Elsevier

Journal of Affective Disorders

Volume 249, 15 April 2019, Pages 336-346
Journal of Affective Disorders

Review article
Moving pharmacoepigenetics tools for depression toward clinical use

https://doi.org/10.1016/j.jad.2019.02.009Get rights and content

Highlights

  • Antidepressant response is associated with multiple epigenetic marks.

  • Many limitations exist in the study base of antidepressant pharmacoepigenetics.

  • Future studies should focus on overcoming these limitations.

  • Integrating multiple predictors into new tools will likely have the greatest utility.

Abstract

Background

Major depressive disorder (MDD) is a leading cause of disability worldwide, and over half of patients do not achieve symptom remission following an initial antidepressant course. Despite evidence implicating a strong genetic basis for the pathophysiology of MDD, there are no adequately validated biomarkers of treatment response routinely used in clinical practice. Pharmacoepigenetics is an emerging field that has the potential to combine both genetic and environmental information into treatment selection and further the goal of precision psychiatry. However, this field is in its infancy compared to the more established pharmacogenetics approaches.

Methods

We prepared a narrative review using literature searches of studies in English pertaining to pharmacoepigenetics and treatment of depressive disorders conducted in PubMed, Google Scholar, PsychINFO, and Ovid Medicine from inception through January 2019. We reviewed studies of DNA methylation and histone modifications in both humans and animal models of depression.

Results

Emerging evidence from human and animal work suggests a key role for epigenetic marks, including DNA methylation and histone modifications, in the prediction of antidepressant response. The challenges of heterogeneity of patient characteristics and loci studied as well as lack of replication that have impacted the field of pharmacogenetics also pose challenges to the development of pharmacoepigenetic tools. Additionally, given the tissue specific nature of epigenetic marks as well as their susceptibility to change in response to environmental factors and aging, pharmacoepigenetic tools face additional challenges to their development.

Limitations

This is a narrative and not systematic review of the literature on the pharmacoepigenetics of antidepressant response. We highlight key studies pertaining to pharmacoepigenetics and treatment of depressive disorders in humans and depressive-like behaviors in animal models, regardless of sample size or methodology. While we discuss DNA methylation and histone modifications, we do not cover microRNAs, which have been reviewed elsewhere recently.

Conclusions

Utilization of genome-wide approaches and reproducible epigenetic assays, careful selection of the tissue assessed, and integration of genetic and clinical information into pharmacoepigenetic tools will improve the likelihood of developing clinically useful tests.

Introduction

Despite a large body of research examining clinical and biological differences among individuals with major depressive disorder (MDD), findings from this work have not yet produced tools to reliably enhance treatment selection. When opting for a medication approach to treating MDD, clinicians’ choices are largely guided by potential side effects, past treatment responses, and clinical characteristics, such as the presence of comorbid anxiety, psychosis, trauma, or substance misuse. However, there is limited replicated evidence showing these characteristics to be robust moderators of antidepressant efficacy (Dunlop, 2015). Evidence-based moderators that some psychiatrists incorporate into antidepressant selection include the presence of melancholic features, which may preferentially respond to tricyclic antidepressants (TCAs) (Perry, 1996), and atypical features, which may respond better to monoamine oxidase inhibitors (MAOIs) than TCAs (Quitkin et al., 1991, Quitkin et al., 1990). Psychotic depression is the only clinical subtype that has strong evidence for a particular prescriptive pathway; that is, the combination of an antidepressant and an antipsychotic (Malhi et al., 2018).

Unguided selection of antidepressants produces response rates of only 50–60% and even lower remission rates for a single treatment course (Papakostas and Fava, 2009, Rush et al., 2006). Failure to achieve remission results in prolonged suffering, greater impairment in psychosocial functioning, and higher levels of health care use. The trial and error approach to treatment selection also suffers from high rates of side effects, with approximately 55% of patients experiencing at least one bothersome side effect from antidepressant treatment (Papakostas, 2008). While successive trial and error treatment trials appear to boost cumulative remission rates to 67% (Rush et al., 2006), the prolonged process to achieve remission leaves patients suffering from illness morbidity and elevates the risk of disengaging from care.

