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.
Keywords: Pharmacoepigenetics, Depression, Antidepressants, Decision Support Tools (DSTs), DNA methylation, Histones
1. 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, 2018; 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 pharmacogenetic 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, pharmacogenetic 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 to 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, which in turn increases expression of methyl-CpG-binding protein, a transcription factor involved in DNA methylation, and a specific histone deacetylase (HDAC), an enzyme that removes acetyl groups from histones (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). In addition to valproic acid, multiple other HDAC inhibitors have antidepressant effects in animal models (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.
2. 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 methylation, acetylation, histone*, antidepressant response, mice, rats, and animal models.
3. Results
3.1. 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 reported. Table 1 lists pharmacoepigenetic studies of antidepressant response in humans and Table 2 shows the same in rodents.
Table 1.
Gene | Epigenetic mark | Baseline or Post-treatment | Antidepressant | Sample | Tissue | Outcome | Time | Reference |
---|---|---|---|---|---|---|---|---|
BDNF | 5mC of 12 CpGs in exon IV promoter | Baseline | SSRIs, SNRIs, atypicals, TCAs, and MAOIs | MDD patients (n=39) | Leukocytes | ↓ 5mC in 1 CpG ↓ response ↓ BDNF expression |
2–6 weeks | Tadic et al. (2014) |
5mC of 9 CpGs in exon VI promoter | Baseline | Escitalopram | ACS patients with depressive disorder (n=255) | Whole blood |
↑ average 5mC ↑ response |
24 weeks | Kim et al. (2015) | |
5mC of 90 CpGs in exon I-IV | Baseline and Post-treatment | Escitalopram | MDD (n=85) | Whole blood |
↑ baseline 5mC in 4 CpGs ↑ response ↑post-treatment average |
8 weeks | Wang et al. (2018b) | |
H3K27me3 at exon IV promoter | Post-treatment | Multiple classes | Medicated MDD patients (n=7) and unmedicated MDD patients (n=11) |
Post-mortem PFC | ↓ H3K27me ↑ BDNF IV expression in medicated group |
n/a | Chen et al. (2011) | |
H3K27me3 at exon IV promoter | Post-treatment | Citalopram | MDD patients (n=25) | Whole blood | ↓ H3K27me and ↑ BDNF IV expression ↑ response |
8 weeks | Lopez et al. (2013) | |
SLC6A4 | 5mC of 9 CpGs within exon 1A | Baseline | Escitalopram | MDD patients (n=94) | Whole blood | ↑average 5mC ↑response |
6 weeks | Domschke et al. (2014) |
5mC of 7 CpGs within promoter | Baseline | Multiple antidepressants | MDD patients (n=108) | Leukocytes | No effect after correction for multiple testing ↑5mC in 1 CpG ↓ response (nominal association) |
12 weeks | Kang et al. (2013) | |
5mC of 10 CpGs within promoter | Post-treatment | SSRIs Dual acting antidepressants |
MDD patients (n=33) | Whole blood | ↑5mC in 2 CpGs associated with SSRI use but not dual acting antidepressants | n/a | Booij et al. (2015) | |
5mC of 81 CpGs within exon 1 promoter | Post-treatment | Paroxetine, fluvoxamine, or milnacipran | MDD patients (n=50) | Whole blood | ↑5mC in 1 CpG ↑response | 6 weeks | Okada et al. (2014) | |
SLC6A2 | 5mC of CpGs within promoter | Post-treatment | Citalopram, fluoxetine, or sertraline | MDD patients (n=5), panic disorder patients (n=4) | Whole blood | No association between 5mc and response | 3 months | Bayles et al. (2013) |
HTR1A | 5mC of CpGs in promoter | Baseline | Escitalopram | MDD (n=85) | Whole blood | ↓5mC in 1 CpG ↓response |
8 weeks | Wang et al. (2018a) |
HTR1B | 5mC of CpGs in promoter | Baseline | Escitalopram | MDD (n=85) | Whole blood | ↓5mC in 1 CpG ↓response |
8 weeks | Wang et al. (2018a) |
5mC of 7 CpGs within promoter | Baseline | Fluoxetine | Children and adolescents with MDD, OCD, or GAD (n=83) | Whole blood | ↓ average 5mC ↑response |
12 weeks | Gasso et al. (2017) | |
IL-11 | 5mC of 11 CpGs in intron | Baseline | Escitalopram and nortriptyline | MDD patients (n=113) | Whole blood | ↑5mC of CpG site 5 ↑response to escitalopram ↑5mC of CpG site 4 ↓response to nortriptyline |
12 weeks | Powell et al. (2013) |
MAO-A | 5mC of 43 CpGs in exon 1, intron 1 | Baseline | Escitalopram | MDD patients (n=94) | Whole blood | No association after correction for multiple testing | 6 weeks | Domschke et al. (2015) |
PPFIA4 and HS3ST1 | Genome-wide 5mC study | Baseline | Paroxetine | MDD patients (n=20) | Leukocytes | ↑5mC of 2 CpGs after multiple test correction ↑response |
6 weeks | Takeuchi et al. (2017) |
Abbreviations: 5mC – 5-methylcytosine; ACS – acute coronary syndrome; BDNF – brain-derived neurotrophic factor; DNMT1 – DNA methyltransferase 1; GAD – generalized anxiety disorder; HTR1A – 5-Hydroxytryptamine receptor 1A; HTR1B – 5-Hydroxytryptamine receptor 1B; HS3ST1 – heparin sulfate-glucosamine 3-sulfotransferase 1; IL11 – interleukin 11; MAO-A – monoamine oxidase A; MAO-I – monoamine oxidase inhibitor; MDD – major depressive disorder; OCD – obsessive compulsive disorder; PFC – prefrontal cortex; PPFIA4 – PTPRF Interacting Protein Alpha 4; SLC6A2 – solute carrier family 6 member 2; SLC6A4 – solute carrier family 6 member 4; SNRI – serotonin-norepinephrine reuptake inhibitor; SSRI – selective serotonin reuptake inhibitor; TCA – tricyclic antidepressants.
Table 2.
Gene | Epigenetic Mark | Baseline or Post-treatment | Antidepressant | Species/strain, model | Tissue | Outcome | Reference |
---|---|---|---|---|---|---|---|
Bdnf | 5mC within promoter IV | Post-treatment | Fluoxetine | C57BL/6 J mice, post-stroke depression model | Hippocampus | ↑5mC in 2 CpG sites ↑ Bdnf expression ↓ depression-like behaviors |
Jin et al. (2017) |
H4K12ac within promoter III | Post-treatment | Fluoxetine + HDAC inhibitor | Balb/c mice, infant maternal separation | Forebrain neocortex | ↑ H4K12ac ↑ Bdnf expression ↓ depression-like behaviors |
Schmauss (2015) | |
H3ac within promoter III and IV | Post-treatment | Imipramine | Adult male Bl6/C57 mice, chronic social stress model | Hippocampus | ↑ H3ac within promoters III and VI ↑ Bdnf IV expression |
Tsankova et al. (2006) | |
H3K9me2 downstream of exon IV | Post-treatment | Tranylcypromine | C57BL/6J mice, social stress model | mPFC | ↓ H3K9me2 ↑ Bdnf IV expression reversed cognitive inflexibility of social stress |
Xu et al. (2018) | |
5mC within promoter | Post-treatment | Genipin | PRS mice (offspring of prenatally stressed dams) | Hippocampus | 5mC demethylation through inhibition of DNMT1 normalized Bdnf expression reversed depressive-like behaviors |
Ye et al. (2018) | |
- | Post-treatment | SAHA | Crtc1(-/) mice | PFC | ↑ Bdnf IV expression partially rescued depressive-like behavior |
Meylan et al. (2016) | |
Htr1a | 5mC within promoter |
Post-treatment | Imipramine | Adult male BALB/c Mice exposed to unpredictable chronic mild stress | PFC, midbrain | ↓ 5mC of 1 CpG reversed depressive-like phenotype |
Le Francois et al. (2015) |
S100a10 | 5mC within promoter | Post-treatment | Escitalopram | Female FSL rodent model of depression | PFC | ↓ 5mC ↑ S100a10 expression |
Melas et al. (2012) |
Crf | 5mC within promoter | Post-treatment | Imipramine | Adult mice exposed to social defeat | Hypothalamus | ↑ 5mC at 5 CpGs ↓ Crf expression ↓ social avoidance behavior |
Elliott et al. (2010) |
Nr2b | H3K9ac and H3K27ac within promoter | Post-treatment | Imipramine | Mouse fetuses | Neocortical neurons | ↑ H3ac ↑ Nrb2 expression ↓ HDAC3 and HDAC4 activity |
Nghia et al. (2015) |
Grm2 | H3K27ac within promoter | Post-treatment | LAC | FSL rodent model of depression | Hippocampus, PFC | ↑ H3K27ac ↑ Grm2 expression |
Nasca et al. (2013) |
Abbreviations: 5mC – 5-methylcytosine; Bdnf – brain-derived neurotrophic factor; Crf – corticotrophin-releasing factor; FSL – Flinders Sensitive Line; Grm2 – glutamate metabotropic receptor type; H3ac – histone 3 acetylation; H3K9me2 – histone 3 lysine 9 dimethylation; H4K12ac – histone 4 lysine 12 acetylation; H3K9ac – histone 3 lysine 9 acetylation; HDAC – histone deacetylase; Htr1a – 5-Hydroxytryptamine receptor 1A; LAC – L-acetylcarnitine; Nr2b – N-methyl D-aspartate receptor subtype 2B; PFC – prefrontal cortex; S100a10 – S100 Calcium Binding Protein A10; SAHA – suberoylanilide hydroxamic acid.
