2013). DNA methylation: A mechanism for embedding early life
Roberts, S., Keers, R., Lester, K. J., Coleman, J. R., Breen, G., Arendt, K., ... Wong, C. C. experiences in the genome. Child Development, 84, 49–57.
(2015). Hpa axis related genes and response to psychological therapies: Genetics and Tay, N., Macare, C., Liu, Y., Ruggeri, B., Jia, T., Chu, C., ... Consortium, I. (2019). Allele-
epigenetics. Depression and Anxiety, 32, 861–870. specific methylation of SPDEF: a novel moderator of psychosocial stress and sub-
Roberts, S., Lester, K. J., Hudson, J. L., Rapee, R. M., Creswell, C., Cooper, P. J., ... Eley, T. stance abuse. The American Journal of Psychiatry, 176, 146–155.
C. (2014). Serotonin transporter [corrected] methylation and response to cognitive Teicher, M. H., & Samson, J. A. (2013). Childhood maltreatment and psychopathology: A
behaviour therapy in children with anxiety disorders. Translational Psychiatry, 4, case for ecophenotypic variants as clinically and neurobiologically distinct subtypes.
e444. The American Journal of Psychiatry, 170, 1114–1133.
Rodgers, A. B., Morgan, C. P., Bronson, S. L., Revello, S., & Bale, T. L. (2013). Paternal Teschler, S., Bartkuhn, M., Kunzel, N., Schmidt, C., Kiehl, S., Dammann, G., & Dammann,
stress exposure alters sperm microRNA content and reprograms offspring HPA stress R. (2013). Aberrant methylation of gene associated CpG sites occurs in borderline
axis regulation. The Journal of Neuroscience, 33, 9003–9012. personality disorder. PLoS One, 8, e84180.
Rosenzweig, S. (1936). Some implicit common factors in diverse methods of psy- Thomas, C. R. (2015). Epigenetics and child psychiatry: Ethical and legal issues.
chotherapy. American Journal of Orthopsychiatry, 6, 412–415. Behavioral Sciences & the Law, 33, 644–652.
Rudenko, A., & Tsai, L. H. (2014). Epigenetic modifications in the nervous system and Thomas, M., Knoblich, N., Wallisch, A., Glowacz, K., Becker-Sadzio, J., Gundel, F., ...
their impact upon cognitive impairments. Neuropharmacology, 80, 70–82. Nieratschker, V. (2018). Increased BDNF methylation in saliva, but not blood, of
Rutten, B. P., Hammels, C., Geschwind, N., Menne-Lothmann, C., Pishva, E., Schruers, K., patients with borderline personality disorder. Clinical Epigenetics, 10, 109.
... Wichers, M. (2013). Resilience in mental health: Linking psychological and neu- Tiwari, A., & Gonzalez, A. (2018). Biological alterations affecting risk of adult psycho-
robiological perspectives. Acta Psychiatrica Scandinavica, 128, 3–20. pathology following childhood trauma: A review of sex differences. Clinical
Sayols-Baixeras, S., Subirana, I., Fernandez-Sanles, A., Senti, M., Lluis-Ganella, C., Psychology Review, 66, 69–79.
Marrugat, J., & Elosua, R. (2017). DNA methylation and obesity traits: An epigenome- Tozzi, L., Farrell, C., Booij, L., Doolin, K., Nemoda, Z., Szyf, M., ... Frodl, T. (2018).

18
M.A. Schiele, et al. Clinical Psychology Review 77 (2020) 101830

Epigenetic changes of FKBP5 as a link connecting genetic and environmental risk Yehuda, R., Lehrner, A., & Bierer, L. M. (2018). The public reception of putative epige-
factors with structural and functional brain changes in major depression. netic mechanisms in the transgenerational effects of trauma. Environmental
Neuropsychopharmacology, 43, 1138–1145. Epigenetics, 4 dvy018.
Tyrka, A. R., Price, L. H., Marsit, C., Walters, O. C., & Carpenter, L. L. (2012). Childhood Yeshurun, S., & Hannan, A. J. (2019). Transgenerational epigenetic influences of paternal
adversity and epigenetic modulation of the leukocyte glucocorticoid receptor: environmental exposures on brain function and predisposition to psychiatric dis-
Preliminary findings in healthy adults. PLoS One, 7, e30148. orders. Molecular Psychiatry, 24, 536–548.
Uher, R. (2014). Gene-environment interactions in severe mental illness. Frontiers in Yousefi, P., Huen, K., Dave, V., Barcellos, L., Eskenazi, B., & Holland, N. (2015). Sex
Psychiatry, 5, 48. differences in DNA methylation assessed by 450 K BeadChip in newborns. BMC
Unternaehrer, E., Bolten, M., Nast, I., Staehli, S., Meyer, A. H., Dempster, E., ... Genomics, 16, 911.