More recent work focusing on biological predictors, including neuroimaging, neurophysiological, neuroendocrine, and genetic measures has identified potential alternative approaches to treatment selection (Busch and Menke, 2019, Fonseka et al., 2018, Olbrich and Arns, 2013, Williams, 2017). The field of pharmacogenetics (PGx) developed with the idea that specific genetic variants can inform decisions about medication choice by helping to predict response to and tolerability of different medications. The potential utility of analyzing common genetic variants in antidepressant response is supported by the finding that 42% of individual variation in medication outcomes derives from genetic factors (Tansey et al., 2013).

Results from candidate gene studies of antidepressant response have been utilized to develop multiple pharmacogenetic-based decision support tools (DSTs), which vary widely in the number and type of genes assessed, included medications, cost, regulation, and method of results delivery. A recent review identified 76 labs in the US that offer PGx testing services (Haga and Kantor, 2018) with 38 DSTs that assess antidepressants (Fabbri et al., 2018). These tools are being marketed directly to patients and clinicians, though current depression treatment guidelines either do not address this type of testing or simply refer to it as an area of future research (Peterson et al., 2017, Zeier et al., 2018). A recent meta-analysis concluded that while there are concerns about the quality of the evidence base, PGx testing does hold promise for improving response and remission rates in MDD, particularly among patients with treatment resistance or intolerability to previous psychotropics (Bousman et al., 2019).

More recently, investigators have begun to examine ways in which epigenetic marks predict antidepressant response as part of the field of pharmacoepigenetics. Epigenetic modifications are changes to DNA structure that do not affect the DNA sequence but mediate alterations in gene expression, which, in turn, can influence protein levels. While some epigenetic marks appear inherited, most can be altered throughout a person's lifetime starting during prenatal development by multiple environmental factors, including exposure to pharmacotherapy (Kanherkar et al., 2014).

Epigenetic factors that have been most studied in the context of antidepressant response include DNA methylation, histone modifications, and the control of gene expression by non-coding RNAs. DNA methylation is the most widely studied and best understood epigenetic modification. It typically involves the addition of a methyl group to a cytosine within a CpG dinucleotide via DNA methyltransferase (DNMT), generating the modified nucleotide 5-methylcytosine (5mC), but less commonly involves adding a methyl group (or other variants of methyl groups) to cytosines within other types of dinucleotides. Generally, the presence of 5mC at gene promoters is associated with decreased gene expression, whereas intragenic methylation can induce gene transcription or silencing (Bonasio et al., 2010, Maunakea et al., 2010). The enzyme DNMT1 preserves DNA methylation patterns during replication, while DNMT3a and DNMT3b lead to de novo methylation of double-stranded DNA (Menke and Binder, 2014).

Histone modification refers to the enzymatic attachment or removal of chemical groups from lysine and arginine residues on histones’ N-terminal tails. Histones are found in nucleosomes, which consist of an octamer of histone proteins (two copies of H2A, H2B, H3, and H4 each) around which DNA is coiled (Sun et al., 2013). Acetylation is the most common histone modification and generally produces an increase in gene expression by inducing the formation of a more loosened and accessible chromatin (‘euchromatin’). N-terminal tails of histones can also be methylated with one, two, or three methyl groups. Methylation of histones can lead to transcriptional activation (H3-lysine (K)4, H3K36) or repression (H3K9, H3K27, H4K20) based on which histone and lysine is being methylated (Lachner et al., 2003).

There are multiple mechanisms by which antidepressants and antidepressant-like compounds have been shown to alter the epigenome. Evidence suggests that the TCAs amitriptyline and imipramine, the selective serotonin reuptake inhibitor (SSRI) paroxetine, and the antidepressant-like compound genipin (a molecule extracted from Gardenia jasminoides Ellis, i.e. cape jasmine) decrease DNA methylation by reducing DNMT1 enzymatic activity both in and ex vivo (Perisic et al., 2010, Ye et al., 2018, Zimmermann et al., 2012). Paroxetine has also been found to alter DNMT1 phosphorylation, which affects the enzyme's activity, in peripheral blood cells obtained from depressed patients (Gassen et al., 2015). Evidence suggests that the SSRI fluoxetine indirectly alters the epigenetic landscape through chronic elevation of serotonin, resulting in increased expression of methyl-CpG-binding proteins and induction of histone deacetylase (HDAC), an enzyme that removes acetyl groups from histones and represses gene transcription (Csoka and Szyf, 2009). Furthermore, the serotonin-norepinephrine reuptake inhibitor (SNRI) venlafaxine (Qiao et al., 2019) and imipramine (Tsankova et al., 2006) selectively down-regulate HDAC5 in rodent models of depression. There is also evidence that imipramine decreases activity of HDAC3 and HDAC4 in fetal mouse neocortical neurons (Nghia et al., 2015). Furthermore, multiple HDAC inhibitors have antidepressant effects in animal models, including valproic acid (Fuchikami et al., 2016).