3.1.1. Locus-specific epigenetic marks
3.1.1.1. Baseline: Brain-Derived Neurotrophic Factor
Brain-derived neurotrophic factor (BDNF) is a neurotrophin belonging to a family of proteins involved in neuronal proliferation, migration, differentiation, and survival (Huang and Reichardt, 2001). For review of epigenetic marks affecting BDNF and their role in depression in both humans and animals, see Duclot and Kabbaj (2015). Multiple human and animal studies have found evidence that effective therapy with antidepressants increases peripheral levels of BDNF (Molendijk et al., 2011) and that an early lack of increase in plasma levels of BDNF predicts non-response to antidepressants (Tadić et al., 2011). Given this background, investigators examined the baseline methylation status of the promoter of BDNF exon IV in the leukocytes of MDD patients (n=39) in a naturalistic setting and found lower levels at this locus to be predictive of non-response to antidepressants belonging to multiple classes, including SSRIs, SNRIs, atypical antidepressants, TCAs, and MAOIs (Tadic et al., 2014). Another study of Korean patients with acute coronary syndrome and depressive disorder (n=255) found that pre-treatment BDNF hypermethylation in the exon VI promoter assessed in whole blood predicted improvement in depressive symptoms with escitalopram treatment (Kim et al., 2015). Using blood, Wang and colleagues found that baseline DNA hypermethylation in 4 CpGs across the promoters of multiple BDNF exons predicted better response to 8 weeks of escitalopram in a cohort of Han Chinese participants with MDD (n=85) (Wang et al., 2018b). Furthermore, they showed that average BDNF methylation across multiple exons was significantly increased after escitalopram treatment in remitters in a subset of participants (n=44). The investigators found that the interaction between lower life stress scores and higher DNA methylation predicted better treatment response, providing an example of how epigenetic marks and environmental exposures may be combined in future pharmacoepigenetic tools.
Figure 1 depicts a truncated version of the human BDNF gene and sites of baseline and post-treatment epigenetic changes related to antidepressant response. The study by Wang and colleagues (2018b) is not included because CpGs across multiple exons in BDNF were evaluated. The BECon application compares DNA methylation levels of 75 CpGs within the BDNF gene or promoters and three Brodmann areas (7,10, and 20) of 16 individuals (Edgar et al., 2017) between blood and brain. Given the relevance of Brodmann area 10 to depression (Bludau et al., 2016), we focused on the correlation between methylation sites in blood and this area of the brain. The analysis revealed that only 2 CpGs (2.7%) had strong positive or negative correlations (r >= 0.5), while 13 CpGs (17.3%) had moderate positive or negative correlations (r >= 0.3).
3.1.1.2. Post-treatment: Brain-Derived Neurotrophic Factor
Multiple antidepressants induce epigenetic effects in the Bdnf gene in animal models of depression. Using hippocampal tissue, fluoxetine has been shown to increase methylation in the Bdnf promoter IV associated with a decrease in depressive-like behaviors in post-stroke mice (Jin et al., 2017) and enrich acetylated histone 4 lysine 12 (H4K12) at the Bdnf promoter III in the forebrain neocortex of maternally separated mice when administered with an HDAC inhibitor (Schmauss, 2015). Bdnf expression was upregulated in both of these studies. Imipramine administration increased histone 3 acetylation at the Bdnf gene promoter III and IV in the hippocampus of mice exposed to social stress (Tsankova et al., 2006). In another study of mice using medial prefrontal cortex (PFC), Xu and colleagues found that the MAOI tranylcypromine reversed stress-induced increases in dimethylation of histone H3 at lysine 9 immediately downstream of the Bdnf exon IV promoter, suppression of gene transcription, and cognitive inflexibility (Xu et al., 2018). Furthermore, evidence suggests that genipin reverses depressive-like behaviors in prenatally stressed mice while inducing DNA demethylation of multiple Bdnf exon promoters in the hippocampus through inhibition of the enzyme DNMT1 (Ye et al., 2018). The authors suggest that genipin may inhibit DNMT1 activity indirectly by interfering with its transcription and/or more directly by disrupting the DNMT1 DNA-binding domain. DNA demethylation was accompanied by the normalization of previously downregulated Bdnf expression in the hippocampus of these mice. Furthermore, chronic systematic administration of the HDAC inhibitor suberoylanilide hydroxamic acid (SAHA) was found to improve depressive-like behavior in a mouse model of treatment-resistant depression and increase the expression of Bdnf in the prefrontal cortex (Meylan et al., 2016). The results of this animal work align well with evidence from humans mentioned previously that increased expression of BDNF correlates with successful antidepressant treatment (Molendijk et al., 2011).
In a small cross-sectional study, Chen and colleagues showed that the use of antidepressant medications from multiple classes was associated with decreased repressive histone methylation at the BDNF IV promoter in post-mortem PFC tissue from medicated MDD subjects (n=7) as compared to unmedicated subjects (n=11) (Chen et al., 2011). Additionally, using whole blood, Lopez and colleagues found a decrease in histone 3 lysine 27 trimethylation (H3K27me3) at the BDNF exon IV promoter correlated negatively with BDNF IV expression and depression severity after treatment with citalopram for 8 weeks as compared to baseline (Lopez et al., 2013).
3.1.1.3. Baseline: Monoaminergic transporter genes
The serotonin transporter, encoded by the solute carrier family 6, member 4 (SLC6A4) gene, is a key target of antidepressant drug action. In one study, higher average baseline methylation of the promotor of the SLC6A4 gene across 9 CpG sites assessed in whole blood was associated with better escitalopram response after 6 weeks of treatment in Caucasian MDD patients (n=94) (Domschke et al., 2014). However, another study found no association between methylation in the SLC6A4 promoter from leukocytes and response in MDD patients (n=108) treated with antidepressants for 12 weeks after correction for multiple testing but nominal associations were found between higher average methylation as well as at a particular CpG and worse response (Kang et al., 2013). Despite examining the same CpG sites, Domschke and colleagues suggest the differing results between these studies may be due to variability in treatment duration, sample ethnicity, or pharmacogenetic heterogeneity in the study by Kang et al. (Domschke et al., 2014). When we assessed the correlation between blood and brain of the 15 CpGs for SLC6A4 listed in BECon in Brodmann area 10 (Edgar et al., 2017), we found that 2 CpGs (13.3%) had strong positive or negative correlations (r >= 0.5), while 11 CpGs (73.3%) had moderate positive or negative correlations (r >= 0.3). To our knowledge, no other monoaminergic transporter genes have been tested with respect to baseline epigenetic status.
3.1.1.4. Post-treatment: Monoaminergic transporter genes
One small cross-sectional study found that two CpG sites assessed in whole blood in the SLC6A4 promoter were more highly methylated in MDD patients (n=33) treated with SSRIs compared to those who were on dual acting antidepressants (Booij et al., 2015). Additionally, post-treatment methylation of a particular CpG in SLC6A4 in whole blood was found to be significantly higher than baseline in a sample of Japanese MDD subjects (n=50) who showed better therapeutic responses after 6 weeks of antidepressant treatment with paroxetine, fluvoxamine, or milnacipran compared to those with poor responses (Okada et al., 2014). Solute carrier family 6, member 2 (SLC6A2) encodes the norepinephrine transporter, another key target of antidepressant drug action. Investigators measured methylation of the promoter of SLC6A2 in whole blood before and after SSRI treatment in a sample of patients with MDD (n=5) and panic disorder (n=4) and found no significant change (Bayles et al., 2013). However, the sample is too small to draw any conclusions about the role of SLC6A2 methylation in response to SSRI treatment.