Meinlschmidt, G. (2016). Maternal adversities during pregnancy and cord blood Youssef, N. A., Lockwood, L., Su, S., Hao, G., & Rutten, B. P. F. (2018). The effects of
oxytocin receptor (OXTR) DNA methylation. Social Cognitive and Affective trauma, with or without PTSD, on the transgenerational DNA methylation alterations
Neuroscience, 11, 1460–1470. in human offsprings. Brain Sciences, 8.
Unternaehrer, E., Luers, P., Mill, J., Dempster, E., Meyer, A. H., Staehli, S., ... Zannas, A. S. (2016). Editorial perspective: Psychological stress and epigenetic aging -
Meinlschmidt, G. (2012). Dynamic changes in DNA methylation of stress-associated What can we learn and how can we prevent? Journal of Child Psychology and
genes (OXTR, BDNF) after acute psychosocial stress. Translational Psychiatry, 2, e150. Psychiatry, 57, 674–675.
Unternaehrer, E., Meyer, A. H., Burkhardt, S. C., Dempster, E., Staehli, S., Theill, N., ... Zannas, A. S., Arloth, J., Carrillo-Roa, T., Iurato, S., Roh, S., Ressler, K. J., ... Mehta, D.
Meinlschmidt, G. (2015). Childhood maternal care is associated with DNA methy- (2015). Lifetime stress accelerates epigenetic aging in an urban, African American
lation of the genes for brain-derived neurotrophic factor (BDNF) and oxytocin re- cohort: Relevance of glucocorticoid signaling. Genome Biology, 16, 266.
ceptor (OXTR) in peripheral blood cells in adult men and women. Stress, 18, 451–461. Zannas, A. S., & Chrousos, G. P. (2017). Epigenetic programming by stress and gluco-
Ursini, G., Bollati, V., Fazio, L., Porcelli, A., Iacovelli, L., Catalani, A., ... Bertolino, A. corticoids along the human lifespan. Molecular Psychiatry, 22, 640–646.
(2011). Stress-related methylation of the catechol-O-methyltransferase Val 158 allele Zannas, A. S., & West, A. E. (2014). Epigenetics and the regulation of stress vulnerability
predicts human prefrontal cognition and activity. The Journal of Neuroscience, 31, and resilience. Neuroscience, 264, 157–170.
6692–6698. Ziegler, C., Dannlowski, U., Brauer, D., Stevens, S., Laeger, I., Wittmann, H., ... Domschke,
Vinkers, C. H., Geuze, E., van Rooij, S. J. H., Kennis, M., Schur, R. R., Nispeling, D. M., ... K. (2015). Oxytocin receptor gene methylation: converging multilevel evidence for a
Boks, M. P. (2019). Successful treatment of post-traumatic stress disorder reverses role in social anxiety. Neuropsychopharmacology, 40, 1528–1538.
DNA methylation marks. Molecular Psychiatry. https://doi.org/10.1038/s41380-019- Ziegler, C., & Domschke, K. (2018). Epigenetic signature of MAOA and MAOB genes in
0549-3. mental disorders. Journal of Neural Transmission (Vienna), 125, 1581–1588.
Voisey, J., Lawford, B., Bruenig, D., Harvey, W., Morris, C. P., Young, R. M., ... Initiative, Ziegler, C., Grundner-Culemann, F., Schiele, M. A., Schlosser, P., Kollert, L., Mahr, M., ...
P. (2019). Differential BDNF methylation in combat exposed veterans and the asso- Domschke, K. (2019). The DNA methylome in panic disorder: A case-control and
ciation with exercise. Gene, 698, 107–112. longitudinal psychotherapy-epigenetic study. Translational Psychiatry, 9, 314.
Voisin, S., Eynon, N., Yan, X., & Bishop, D. J. (2015). Exercise training and DNA me- Ziegler, C., Richter, J., Mahr, M., Gajewska, A., Schiele, M. A., Gehrmann, A., ...
thylation in humans. Acta Physiologica (Oxford, England), 213, 39–59. Domschke, K. (2016). MAOA gene hypomethylation in panic disorder-reversibility of
Wampold, B. E., Mondin, G. W., Moody, M., Stich, F., Benson, K., & Ahn, H. N. (1997). A an epigenetic risk pattern by psychotherapy. Translational Psychiatry, 6, e773.
meta-analysis of outcome studies comparing bona fide psychotherapies: Empirically, Zubin, J., & Spring, B. (1977). Vulnerability–A new view of schizophrenia. Journal of
“all must have prizes” Psychological Bulletin, 122, 203–215. Abnormal Psychology, 86, 103–126.
Wang, C., Schroeder, F. A., & Hooker, J. M. (2014). Visualizing epigenetics: Current
advances and advantages in HDAC PET imaging techniques. Neuroscience, 264, Katharina Domschke, MA, MD, PhD, is Full Professor of Psychiatry and Chair of the
186–197. Department of Psychiatry and Psychotherapy, University of Freiburg, Germany.