Although the field of pharmacoepigenetics is quite young compared to the more established pharmacogenetics approach, an increasing body of preclinical and clinical work indicates that epigenetic marks may be useful for the prediction of treatment response in patients with MDD. Here, we review the current state of the field of pharmacoepigenetics of oral antidepressant response in human and animal models of depression with a focus on DNA methylation and histone modifications. We will not discuss non-coding RNAs, as they have recently been reviewed elsewhere in relation to antidepressant response (Belzeaux et al., 2018, Fiori et al., 2018). Given the issue of high tissue specificity in DNA methylation patterns (Ziller et al., 2013) and the fact that most DNA methylation studies in humans use blood, we used the freely available web application Blood-Brain Epigenetic Concordance, BECon (Edgar et al., 2017), to obtain a sense of the concordance of CpGs between blood and brain in genes for which we found two or more studies, including BDNF, SLC6A4, and HTR1B. We then discuss key issues with the current antidepressant pharmacoepigenetics literature and provide suggestions for future studies to aid in the development of clinically useful pharmacoepigenetic DSTs.

Section snippets

Methods

Studies were identified for inclusion in this narrative review by searching PubMed, Google Scholar, PsychINFO, and Ovid Medicine from inception through January 2019 (LMH, GRF, HAE, BTB, and BWD). Articles were limited to those published in English and could include either humans or animals as subjects. The literature search strategy was based on using combinations of the following keywords: epigenetic*, pharmacoepigenetic*, pharmacoepigenomic*, depression, major depressive disorder, DNA

Pharmacoepigenetic studies

The majority of studies of antidepressant pharmacoepigenetics have examined SSRIs in relation to epigenetic marks, although SNRIs, TCAs, MAOIs, and mirtazapine have also been assessed. Furthermore, compounds not traditionally considered to be antidepressants, including HDAC inhibitors, DNMT inhibitors, and genipin have also been studied. Below, we discuss the animal and human literature findings, structured by the gene studied and the observed baseline predictors or post-treatment changes

Discussion

How can pharmacoepigenetic tools be prepared for clinical use?

Unlike genetic assays, which measure static allele states that are immutable and constant across all cell types, epigenetic measures are tissue specific and susceptible to change over time with aging and environmental exposures (both physical and psychological). These tissue specific and dynamic aspects of epigenetic profiles increase both the challenge and the opportunities for developing pharmacoepigenetics tools to aid in

Acknowledgement

There are no acknowledgements for this manuscript.

Role of funding source

LMH received training support from the National Institutes of Health Grant R25MH101079.

Contributors

LMH, GRF, HAE, BTB, and BWD conducted the literature review and LMH wrote the original draft of the manuscript. GRF, HAE, CAB, ABS, JQ, VPJ, BTB, and BWD provided critical feedback and editing of the manuscript. All authors approved of the final submitted version.

Conflict of interest

GRF, VPJ, and JQ have no conflicts of interest to declare. LMH has received material support from CNSDose for research purposes but does not have equity, stocks, or options in this company or any other pharmacogenetic companies. ABS and HAE have equity in CNSDose LLC. CAB has received material support from Assurex, CNSDose, Genomind, and AB-Biotics for research purposes and has active research collaborations with CNSDose and MyDNA but does not have equity, stocks, or options in these companies

References (107)

  • KimJ.M. et al.

    BDNF methylation and depressive disorder in acute coronary syndrome: the K-DEPACS and EsDEPACS studies

    Psychoneuroendocrinology

    (2015)
  • B. Le Francois et al.

    Chronic mild stress and antidepressant treatment alter 5-HT1A receptor expression by modifying DNA methylation of a conserved Sp4 site

    Neurobiol. Dis.

    (2015)
  • A.F. Leuchter et al.