3.1.1.5. Baseline: Serotonin receptor genes
Baseline hypomethylation of two CpG sites within serotonin receptor type 1A (HTR1A) and 1B (HTR1B) genes derived from whole blood was predictive of poor escitalopram response in a sample of Han Chinese patients with MDD (n=85) after 8 weeks of treatment (Wang et al., 2018a). This was the same group of subjects tested in the Wang et al. (2018b) BDNF article previously discussed. The authors further showed that the interaction of high recent life stress with low methylation in 3 CpG sites in the HTR1A gene and 1 CpG site in the HTR1B gene predicted poor escitalopram response. Another group showed that improvement in depressive symptoms after treatment with fluoxetine was correlated with lower baseline average methylation of the HTR1B gene promoter assessed in whole blood in 83 children and adolescents with MDD, obsessive compulsive disorder, or generalized anxiety disorder (Gasso et al., 2017). Wang and colleagues provide several possible reasons for the discrepancy in findings between their study and that of Gasso et al., including differences in ethnicity, diagnosis, antidepressant studied, and duration of treatment (Wang et al., 2018a). Other possible reasons for the contrasting results include differences in the 5mC analysis method and ages of the participants. Using the BECon application (Edgar et al., 2017) to evaluate the correlation between blood and brain of the 12 CpGs for HTR1B in Brodmann area 10, there were 0 CpGs (0%) with a strong positive or negative correlations (r >= 0.5) and 5 CpGs (41.7%) with a moderate positive or negative correlations (r >= 0.3).
3.1.1.6. Post-treatment: Serotonin receptor genes
One preclinical study assessed the effect of an antidepressant on methylation status of the Htr1a gene. Chronic imipramine administration reversed chronic stress-induced increases in the methylation of a CpG site within the promoter of the Htr1a gene in the prefrontal cortex and midbrain of mice, along with an associated increase in Htr1a expression (Le Francois et al., 2015).
3.1.1.7. Baseline: Other genes
A potential epigenetic moderator for choosing between nortriptyline and escitalopram was identified from the Genome-based Therapeutic Drugs for Depression study. MDD patients with baseline hypermethylation of a CpG site in the interleukin-11 (IL11) gene assessed in whole blood achieved better antidepressant response to escitalopram (n=80) but worse response to nortriptyline (n=33) (Powell et al., 2013). If replicated, this finding is an example of the clinical utility pharmacoepigenetic approaches could bring to clinical care in predicting differential response to treatment. The authors further demonstrated that a variant in the IL11 gene interacted with a CpG site to predict antidepressant response, providing an example of how both genetic and epigenetic information could be incorporated into future tools. Takeuchi and colleagues conducted a genome-wide DNA methylation analysis using peripheral leukocytes in a small sample of Japanese MDD patients (n=20) treated with paroxetine for 6 weeks (Takeuchi et al., 2017). The investigators showed that 218 CpGs were nominally significantly different between the 10 best responders and 10 worst responders, while 2 CpGs passed multiple test correction. These 2 sites were located in the PPFIA4 gene, which encodes the protein liprin-alpha-4 shown to be important in neural transmission and HS3ST1, which encodes the enzyme heparin sulfate glucosamine 3-O sulfotransferase 1 and has no known relationship with MDD or antidepressant response.
3.1.1.8. Post-treatment: Other genes
Epigenetic changes induced by antidepressants have been studied in multiple other genes in animal models of depression. Chronic escitalopram treatment reversed hypermethylation of the promoter of the S100 Calcium Binding Protein A10 (S100a10) gene in the PFC of the Flinders Sensitive Line (FSL) genetic rat model of depression (Melas et al., 2012), suggesting a potentially useful novel target worthy of examination in clinical MDD samples treated with escitalopram. Furthermore, investigators reported that imipramine reversed stress-induced methylation and expression changes of the corticotropin-releasing factor (Crf) gene in the hypothalami of mice exposed to social defeat and attenuated their social avoidance behavior (Elliott et al., 2010). In FSL rats, another HDAC inhibitor, L-acetylcarnitine (LAC), acted as a rapid and long-lasting antidepressant while inducing increased levels of acetylated H3K27 bound to the promotor of the glutamate metabotropic receptor type (Grm2) gene, thus enhancing transcription of this gene in the hippocampus and prefrontal cortex (Nasca et al., 2013).
3.1.2. Global epigenetic marks
Multiple preclinical studies have examined global epigenetic changes in response to antidepressant administration in animal models of depression. Chronic but not acute imipramine administration decreased stress-induced increases in global DNA methylation as well as DNMT3a and DNMT3b expression in the PFC of rats (Sales and Joca, 2018). The DNMT inhibitors 5-aza-2′-deoxycytidine (5-AzaD) and 5-azacytidine (5-AzaC) were found to improve depressive-like behaviors in mice, both when administered directly into the hippocampus and systemically (Sales et al., 2011). Moreover, the same authors demonstrated that DNMT inhibitors potentiate the behavioral effects of known antidepressant drugs, such as desipramine and fluoxetine, in a preclinical model of stress (Sales and Joca, 2016). Similar evidence exists with drugs that target histone modifications, including HDAC inhibitors. Investigators showed that chronic administration of the HDAC inhibitor sodium butyrate induced antidepressant-like effects in FSL rats with accompanying upregulation of the enzyme ten-eleven translocation methylcytosine dioxygenase 1 (TET1) in PFC (Wei et al., 2014). This enzyme converts 5mC into 5-hydroxymethycytosine (5hmC), an important step in the DNA demethylation process. Qiao and colleagues treated rats exposed to unpredictable stress with venlafaxine for 4 weeks and found the medication rescued depressive-like behaviors, significantly inhibited the global decrease of H3K9ac in the hippocampus, and downregulated the elevated expression of HDAC5 induced by stress (Qiao et al., 2019). In a human study, researchers demonstrated a significant association between clinical response to paroxetine after 6 weeks of treatment in MDD patients and phosphorylation of DNMT1 in isolated peripheral blood mononuclear cells from the same patients (Gassen et al., 2015).
3.2. Summary of current pharmacoepigenetic studies
Overall results of published antidepressant pharmacoepigenetic studies in humans and animal models of depression are of limited power and have inconsistent findings. This may partially be because the studies are heterogeneous in terms of genetic loci, tissue source, sample clinical characteristics, and type of antidepressant assessed. Given the limited number of studies and relatively small sample sizes to date, it is premature to make firm conclusions about the potential clinical utility of such as treatment biomarkers. However, preliminary data suggests that antidepressant medications from multiple classes modulate epigenetic mechanisms. Some of these changes have been correlated with antidepressant response in samples of depressed patients, providing a valuable opportunity for potential clinical application. Additionally, baseline epigenetic differences have been found between responders and non-responders to antidepressants. Thus, the emerging field of psychiatric pharmacoepigenetics may complement that of pharmacogenetics and offer ways to integrate genetic and environmental information. This may be particularly relevant in MDD, as there is evidence that environmental exposures, such as early-life adversity, affect treatment outcomes and progression of illness in some patients (Klein et al., 2009), probably partially through epigenetic modulation (Jawahar et al., 2015; Turecki and Meaney, 2016).
4. 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 antidepressant treatment selection. Much work needs to be done before pharmacoepigenetic measures could be incorporated into DSTs to guide treatment selection for patients with MDD. Key issues with the current evidence base and suggestions for future research are outlined in Table 3.
Table 3.
Key Issues | Suggestions for future research |
---|---|
1. May be missing relevant genes/loci due to lack of epigenome-wide association studies of antidepressant response | ● Conduct whole genome epigenetic studies in humans by leveraging consortia data in order to have the sample sizes needed for appropriate power. |
2. Lack of replication of particular epigenetic marks | ● Test for replication of epigenetic marks in large controlled clinical trials and determine validity of these marks by expert group |
3. Inconsistent methods for measuring epigenetic marks | ● Avoid methods known to have poor performance compared to other assays, such as enrichment bisulfite sequencing. |
4. Use of blood given low concordance between brain and blood methylation patterns | ● Use saliva or buccal samples given higher concordance with brain methylation patterns as compared to blood. ● If blood is used, limit to markers showing high concordance between brain and blood or generate iPSC-derived neurons. ● Blood may be appropriate when assessing pharmacokinetic genes. |
5. Lack of studies assessing non-CpG dinucleotides and 5hmC | ● Incorporate testing of non-CpG dinucleotides and 5hmC into future studies. |
6. Lack of studies with a comparator arm | ● Design studies to determine if differential response to antidepressants can be predicted by epigenetic marks. |
7. Variability in treatment duration, covariates included in the models, and use of correction for multiple testing | ● Standardize treatment duration and covariates in order to increase comparability between studies and correct for multiple testing. |
Abbreviations: 5hmC – 5-hydroxymethylcytosine; iPSC – induced pluripotent stem cell
One concern is that most studies thus far of antidepressant pharmacoepigenetics have been conducted using a candidate approach. Just as genome-wide association studies (GWAS) of antidepressant response have suggested regions and associated molecular pathways not considered using hypothesis-driven approaches (Gadad et al., 2018), there are likely candidates in the epigenetics of antidepressant response that have not been evaluated based on our understanding of the biology of depression. Data from individual studies will likely need to be pooled into consortia to have adequate power to detect effects significant at the epigenome-wide level. The aggregation of epigenetic variants significant at sub-threshold levels (i.e. polyepigenetic risk scores) may prove useful in predicting antidepressant response even if epigenome-wide significant marks are not initially identified, as has been the case with genetic variants of antidepressant response (GENDEP Investigators et al., 2013). As the field begins to accumulate evidence from EWAS of antidepressant response, it will benefit from curation of associated epigenetic marks into a knowledgebase like the EWAS Atlas (Li et al., 2018). Incorporating findings from EWAS along with well replicated loci from candidate studies with convergent evidence (e.g. from gene expression studies) into future DSTs will increase the likelihood of clinical utility..