Weaver, I. C., D'Alessio, A. C., Brown, S. E., Hellstrom, I. C., Dymov, S., Sharma, S., ... Additionally, she holds an Adjunct Professorship at the Medical University of Vienna,
Meaney, M. J. (2007). The transcription factor nerve growth factor-inducible protein Austria. Prof. Domschke completed her studies and postgraduate education in medicine
a mediates epigenetic programming: altering epigenetic marks by immediate-early and psychology at the University of Muenster, Germany, and Trinity College Dublin,
genes. The Journal of Neuroscience, 27, 1756–1768. Ireland, as well as at Boston University, Boston, MA, USA, and Maastricht University, The
Weder, N., Zhang, H., Jensen, K., Yang, B. Z., Simen, A., Jackowski, A., ... Kaufman, J. Netherlands. After her residency in psychiatry at the Universities of Bonn and Muenster,
(2014). Child abuse, depression, and methylation in genes involved with stress, Germany, she worked as an attending physician and associate professor at the
neural plasticity, and brain circuitry. Journal of the American Academy of Child and Department of Psychiatry, University of Muenster, Germany. In 2012, she was appointed
Adolescent Psychiatry, 53(417-24), e5. Full Professor of Psychiatry at the University of Wuerzburg, Germany, where she worked
Wells, A. (2002). Emotional disorders and metacognition: Innovative cognitive therapy. John as attending physician and from 2014 through 2016 as vice chair. Prof. Domschke’s
Wiley & Sons. clinical expertise is on diagnostics and therapy in adult psychiatry, particularly depres-
Whittle, N., & Singewald, N. (2014). HDAC inhibitors as cognitive enhancers in fear, sion, anxiety disorders, stress-related disorders and obsessive-compulsive disorders.
anxiety and trauma therapy: Where do we stand? Biochemical Society Transactions, 42, Scientifically, applying genetic/epigenetic, imaging genetic and therapy(epi)genetic ap-
569–581. proaches she is interested in the identification of multilevel markers for the prediction,
Wiegand, C., Heusser, P., Klinger, C., Cysarz, D., Bussing, A., Ostermann, T., & indicated prevention and personalized treatment of mental disorders. Her work is re-
Savelsbergh, A. (2018). Stress-associated changes in salivary microRNAs can be de- flected by to date over 260 publications in peer-reviewed journals, two books and 30 book
tected in response to the Trier Social Stress Test: An exploratory study. Scientific chapters. Prof. Domschke is a member of the National Academy of Sciences Leopoldina
Reports, 8, 7112. and has received the Research Award of the World Federation of Societies of Biological
Wochnik, G. M., Ruegg, J., Abel, G. A., Schmidt, U., Holsboer, F., & Rein, T. (2005). Psychiatry (WFSBP), the Fellowship Award of the World Psychiatric Association (WPA),
FK506-binding proteins 51 and 52 differentially regulate dynein interaction and the Fellowship Award of the European College of Neuropsychopharmacology (ECNP), the
nuclear translocation of the glucocorticoid receptor in mammalian cells. The Journal Research Award of the German Society of Biological Psychiatry (DGBP), and the Research
of Biological Chemistry, 280, 4609–4616. Award of the German Society of Psychiatry, Psychotherapy and Neurology (DGPPN).
Xiao, F. H., Wang, H. T., & Kong, Q. P. (2019). Dynamic DNA methylation during aging: A Prof. Domschke has received funding from, e.g., the EU, the German Research Foundation
"prophet" of age-related outcomes. Frontiers in Genetics, 10, 107. (DFG), and the German Ministry of Research and Education (BMBF). She is a fellow of the
Yang, B. Z., Zhang, H., Ge, W., Weder, N., Douglas-Palumberi, H., Perepletchikova, F., ... ACNP, full member of the ECNP, SOBP, ISPG and the German Society of Psychiatry,
Kaufman, J. (2013). Child abuse and epigenetic mechanisms of disease risk. American Psychotherapy and Neurology (DGPPN), executive secretary of the German Society of
Journal of Preventive Medicine, 44, 101–107. Biological Psychiatry (DGBP) and vice president of the German Society of Anxiety
Yehuda, R., Daskalakis, N. P., Bierer, L. M., Bader, H. N., Klengel, T., Holsboer, F., & Research (GAF). Prof. Domschke serves on the editorial boards of the International
Binder, E. B. (2016). Holocaust exposure induced intergenerational effects on FKBP5 Journal of Neuropsychopharmacology, Journal of Psychiatric Research, Nervenarzt,
methylation. Biological Psychiatry, 80, 372–380. PLoSONE, Pharmacopsychiatry, Progress in Neuropsychopharmacology & Biological
Yehuda, R., Daskalakis, N. P., Desarnaud, F., Makotkine, I., Lehrner, A. L., Koch, E., ... Psychiatry, Acta Neuropsychiatrica, World Journal of Biological Psychiatry, International
Bierer, L. M. (2013). Epigenetic biomarkers as predictors and correlates of symptom Journal of Methods in Psychiatric Research as well as Frontiers in Molecular Psychiatry,
improvement following psychotherapy in combat veterans with PTSD. Frontiers in and is a regular reviewer for over 60 SCI-listed international journals.
Psychiatry, 4, 118.

19

You might also like