    Comparative effectiveness of biomarkers and clinical indicators for predicting outcomes of SSRI treatment in Major Depressive Disorder: results of the BRITE-MD study

    Psychiatry Res.

    (2009)
  • A.J. Lisoway et al.

    DNA methylation and clinical response to antidepressant medication in major depressive disorder: a review and recommendations

    Neurosci. Lett.

    (2018)
  • E.M. Meylan et al.

    The HDAC inhibitor SAHA improves depressive-like behavior of CRTC1-deficient mice: possible relevance for treatment-resistant depression

    Neuropharmacology

    (2016)
  • N.A. Nghia et al.

    Long-term imipramine treatment increases N-methyl-d-aspartate receptor activity and expression via epigenetic mechanisms

    Eur. J. Pharmacol.

    (2015)
  • G. Oh et al.

    DNA modification study of major depressive disorder: beyond locus-by-locus comparisons

    Biol. Psychiatry

    (2015)
  • S. Okada et al.

    The potential of SLC6A4 gene methylation analysis for the diagnosis and treatment of major depression

    J. Psychiatr. Res.

    (2014)
  • G.I. Papakostas et al.

    Does the probability of receiving placebo influence clinical trial outcome? A meta-regression of double-blind, randomized clinical trials in MDD

    Eur. Neuropsychopharmacol.

    (2009)
  • P.J. Perry

    Pharmacotherapy for major depression with melancholic features: relative efficacy of tricyclic versus selective serotonin reuptake inhibitor antidepressants

    J. Affect. Disord.

    (1996)
  • A.J. Sales et al.

    Antidepressant administration modulates stress-induced DNA methylation and DNA methyltransferase expression in rat prefrontal cortex and hippocampus

    Behav. Brain Res.

    (2018)
  • A. Tadić et al.

    The early non-increase of serum BDNF predicts failure of antidepressant treatment in patients with major depression: a pilot study

    Prog. Neuropsychopharmacol. Biol. Psychiatry

    (2011)
  • K.E. Tansey et al.

    Contribution of common genetic variants to antidepressant response

    Biol. Psychiatry

    (2013)
  • G. Turecki et al.

    Effects of the social environment and stress on glucocorticoid receptor gene methylation: a systematic review

    Biol. Psychiatry

    (2016)
  • WangP. et al.

    HTR1A/1B DNA methylation may predict escitalopram treatment response in depressed Chinese Han patients

    J. Affect. Disord.

    (2018)
  • R. Bayles et al.

    Methylation of the SLC6a2 gene promoter in major depression and panic disorder

    PloS One

    (2013)
  • S. Bludau et al.

    Medial prefrontal aberrations in major depressive disorder revealed by cytoarchitectonically informed voxel-based morphometry

    Am. J. Psychiatry

    (2016)
  • C. Bock et al.

    Quantitative comparison of DNA methylation assays for biomarker development and clinical applications

    Nat. Biotechnol.

    (2016)
  • R. Bonasio et al.

    Molecular signals of epigenetic states

    Science

    (2010)
  • L. Booij et al.

    DNA methylation of the serotonin transporter gene in peripheral cells and stress-related changes in hippocampal volume: a study in depressed patients and healthy controls

    PloS One

    (2015)
  • C.A. Bousman et al.

    Pharmacogenetic tests and depressive symptom remission: a meta-analysis of randomized controlled trials

    Pharmacogenomics

    (2019)
  • Y. Busch et al.

    Blood-based biomarkers predicting response to antidepressants

    J. Neural Transm. (Vienna)

    (2019)
  • M. Bysani et al.

    Epigenetic alterations in blood mirror age-associated DNA methylation and gene expression changes in human liver

    Epigenomics

    (2017)
  • ChenE.S. et al.

    The epigenetic effects of antidepressant treatment on human prefrontal cortex BDNF expression

    Int. J. Neuropsychopharmacol.

    (2011)
  • L. de Boni et al.

    DNA methylation alterations in iPSC- and hESC-derived neurons: potential implications for neurological disease modeling

    Clin. Epigenet.

    (2018)
  • J.O. de Jong et al.

    Epigenetic effects of electroconvulsive seizures

    J. ECT

    (2014)
  • K. Domschke et al.

    Serotonin transporter gene hypomethylation predicts impaired antidepressant treatment response

    Int. J. Neuropsychopharmacol.