Significant, clinically-useful marks must be validated by large controlled clinical trials with replication of findings in independent populations. In doing so, we will gain a better sense of whether epigenetic marks that predict treatment response are the same as those that change in response to treatment. As more evidence accumulates for particular epigenetic marks, inclusion in the pharmacogenetic knowledgebase (PharmGKB) (Whirl-Carrillo et al., 2012) would facilitate the integration and implementation of this information in the future. It would be ideal if the validity of replicated marks were determined by an international, expert group, like the Clinical Pharmacogenetics Implementation Consortium (CPIC) has done for the pharmacogenetics of depression (Hicks et al., 2015; Hicks et al., 2017), prior to incorporation into future tools.
It will be important for investigators who conduct future pharmacoepigenetic studies to select reproducible assays to ensure that pharmacoepigenetic tools have high analytic validity. For DNA methylation, extant pharmacoepigenetic studies of antidepressant response have also used a variety of assays. A study comparing 21 locus-specific DNA and 6 global methylation methods for clinical applications found good agreement across all approaches, although the amplicon bisulfite sequencing and bisulfite pyrosequencing showed the best all-round performance (Bock et al., 2016). Forthcoming studies should avoid methylation assays found to have poorer performance, such as enrichment bisulfite sequencing, which produced a high number of outliers compared to other assays (Bock et al., 2016). Furthermore, investigators who engage in future epigenome-wide studies of antidepressant response should be aware of the low concordance found for some CpGs between two widely used Illumina BeadChips and the potential need to filter out discordant probes (Logue et al., 2017).
An additional concern for pharmacoepigenetic studies is tissue source. Most human studies of epigenetic variation in relation to antidepressant treatment response have been conducted using blood samples, as neural tissue is unlikely to be obtained from live subjects. This is a concern given that, while some epigenetic marks have been shown to be highly correlated between brain and blood in the context of MDD (Oh et al., 2015), it is known that DNA methylation generally has strong tissue specificity (Farré et al., 2015). Because only easily accessible tissues could be collected in the clinical setting for use in a pharmacoepigenetics panel, we suggest the use of buccal cells (Lowe et al., 2013) or saliva samples (Smith et al., 2015), given that they show more similar methylation patterns than blood tissue. If blood is used in future studies for identifying additional loci, one possibility is to limit the selected CpGs to those that show high correlation between blood and brain using the freely available web application BECon (Edgar et al., 2017). The NIH’s Epigenomics Roadmap is another resource for identifying epigenetic marks in blood that may serve as proxies for those in brain (Kundaje et al., 2015). An additional possibility for future studies to identify additional loci of interest is to generate induced pluripotent stem cell (iPSC)-derived neurons from skin fibroblasts or from peripheral blood cells and expose these to antidepressants in vitro (Wang et al., 2015). Research has demonstrated that iPSC-derived neurons show similar methylation patterns as compared to human embryonic stem cells (de Boni et al., 2018); however, it is still unknown how similar their epigenetic profiles are compared to native neurons. Another limitation of this approach is the time required to generate iPSCs. Nevertheless, treating cells derived from depressed patients ex vivo and tracking epigenetic changes has already been shown to be a viable approach (Gassen et al., 2015). Researchers demonstrated that epigenetic changes observed in peripheral blood mononuclear cells derived from depressed patients and treated ex vivo with paroxetine mirrored changes seen in the same patients treated with various antidepressants. Furthermore, these changes correlated with clinical response.
While extant antidepressant pharmacoepigenetic studies assessing DNA methylation and histone modifications have only examined pharmacodynamic genes, we recommend future work also assess pharmacokinetic genes, given evidence that epigenetic marks affect the expression of cytochrome P450 genes (Ingelman-Sundberg et al., 2007). Blood would be a good tissue for this purpose, as there is evidence that blood methylation patterns are similar to those in the liver (Bysani et al., 2017). A concern related to tissue specificity is that, to our knowledge, no published studies of antidepressant pharmacoepigenetics have corrected for cell type, although DNA methylation is known to vary by cell type within the blood (Jaffe and Irizarry, 2014), saliva (Langie et al., 2017), and brain (Iwamoto et al., 2011). Thus, it will be important for future studies to correct for cellular proportions within the studied tissue.
Another limitation of the current pharmacoepigenetics literature is the focus on methylation of CpG dinucleotides given that approximately 25% of methylation is of cytosines within CpH (H = A/C/T) dinucleotides in the adult brain (Guo et al., 2014). Non-CpG dinucleotides display varying levels of methylation in different parts of the brain and there is evidence that they play a role in multiple neurological disorders, including Alzheimer’s Disease and Parkinson’s Disease (Jang et al., 2017; Oh et al., 2015), so it is reasonable to assume that they may be important in psychiatric disorders as well. Furthermore, common methods for assessing DNA methylation (i.e. bisulfite-based technologies) do not differentiate between 5mC and 5hmC, an intermediate in the active demethylation pathway. 5hmC is most abundant in the brain of mammals (Yardimci and Zhang, 2015), enriched in genes involved in neurodevelopment (Gross et al., 2015), and is modulated by stress (Hack et al., 2016), supporting an independent (and possibly stable) role for this epigenetic mark, particularly in psychiatric disorders.
Furthermore, most extant pharmacoepigenetics studies lack a comparator arm. Head-to-head trials of different antidepressants are needed to determine if epigenetic marks predict differential response to medications. Moreover, there needs to be greater consistency among studies in terms of patient characteristics, treatment duration, use of covariates, and multiple testing correction. This standardization will help decrease the possibility that lack of replication among studies assessing the same variants is due to heterogeneity in these variables. Future studies must also consider the impact of age, gender, other medications, and environmental factors on epigenetic marks.
Given evidence that antidepressant response likely arises from a complex interplay of genetic, epigenetic, and environmental factors, it would be advantageous to the field of pharmacoepigenetics to integrate genetic background, high-quality environmental information, and epigenetic marks in the same analysis and assess interactions between these variables. An interesting approach to address some limitations of currently available commercial pharmacogenetic panels would be to incorporate epigenetic marks in them along with clinical and environmental exposures, as opposed to creating new independent pharmacoepigenetic tools. This will be highly dependent upon clinical trials integrating multiple genetic and epigenetic marks at once. To take this one step further, given evidence from animal and human work that other therapies for depression, including TMS (Etiévant et al., 2015), ECT (de Jong et al., 2014), and cognitive behavioral therapy (Roberts et al., 2014) induce epigenetic changes, it is reasonable to think that there is a potential to incorporate marks related to these treatments in future tools.
A unique opportunity of pharmacoepigenetic approaches is the potential to use early changes (1–2 weeks) after initiating as a means of predicting eventual likely response. Most predictor studies of outcomes in MDD examine pre-treatment (baseline) variables. However, because unguided assessment of efficacy of antidepressants in individuals usually requires a six-week trial, tools that identify changes in a biomarker early in treatment could prove very valuable for indicating whether a medication should be continued or whether a switch should be made to an alternative treatment, thereby aborting the unnecessary time and potential side effects associated with a full six-week trial. Unreplicated studies using electroencephalogram measures (Hunter et al., 2010; Leuchter et al., 2009) or metabolomic (Kaddurah-Daouk et al., 2013; Zhu et al., 2013) changes have demonstrated the possibility of applying early change biomarkers for personalizing treatment. However, developing this application of pharmacoepigenetics will require improved understanding of the time course of change in epigenetic marks after initiating treatment. This is an area where human iPSCs may be particular valuable. Epigenetic assessments may also provide insights into the tachyphylaxis observed in some patients where a previously effective antidepressant loses benefit over time (Lisoway et al., 2018). This is a serious and understudied problem that affects a significant percentage of antidepressant-treated patients (Rothschild et al., 2009).
Because the field of antidepressant pharmacoepigenetics is only in its infancy and there are many pending milestones as reviewed in this manuscript, it will likely require several more years for pharmacoepigenetic tools to become clinically available for patients with MDD. Nevertheless, initial results are promising and warrant more substantial translational research. The development of DSTs that incorporate epigenetic marks is poised to be part of the exciting and rapidly growing area known as precision or personalized psychiatry, which holds the promise of providing personalized, patient-centered treatment based on an individual’s unique biological and environmental characteristics (Fernandes et al., 2017).
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.
Acknowledgements:
There are no acknowledgements for this manuscript.
Role of Funding Source: LMH was supported by the National Institutes of Health Grant MH101079.
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 or any other pharmacogenetic companies. BTB received speaker/consultation fees from AstraZeneca, Lundbeck, Pfizer, Takeda, Servier, Bristol Myers Squibb, Otsuka, and Janssen-Cilag. BWD has received research support from Acadia, Assurex, Axsome, Janssen, NIMH, Otsuka, and Takeda, and serves as a consultant to Aptinyx and Assurex Health, Inc.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of Interest: GRF, VPJ, and JQ have no conflicts of interest to declare.
References
- Bayles R, Baker EK, Jowett JB, Barton D, Esler M, El-Osta A, Lambert G, 2013. Methylation of the SLC6a2 gene promoter in major depression and panic disorder. PloS one 8, e83223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Belzeaux R, Lin R, Ju C, Chay MA, Fiori LM, Lutz PE, Turecki G, 2018. Transcriptomic and epigenomic biomarkers of antidepressant response. J Affect Disord 233, 36–44. [DOI] [PubMed] [Google Scholar]
- Bludau S, Bzdok D, Gruber O, Kohn N, Riedl V, Sorg C, Palomero-Gallagher N, Müller VI, Hoffstaedter F, Amunts K, Eickhoff SB, 2016. Medial Prefrontal Aberrations in Major Depressive Disorder Revealed by Cytoarchitectonically Informed Voxel-Based Morphometry. Am J Psychiatry 173, 291–298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bock C, Halbritter F, Carmona F, Tierling S, Datlinger P, Assenov Y, Berdasco M, Bergmann A, Booher K, Busato F, Campan M, Dahl C, Dahmcke C, Diep D, Fernández A, Gerhauser C, Haake A, Heilmann K, Holcomb T, Hussmann D, Ito M, Kläver R, Kreutz M, Kulis M, Lopez V, Nair S, Paul D, Plongthongkum N, Qu W, Queirós A, Reinicke F, Sauter G, Schlomm T, Statham A, Stirzaker C, Strogantsev R, Urdinguio R, Walter K, Weichenhan D, Weisenberger D, Beck S, Clark S, Esteller M, Ferguson-Smith A, Fraga M, Guldberg P, Hansen L, Laird P, Martín-Subero J, Nygren A, Peist R, Plass C, Shames D, Siebert R, Sun X, Tost J, Walter J, Zhang K, 2016. Quantitative comparison of DNA methylation assays for biomarker development and clinical applications. Nat Biotechnol 34, 726–737. [DOI] [PubMed] [Google Scholar]
- Bonasio R, Tu S, Reinberg D, 2010. Molecular signals of epigenetic states. Science 330, 612–616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Booij L, Szyf M, Carballedo A, Frey EM, Morris D, Dymov S, Vaisheva F, Ly V, Fahey C, Meaney J, Gill M, Frodl T, 2015. 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 10, e0119061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bousman CA, Arandjelovic K, Mancuso SG, Eyre HA, Dunlop BW, 2019. Pharmacogenetic tests and depressive symptom remission: a meta-analysis of randomized controlled trials. Pharmacogenomics 20, 37–47. [DOI] [PubMed] [Google Scholar]
- Busch Y, Menke A, 2018. Blood-based biomarkers predicting response to antidepressants. J Neural Transm (Vienna), doi: 10.1007/s00702-00018-01844-x. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
- Bysani M, Perfilyev A, de Mello VD, Rönn T, Nilsson E, Pihlajamäki J, Ling C, 2017. Epigenetic alterations in blood mirror age-associated DNA methylation and gene expression changes in human liver. Epigenomics 9, 105–122. [DOI] [PubMed] [Google Scholar]
- Chen ES, Ernst C, Turecki G, 2011. The epigenetic effects of antidepressant treatment on human prefrontal cortex BDNF expression. The international journal of neuropsychopharmacology 14, 427–429. [DOI] [PubMed] [Google Scholar]
- Csoka AB, Szyf M, 2009. Epigenetic side-effects of common pharmaceuticals: a potential new field in medicine and pharmacology. Med Hypotheses 73, 770–780. [DOI] [PubMed] [Google Scholar]
- de Boni L, Gasparoni G, Haubenreich C, Tierling S, Schmitt I, Peitz M, Koch P, Walter J, Wüllner U, Brüstle O, 2018. DNA methylation alterations in iPSC- and hESC-derived neurons: potential implications for neurological disease modeling. Clin Epigenetics 10, 13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Jong JO, Arts B, Boks MP, Sienaert P, van den Hove DL, Kenis G, van Os J, Rutten BP, 2014. Epigenetic effects of electroconvulsive seizures. J ECT 30, 152–159. [DOI] [PubMed] [Google Scholar]
- Domschke K, Tidow N, Schwarte K, Deckert J, Lesch KP, Arolt V, Zwanzger P, Baune BT, 2014. Serotonin transporter gene hypomethylation predicts impaired antidepressant treatment response. The international journal of neuropsychopharmacology 17, 1167–1176. [DOI] [PubMed] [Google Scholar]
- Duclot F, Kabbaj M, 2015. Epigenetic mechanisms underlying the role of brain-derived neurotrophic factor in depression and response to antidepressants. J Exp Biol 218, 21–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dunlop BW, 2015. Prediction of treatment outcomes in major depressive disorder. Expert Rev Clin Pharmacol 8, 669–672. [DOI] [PubMed] [Google Scholar]
- Edgar RD, Jones MJ, Meaney MJ, Turecki G, Kobor MS, 2017. BECon: a tool for interpreting DNA methylation findings from blood in the context of brain. Transl Psychiatry 7, e1187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elliott E, Ezra-Nevo G, Regev L, Neufeld-Cohen A, Chen A, 2010. Resilience to social stress coincides with functional DNA methylation of the Crf gene in adult mice. Nature neuroscience 13, 1351–1353. [DOI] [PubMed] [Google Scholar]
- Etiévant A, Manta S, Latapy C, Magno LA, Fecteau S, Beaulieu JM, 2015. Repetitive transcranial magnetic stimulation induces long-lasting changes in protein expression and histone acetylation. Sci Rep 5, 16873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fabbri C, Zohar J, Serretti A, 2018. Pharmacogenetic tests to guide drug treatment in depression: Comparison of the available testing kits and clinical trials. Prog Neuropsychopharmacol Biol Psychiatry 86, 36–44. [DOI] [PubMed] [Google Scholar]
- Farré P, Jones MJ, Meaney MJ, Emberly E, Turecki G, Kobor MS, 2015. Concordant and discordant DNA methylation signatures of aging in human blood and brain. Epigenetics Chromatin 8, 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fernandes BS, Williams LM, Steiner J, Leboyer M, Carvalho AF, Berk M, 2017. The new field of ‘precision psychiatry’. BMC Med 15, 80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fiori LM, Lin R, Ju C, Belzeaux R, Turecki G, 2018. Using Epigenetic Tools to Investigate Antidepressant Response. Prog Mol Biol Transl Sci 158, 255–272. [DOI] [PubMed] [Google Scholar]
- Fonseka TM, MacQueen GM, Kennedy SH, 2018. Neuroimaging biomarkers as predictors of treatment outcome in Major Depressive Disorder. J Affect Disord 233, 21–35. [DOI] [PubMed] [Google Scholar]
- Fuchikami M, Yamamoto S, Morinobu S, Okada S, Yamawaki Y, Yamawaki S, 2016. The potential use of histone deacetylase inhibitors in the treatment of depression. Prog Neuropsychopharmacol Biol Psychiatry 64, 320–324. [DOI] [PubMed] [Google Scholar]