    (2014)
  • K. Domschke et al.

    Pharmacoepigenetics of depression: no major influence of MAO-A DNA methylation on treatment response

    J. Neural. Transm. (Vienna)

    (2015)
  • F. Duclot et al.

    Epigenetic mechanisms underlying the role of brain-derived neurotrophic factor in depression and response to antidepressants

    J. Exp. Biol.

    (2015)
  • B.W. Dunlop

    Prediction of treatment outcomes in major depressive disorder

    Expert Rev Clin Pharmacol

    (2015)
  • R.D. Edgar et al.

    BECon: a tool for interpreting DNA methylation findings from blood in the context of brain

    Transl. Psychiatry

    (2017)
  • E. Elliott et al.

    Resilience to social stress coincides with functional DNA methylation of the Crf gene in adult mice

    Nat. Neurosci.

    (2010)
  • A. Etiévant et al.

    Repetitive transcranial magnetic stimulation induces long-lasting changes in protein expression and histone acetylation

    Sci. Rep.

    (2015)
  • P. Farré et al.

    Concordant and discordant DNA methylation signatures of aging in human blood and brain

    Epigenet. Chromatin

    (2015)
  • B.S. Fernandes et al.

    The new field of 'precision psychiatry'

    BMC Med.

    (2017)
  • B.S. Gadad et al.

    Association of Novel ALX4 Gene Polymorphisms with Antidepressant Treatment Response: findings from the CO-MED Trial

    Mol. Neuropsychiatry

    (2018)
  • N.C. Gassen et al.

    Chaperoning epigenetics: FKBP51 decreases the activity of DNMT1 and mediates epigenetic effects of the antidepressant paroxetine

    Sci. Signal

    (2015)
  • Common genetic variation and antidepressant efficacy in major depressive disorder: a meta-analysis of three genome-wide pharmacogenetic studies

    Am. J. Psychiatry

    (2013)
  • J.A. Gross et al.

    Characterizing 5-hydroxymethylcytosine in human prefrontal cortex at single base resolution

    BMC Genom.

    (2015)
  • GuoJ.U. et al.

    Distribution, recognition and regulation of non-CpG methylation in the adult mammalian brain

    Nat. Neurosci.

    (2014)
  • Cited by (22)

    • Integrated genome-wide methylation and expression analyses provide predictors of diagnosis and early response to antidepressant in panic disorder

      2023, Journal of Affective Disorders
      Citation Excerpt :

      Epigenetic mechanism participates in the pathogenesis of disease but also may affect drug efficacy. Evidence from human and animal studies showed that epigenetic markers (DNA methylation and histone modification) play a key role in predicting the efficacy response of antidepressants (Hack et al., 2019). These studies focused mainly on depressive disorder (Lisoway et al., 2018).

    • Swimming exercise ameliorates depressive-like behavior by anti-inflammation activity, rebalancing gut Escherichia coli and Lactobacilli

      2022, Brain Research
      Citation Excerpt :

      However, less than half of patients globally receive anti-depression therapy, and there are still great difficulties and challenges regarding the diagnosis and treatment of depression (Tiller, 2013). Similar limitations exist for treatment of major depression (Hack et al., 2019). Antidepressants have some disadvantages, such as delayed onset of therapeutic effect, high ratio of non-responding patients and undesirable side effects (Burcu et al., 2017; Coupland et al., 2018; Dragioti et al., 2019).

    • The interaction of P11 methylation and early-life stress impacts the antidepressant response in patients with major depressive disorder

      2022, Journal of Affective Disorders
      Citation Excerpt :

      In addition, the interactions between DNA methylation and stress affect the response to antidepressants (Shen et al., 2020; Wang et al., 2018a,b). These findings suggest that pharmacoepigenetics is an emerging field showing the potential to integrate genetic and environmental information into treatment selection and advance the goal of precision psychiatry (Hack et al., 2019). Protein P11 (also named S100A10) is a critical member of the S100 calcium-binding protein family (Oh et al., 2013; Svenningsson et al., 2013).

    • Depression management and pharmacoepigenetics

      2022, Epigenetics of Stress and Stress Disorders
    • Epigenetics of bipolar disorder

      2021, Epigenetics in Psychiatry
    View all citing articles on Scopus
    View full text