- Gadad BS, Raj P, Jha MK, Carmody T, Dozmorov I, Mayes TL, Wakeland EK, Trivedi MH, 2018. Association of Novel ALX4 Gene Polymorphisms with Antidepressant Treatment Response: Findings from the CO-MED Trial. Mol Neuropsychiatry 4, 7–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gassen NC, Fries GR, Zannas AS, Hartmann J, Zschocke J, Hafner K, Carrillo-Roa T, Steinbacher J, Preissinger SN, Hoeijmakers L, Knop M, Weber F, Kloiber S, Lucae S, Chrousos GP, Carell T, Ising M, Binder EB, Schmidt MV, Ruegg J, Rein T, 2015. Chaperoning epigenetics: FKBP51 decreases the activity of DNMT1 and mediates epigenetic effects of the antidepressant paroxetine. Sci Signal 8, ra119. [DOI] [PubMed] [Google Scholar]
- Gasso P, Rodriguez N, Blazquez A, Monteagudo A, Boloc D, Plana MT, Lafuente A, Lazaro L, Arnaiz JA, Mas S, 2017. Epigenetic and genetic variants in the HTR1B gene and clinical improvement in children and adolescents treated with fluoxetine. Progress in neuro-psychopharmacology & biological psychiatry 75, 28–34. [DOI] [PubMed] [Google Scholar]
- GENDEP Investigators, MARS Investigators, STAR*D Investigators, 2013. Common genetic variation and antidepressant efficacy in major depressive disorder: a meta-analysis of three genome-wide pharmacogenetic studies. Am J Psychiatry 170, 207–217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gross JA, Pacis A, Chen GG, Barreiro LB, Ernst C, Turecki G, 2015. Characterizing 5-hydroxymethylcytosine in human prefrontal cortex at single base resolution. BMC Genomics 16, 672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guo JU, Su Y, Shin JH, Shin J, Li H, Xie B, Zhong C, Hu S, Le T, Fan G, Zhu H, Chang Q, Gao Y, Ming GL, Song H, 2014. Distribution, recognition and regulation of non-CpG methylation in the adult mammalian brain. Nat Neurosci 17, 215–222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hack LM, Dick ALW, Provençal N, 2016. Epigenetic mechanisms involved in the effects of stress exposure: focus on 5-hydroxymethylcytosine. Environ Epigenet 2, dvw016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haga SB, Kantor A, 2018. Horizon Scan Of Clinical Laboratories Offering Pharmacogenetic Testing. Health Aff (Millwood) 37, 717–723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hicks JK, Bishop JR, Sangkuhl K, Müller DJ, Ji Y, Leckband SG, Leeder JS, Graham RL, Chiulli DL, LLerena A, Skaar TC, Scott SA, Stingl JC, Klein TE, Caudle KE, Gaedigk A, Consortium CPI, 2015. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for CYP2D6 and CYP2C19 Genotypes and Dosing of Selective Serotonin Reuptake Inhibitors. Clin Pharmacol Ther 98, 127–134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hicks JK, Sangkuhl K, Swen JJ, Ellingrod VL, Müller DJ, Shimoda K, Bishop JR, Kharasch ED, Skaar TC, Gaedigk A, Dunnenberger HM, Klein TE, Caudle KE, Stingl JC, 2017. Clinical pharmacogenetics implementation consortium guideline (CPIC) for CYP2D6 and CYP2C19 genotypes and dosing of tricyclic antidepressants: 2016 update. Clin Pharmacol Ther 102, 37–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang EJ, Reichardt LF, 2001. Neurotrophins: roles in neuronal development and function. Annu Rev Neurosci 24, 677–736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hunter AM, Muthén BO, Cook IA, Leuchter AF, 2010. Antidepressant response trajectories and quantitative electroencephalography (QEEG) biomarkers in major depressive disorder. J Psychiatr Res 44, 90–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ingelman-Sundberg M, Sim SC, Gomez A, Rodriguez-Antona C, 2007. Influence of cytochrome P450 polymorphisms on drug therapies: pharmacogenetic, pharmacoepigenetic and clinical aspects. Pharmacol Ther 116, 496–526. [DOI] [PubMed] [Google Scholar]
- Iwamoto K, Bundo M, Ueda J, Oldham MC, Ukai W, Hashimoto E, Saito T, Geschwind DH, Kato T, 2011. Neurons show distinctive DNA methylation profile and higher interindividual variations compared with non-neurons. Genome Res 21, 688–696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jaffe AE, Irizarry RA, 2014. Accounting for cellular heterogeneity is critical in epigenome-wide association studies. Genome Biol 15, R31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jang HS, Shin WJ, Lee JE, Do JT, 2017. CpG and Non-CpG Methylation in Epigenetic Gene Regulation and Brain Function. Genes (Basel) 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jawahar MC, Murgatroyd C, Harrison EL, Baune BT, 2015. Epigenetic alterations following early postnatal stress: a review on novel aetiological mechanisms of common psychiatric disorders. Clinical epigenetics 7, 122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jin HJ, Pei L, Li YN, Zheng H, Yang S, Wan Y, Mao L, Xia YP, He QW, Li M, Yue ZY, Hu B, 2017. Alleviative effects of fluoxetine on depressive-like behaviors by epigenetic regulation of BDNF gene transcription in mouse model of post-stroke depression. Scientific reports 7, 14926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaddurah-Daouk R, Bogdanov MB, Wikoff WR, Zhu H, Boyle SH, Churchill E, Wang Z, Rush AJ, Krishnan RR, Pickering E, Delnomdedieu M, Fiehn O, 2013. Pharmacometabolomic mapping of early biochemical changes induced by sertraline and placebo. Transl Psychiatry 3, e223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kang HJ, Kim JM, Stewart R, Kim SY, Bae KY, Kim SW, Shin IS, Shin MG, Yoon JS, 2013. Association of SLC6A4 methylation with early adversity, characteristics and outcomes in depression. Progress in neuro-psychopharmacology & biological psychiatry 44, 23–28. [DOI] [PubMed] [Google Scholar]
- Kanherkar RR, Bhatia-Dey N, Csoka AB, 2014. Epigenetics across the human lifespan. Front Cell Dev Biol 2, 49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim JM, Stewart R, Kang HJ, Bae KY, Kim SW, Shin IS, Hong YJ, Ahn Y, Jeong MH, Yoon JS, 2015. BDNF methylation and depressive disorder in acute coronary syndrome: The K-DEPACS and EsDEPACS studies. Psychoneuroendocrinology 62, 159–165. [DOI] [PubMed] [Google Scholar]
- Klein DN, Arnow BA, Barkin JL, Dowling F, Kocsis JH, Leon AC, Manber R, Rothbaum BO, Trivedi MH, Wisniewski SR, 2009. Early adversity in chronic depression: clinical correlates and response to pharmacotherapy. Depression and anxiety 26, 701–710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kundaje A, Meuleman W, Ernst J, Bilenky M, Yen A, Heravi-Moussavi A, Kheradpour P, Zhang Z, Wang J, Ziller MJ, Amin V, Whitaker JW, Schultz MD, Ward LD, Sarkar A, Quon G, Sandstrom RS, Eaton ML, Wu YC, Pfenning AR, Wang X, Claussnitzer M, Liu Y, Coarfa C, Harris RA, Shoresh N, Epstein CB, Gjoneska E, Leung D, Xie W, Hawkins RD, Lister R, Hong C, Gascard P, Mungall AJ, Moore R, Chuah E, Tam A, Canfield TK, Hansen RS, Kaul R, Sabo PJ, Bansal MS, Carles A, Dixon JR, Farh KH, Feizi S, Karlic R, Kim AR, Kulkarni A, Li D, Lowdon R, Elliott G, Mercer TR, Neph SJ, Onuchic V, Polak P, Rajagopal N, Ray P, Sallari RC, Siebenthall KT, Sinnott-Armstrong NA, Stevens M, Thurman RE, Wu J, Zhang B, Zhou X, Beaudet AE, Boyer LA, De Jager PL, Farnham PJ, Fisher SJ, Haussler D, Jones SJ, Li W, Marra MA, McManus MT, Sunyaev S, Thomson JA, Tlsty TD, Tsai LH, Wang W, Waterland RA, Zhang MQ, Chadwick LH, Bernstein BE, Costello JF, Ecker JR, Hirst M, Meissner A, Milosavljevic A, Ren B, Stamatoyannopoulos JA, Wang T, Kellis M, Consortium RE, 2015. Integrative analysis of 111 reference human epigenomes. Nature 518, 317–330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lachner M, O’Sullivan RJ, Jenuwein T, 2003. An epigenetic road map for histone lysine methylation. J Cell Sci 116, 2117–2124. [DOI] [PubMed] [Google Scholar]
- Langie SAS, Moisse M, Declerck K, Koppen G, Godderis L, Vanden Berghe W, Drury S, De Boever P, 2017. Salivary DNA Methylation Profiling: Aspects to Consider for Biomarker Identification. Basic Clin Pharmacol Toxicol 121 Suppl 3, 93–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Le Francois B, Soo J, Millar AM, Daigle M, Le Guisquet AM, Leman S, Minier F, Belzung C, Albert PR, 2015. Chronic mild stress and antidepressant treatment alter 5-HT1A receptor expression by modifying DNA methylation of a conserved Sp4 site. Neurobiology of disease 82, 332–341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leuchter AF, Cook IA, Marangell LB, Gilmer WS, Burgoyne KS, Howland RH, Trivedi MH, Zisook S, Jain R, McCracken JT, Fava M, Iosifescu D, Greenwald S, 2009. 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 169, 124–131. [DOI] [PubMed] [Google Scholar]
- Li M, Zou D, Li Z, Gao R, Sang J, Zhang Y, Li R, Xia L, Zhang T, Niu G, Bao Y, Zhang Z, 2018. EWAS Atlas: a curated knowledgebase of epigenome-wide association studies. Nucleic Acids Res [DOI] [PMC free article] [PubMed]
- Lisoway AJ, Zai CC, Tiwari AK, Kennedy JL, 2018. DNA methylation and clinical response to antidepressant medication in major depressive disorder: A review and recommendations. Neurosci Lett 669, 14–23. [DOI] [PubMed] [Google Scholar]
- Logue MW, Smith AK, Wolf EJ, Maniates H, Stone A, Schichman SA, McGlinchey RE, Milberg W, Miller MW, 2017. The correlation of methylation levels measured using Illumina 450K and EPIC BeadChips in blood samples. Epigenomics 9, 1363–1371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lopez JP, Mamdani F, Labonte B, Beaulieu MM, Yang JP, Berlim MT, Ernst C, Turecki G, 2013. Epigenetic regulation of BDNF expression according to antidepressant response. Mol Psychiatry 18, 398–399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lowe R, Gemma C, Beyan H, Hawa MI, Bazeos A, Leslie RD, Montpetit A, Rakyan VK, Ramagopalan SV, 2013. Buccals are likely to be a more informative surrogate tissue than blood for epigenome-wide association studies. Epigenetics : official journal of the DNA Methylation Society 8, 445–454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Malhi GS, Outhred T, Hamilton A, Boyce PM, Bryant R, Fitzgerald PB, Lyndon B, Mulder R, Murray G, Porter RJ, Singh AB, Fritz K, 2018. Royal Australian and New Zealand College of Psychiatrists clinical practice guidelines for mood disorders: major depression summary. Med J Aust 208, 175–180. [DOI] [PubMed] [Google Scholar]
- Maunakea AK, Nagarajan RP, Bilenky M, Ballinger TJ, D’Souza C, Fouse SD, Johnson BE, Hong C, Nielsen C, Zhao Y, Turecki G, Delaney A, Varhol R, Thiessen N, Shchors K, Heine VM, Rowitch DH, Xing X, Fiore C, Schillebeeckx M, Jones SJ, Haussler D, Marra MA, Hirst M, Wang T, Costello JF, 2010. Conserved role of intragenic DNA methylation in regulating alternative promoters. Nature 466, 253–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Melas PA, Rogdaki M, Lennartsson A, Björk K, Qi H, Witasp A, Werme M, Wegener G, Mathé AA, Svenningsson P, Lavebratt C, 2012. Antidepressant treatment is associated with epigenetic alterations in the promoter of P11 in a genetic model of depression. Int J Neuropsychopharmacol 15, 669–679. [DOI] [PubMed] [Google Scholar]
- Menke A, Binder EB, 2014. Epigenetic alterations in depression and antidepressant treatment. Dialogues Clin Neurosci 16, 395–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meylan EM, Halfon O, Magistretti PJ, Cardinaux JR, 2016. The HDAC inhibitor SAHA improves depressive-like behavior of CRTC1-deficient mice: Possible relevance for treatment-resistant depression. Neuropharmacology 107, 111–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Molendijk ML, Bus BA, Spinhoven P, Penninx BW, Kenis G, Prickaerts J, Voshaar RC, Elzinga BM, 2011. Serum levels of brain-derived neurotrophic factor in major depressive disorder: state-trait issues, clinical features and pharmacological treatment. Mol Psychiatry 16, 1088–1095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nasca C, Xenos D, Barone Y, Caruso A, Scaccianoce S, Matrisciano F, Battaglia G, Mathe AA, Pittaluga A, Lionetto L, Simmaco M, Nicoletti F, 2013. L-acetylcarnitine causes rapid antidepressant effects through the epigenetic induction of mGlu2 receptors. Proceedings of the National Academy of Sciences of the United States of America 110, 4804–4809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nghia NA, Hirasawa T, Kasai H, Obata C, Moriishi K, Mochizuki K, Koizumi S, Kubota T, 2015. Long-term imipramine treatment increases N-methyl-d-aspartate receptor activity and expression via epigenetic mechanisms. Eur J Pharmacol 752, 69–77. [DOI] [PubMed] [Google Scholar]
- Oh G, Wang SC, Pal M, Chen ZF, Khare T, Tochigi M, Ng C, Yang YA, Kwan A, Kaminsky ZA, Mill J, Gunasinghe C, Tackett JL, Gottesman II, Willemsen G, de Geus EJC, Vink JM, Slagboom PE, Wray NR, Heath AC, Montgomery GW, Turecki G, Martin NG, Boomsma DI, McGuffin P, Kustra R, Petronis A, 2015. DNA modification study of major depressive disorder: beyond locus-by-locus comparisons. Biol Psychiatry 77, 246–255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Okada S, Morinobu S, Fuchikami M, Segawa M, Yokomaku K, Kataoka T, Okamoto Y, Yamawaki S, Inoue T, Kusumi I, Koyama T, Tsuchiyama K, Terao T, Kokubo Y, Mimura M, 2014. The potential of SLC6A4 gene methylation analysis for the diagnosis and treatment of major depression. Journal of psychiatric research 53, 47–53. [DOI] [PubMed] [Google Scholar]
- Olbrich S, Arns M, 2013. EEG biomarkers in major depressive disorder: discriminative power and prediction of treatment response. Int Rev Psychiatry 25, 604–618. [DOI] [PubMed] [Google Scholar]
- Papakostas GI, 2008. Tolerability of modern antidepressants. J Clin Psychiatry 69 Suppl E1, 8–13. [PubMed] [Google Scholar]
- Papakostas GI, Fava M, 2009. Does the probability of receiving placebo influence clinical trial outcome? A meta-regression of double-blind, randomized clinical trials in MDD. Eur Neuropsychopharmacol 19, 34–40. [DOI] [PubMed] [Google Scholar]
- Perisic T, Zimmermann N, Kirmeier T, Asmus M, Tuorto F, Uhr M, Holsboer F, Rein T, Zschocke J, 2010. Valproate and amitriptyline exert common and divergent influences on global and gene promoter-specific chromatin modifications in rat primary astrocytes. Neuropsychopharmacology 35, 792–805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perry PJ, 1996. Pharmacotherapy for major depression with melancholic features: relative efficacy of tricyclic versus selective serotonin reuptake inhibitor antidepressants. J Affect Disord 39, 1–6. [DOI] [PubMed] [Google Scholar]
- Peterson K, Dieperink E, Anderson J, Boundy E, Ferguson L, Helfand M, 2017. Rapid evidence review of the comparative effectiveness, harms, and cost-effectiveness of pharmacogenomics-guided antidepressant treatment versus usual care for major depressive disorder. Psychopharmacology (Berl) 234, 1649–1661. [DOI] [PubMed] [Google Scholar]
- Powell TR, Smith RG, Hackinger S, Schalkwyk LC, Uher R, McGuffin P, Mill J, Tansey KE, 2013. DNA methylation in interleukin-11 predicts clinical response to antidepressants in GENDEP. Translational psychiatry 3, e300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qiao M, Jiang QS, Liu YJ, Hu XY, Wang LJ, Zhou QX, Qiu HM, 2019. Antidepressant mechanisms of venlafaxine involving increasing histone acetylation and modulating tyrosine hydroxylase and tryptophan hydroxylase expression in hippocampus of depressive rats. Neuroreport [DOI] [PubMed]
- Quitkin FM, Harrison W, Stewart JW, McGrath PJ, Tricamo E, Ocepek-Welikson K, Rabkin JG, Wager SG, Nunes E, Klein DF, 1991. Response to phenelzine and imipramine in placebo nonresponders with atypical depression. A new application of the crossover design. Arch Gen Psychiatry 48, 319–323. [DOI] [PubMed] [Google Scholar]
- Quitkin FM, McGrath PJ, Stewart JW, Harrison W, Tricamo E, Wager SG, Ocepek-Welikson K, Nunes E, Rabkin JG, Klein DF, 1990. Atypical depression, panic attacks, and response to imipramine and phenelzine. A replication. Arch Gen Psychiatry 47, 935–941. [DOI] [PubMed] [Google Scholar]
- Roberts S, Lester KJ, Hudson JL, Rapee RM, Creswell C, Cooper PJ, Thirlwall KJ, Coleman JR, Breen G, Wong CC, Eley TC, 2014. Serotonin transporter [corrected] methylation and response to cognitive behaviour therapy in children with anxiety disorders. Transl Psychiatry 4, e444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rothschild AJ, Dunlop BW, Dunner DL, Friedman ES, Gelenberg A, Holland P, Kocsis JH, Kornstein SG, Shelton R, Trivedi MH, Zajecka JM, Goldstein C, Thase ME, Pedersen R, Keller MB, 2009. Assessing rates and predictors of tachyphylaxis during the prevention of recurrent episodes of depression with venlafaxine ER for two years (PREVENT) study. Psychopharmacol Bull 42, 5–20. [PMC free article] [PubMed] [Google Scholar]
- Rush AJ, Trivedi MH, Wisniewski SR, Nierenberg AA, Stewart JW, Warden D, Niederehe G, Thase ME, Lavori PW, Lebowitz BD, McGrath PJ, Rosenbaum JF, Sackeim HA, Kupfer DJ, Luther J, Fava M, 2006. Acute and longer-term outcomes in depressed outpatients requiring one or several treatment steps: a STAR*D report. Am J Psychiatry 163, 1905–1917. [DOI] [PubMed] [Google Scholar]
- Sales AJ, Biojone C, Terceti MS, Guimaraes FS, Gomes MV, Joca SR, 2011. Antidepressant-like effect induced by systemic and intra-hippocampal administration of DNA methylation inhibitors. British journal of pharmacology 164, 1711–1721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sales AJ, Joca SR, 2016. Effects of DNA methylation inhibitors and conventional antidepressants on mice behaviour and brain DNA methylation levels. Acta neuropsychiatrica 28, 11–22. [DOI] [PubMed] [Google Scholar]
- Sales AJ, Joca SRL, 2018. Antidepressant administration modulates stress-induced DNA methylation and DNA methyltransferase expression in rat prefrontal cortex and hippocampus. Behavioural brain research 343, 8–15. [DOI] [PubMed] [Google Scholar]
- Schmauss C, 2015. An HDAC-dependent epigenetic mechanism that enhances the efficacy of the antidepressant drug fluoxetine. Scientific reports 5, 8171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith AK, Kilaru V, Klengel T, Mercer KB, Bradley B, Conneely KN, Ressler KJ, Binder EB, 2015. DNA extracted from saliva for methylation studies of psychiatric traits: evidence tissue specificity and relatedness to brain. American journal of medical genetics. Part B, Neuropsychiatric genetics : the official publication of the International Society of Psychiatric Genetics 168B, 36–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun H, Kennedy PJ, Nestler EJ, 2013. Epigenetics of the depressed brain: role of histone acetylation and methylation. Neuropsychopharmacology 38, 124–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tadic A, Muller-Engling L, Schlicht KF, Kotsiari A, Dreimuller N, Kleimann A, Bleich S, Lieb K, Frieling H, 2014. Methylation of the promoter of brain-derived neurotrophic factor exon IV and antidepressant response in major depression. Molecular psychiatry 19, 281–283. [DOI] [PubMed] [Google Scholar]
- Tadić A, Wagner S, Schlicht KF, Peetz D, Borysenko L, Dreimüller N, Hiemke C, Lieb K, 2011. The early non-increase of serum BDNF predicts failure of antidepressant treatment in patients with major depression: a pilot study. Prog Neuropsychopharmacol Biol Psychiatry 35, 415–420. [DOI] [PubMed] [Google Scholar]
- Takeuchi N, Nonen S, Kato M, Wakeno M, Takekita Y, Kinoshita T, Kugawa F, 2017. Therapeutic Response to Paroxetine in Major Depressive Disorder Predicted by DNA Methylation. Neuropsychobiology 75, 81–88. [DOI] [PubMed] [Google Scholar]
- Tansey KE, Guipponi M, Hu X, Domenici E, Lewis G, Malafosse A, Wendland JR, Lewis CM, McGuffin P, Uher R, 2013. Contribution of common genetic variants to antidepressant response. Biol Psychiatry 73, 679–682. [DOI] [PubMed] [Google Scholar]
- Tsankova NM, Berton O, Renthal W, Kumar A, Neve RL, Nestler EJ, 2006. Sustained hippocampal chromatin regulation in a mouse model of depression and antidepressant action. Nature neuroscience 9, 519–525. [DOI] [PubMed] [Google Scholar]
- Turecki G, Meaney MJ, 2016. Effects of the Social Environment and Stress on Glucocorticoid Receptor Gene Methylation: A Systematic Review. Biol Psychiatry 79, 87–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang P, Lv Q, Mao Y, Zhang C, Bao C, Sun H, Chen H, Yi Z, Cai W, Fang Y, 2018a. HTR1A/1B DNA methylation may predict escitalopram treatment response in depressed Chinese Han patients. J Affect Disord 228, 222–228. [DOI] [PubMed] [Google Scholar]
- Wang P, Zhang C, Lv Q, Bao C, Sun H, Ma G, Fang Y, Yi Z, Cai W, 2018b. Association of DNA methylation in BDNF with escitalopram treatment response in depressed Chinese Han patients. Eur J Clin Pharmacol 74, 1011–1020. [DOI] [PubMed] [Google Scholar]
- Wang T, Choi E, Monaco MC, Major EO, Medynets M, Nath A, 2015. Direct induction of human neural stem cells from peripheral blood hematopoietic progenitor cells. J Vis Exp, 52298. [DOI] [PMC free article] [PubMed]
- Wei Y, Melas PA, Wegener G, Mathe AA, Lavebratt C, 2014. Antidepressant-like effect of sodium butyrate is associated with an increase in TET1 and in 5-hydroxymethylation levels in the Bdnf gene. The international journal of neuropsychopharmacology 18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Whirl-Carrillo M, McDonagh EM, Hebert JM, Gong L, Sangkuhl K, Thorn CF, Altman RB, Klein TE, 2012. Pharmacogenomics knowledge for personalized medicine. Clin Pharmacol Ther 92, 414–417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Williams LM, 2017. Defining biotypes for depression and anxiety based on large-scale circuit dysfunction: a theoretical review of the evidence and future directions for clinical translation. Depress Anxiety 34, 9–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu H, Wang J, Zhang K, Zhao M, Ellenbroek B, Shao F, Wang W, 2018. Effects of adolescent social stress and antidepressant treatment on cognitive inflexibility and Bdnf epigenetic modifications in the mPFC of adult mice. Psychoneuroendocrinology 88, 92–101. [DOI] [PubMed] [Google Scholar]
- Yardimci H, Zhang Y, 2015. Charting oxidized methylcytosines at base resolution. Nat Struct Mol Biol 22, 656–661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ye D, Zhang L, Fan W, Zhang X, Dong E, 2018. Genipin normalizes depression-like behavior induced by prenatal stress through inhibiting DNMT1. Epigenetics, 1–22. [DOI] [PMC free article] [PubMed]
- Zeier Z, Carpenter LL, Kalin NH, Rodriguez CI, McDonald WM, Widge AS, Nemeroff CB, 2018. Clinical Implementation of Pharmacogenetic Decision Support Tools for Antidepressant Drug Prescribing. Am J Psychiatry, appiajp201817111282. [DOI] [PMC free article] [PubMed]
- Zhu H, Bogdanov MB, Boyle SH, Matson W, Sharma S, Matson S, Churchill E, Fiehn O, Rush JA, Krishnan RR, Pickering E, Delnomdedieu M, Kaddurah-Daouk R, Network PR, 2013. Pharmacometabolomics of response to sertraline and to placebo in major depressive disorder - possible role for methoxyindole pathway. PLoS One 8, e68283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ziller MJ, Gu H, Müller F, Donaghey J, Tsai LT, Kohlbacher O, De Jager PL, Rosen ED, Bennett DA, Bernstein BE, Gnirke A, Meissner A, 2013. Charting a dynamic DNA methylation landscape of the human genome. Nature 500, 477–481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zimmermann N, Zschocke J, Perisic T, Yu S, Holsboer F, Rein T, 2012. Antidepressants inhibit DNA methyltransferase 1 through reducing G9a levels. Biochem J 448, 93–102. [DOI] [PubMed] [Google Scholar]