No evidence for intervention-associated DNA methylation changes in monocytes of patients with posttraumatic stress disorder or anorexia nervosa =============================================================================================================================================== * Elisabeth Hummel * Magdeldin Elgizouli * Jasmin Beygo * Johanna Giuranna * Maurizio Sicorello * Elsa Leitão * Christopher Schröder * Michael Zeschnigk * Svenja Müller * Dana Öztürk * Manuel Föcker * Stephan Herpertz * Johannes Hebebrand * Dirk Moser * Henrik Kessler * Bernhard Horsthemke * Anke Hinney * Robert Kumsta ## Abstract **Background** DNA methylation patterns can be responsive to environmental influences. This observation has sparked interest in the potential for psychological interventions to influence epigenetic processes. Recent studies have observed correlations between DNA methylation changes and therapy outcome. However, most did not control for changes in cell composition from pre- to post-therapy. Here, we addressed this limitation and analyzed DNA methylation patterns by targeted deep bisulfite sequencing (DBS) of commonly assessed candidate genes in isolated monocytes from 60 female patients with post-traumatic stress disorder (PTSD) before and after in-patient treatment. Furthermore, in two patients with PTSD and three patients with anorexia nervosa (AN), whole-genome bisulfite sequencing (WGBS) was performed before and after intervention with the aim of identifying novel genomic regions with notable pre-to-post intervention DNA methylation changes. **Results** WGBS yielded only a limited set of candidate regions with suggestive evidence of differential methylation pre- to post-therapy. These differential methylation patterns did not prove replicable when investigated in the entire cohort. Furthermore, equivalence testing and Bayesian analyses provided evidence against physiologically meaningful intervention associated DNA methylation changes in monocytes of PTSD patients in commonly investigated target genes (*NR3C1, FKBP5, SLC6A4, OXTR)*. **Conclusions** We conclude that there is no evidence for major, recurrent intervention-associated DNA methylation changes in the investigated genes in monocytes of patients with either PTSD or anorexia nervosa. Keywords * DNA methylation * whole-genome bisulfite sequencing * posttraumatic stress disorder * anorexia nervosa * monocytes * psychotherapy ## 1. Background The concept of an environmentally regulated epigenome has attracted considerable attention in the mental health field [1, 2]. Many psychopathologies have developmental origins, and epigenetic processes have been suggested as targets for the effects of psychosocial adversity particularly in early life. The epigenome is an umbrella term for a range of mechanisms involved in gene regulation, including DNA methylation, histone modifications and regulation through non-coding RNA molecules [3]. Epigenetic processes are essential for normal cellular differentiation and development. Perturbations in these processes have been linked to different pathologies [4], including mental disorders [5]. Both animal models and human studies have pointed to persistent epigenetic changes in response to various stimuli. These changes, it has been argued, might reflect or mediate the long-term effects of early risk exposure such as prenatal malnutrition on metabolic outcomes [6]. Another often quoted example of epigenetic mediation is the programming of the neuroendocrine stress response by the degree of maternal care in rodents [7]. DNA methylation, which involves methylation of cytosines predominantly in cytosine-guanine (CpG) dinucleotides, has long been thought to represent an early-established and rather stable epigenetic marker. A number of recent studies have shown, however, that DNA methylation patterns are more dynamic than previously thought, and can change in response to internal signals or environmental influences [8-12]. These observations have sparked interest in the potential for psychological and nutritional interventions (e.g. in Anorexia Nervosa) to influence these biological processes. Several studies have been published that assessed DNA methylation before and after therapeutic intervention [see 13 for review]. Most followed a candidate-gene approach and investigated genes involved in stress regulation, such as the Glucocorticoid Receptor (*NR3C1*) or FKBP Prolyl Isomerase 5 (*FKBP5*) [14-16], or genes commonly investigated in psychiatric genetics studies, such as the Serotonin Transporter (*SLC6A4*; [17]), Monoamine Oxidase A (*MAOA*; [18]), and the Brain Derived Neurotrophic Factor (*BDNF*; [19]). The studies conducted so far are very heterogeneous in terms of investigated disorders (post-traumatic stress disorder, PTSD; anorexia nervosa, AN; anxiety disorders, borderline personality disorder), analyzed tissue (buccal cells, whole blood, peripheral blood mononuclear cells (PBMCs), type of intervention and methodology for exploring DNA methylation. A common feature of all investigations is the observation that therapy responders and non-responders showed differential DNA methylation patterns pre- to post-intervention. For instance, in combat veterans with PTSD, DNA methylation levels of *FKBP5* in PBMC DNA decreased in responders but increased in non responders comparing pre- to post-therapy measurements [16]. The first epigenome-wide investigation of intervention-associated DNA methylation changes in PTSD further supported this notion. Successful psychotherapeutic treatment of PTSD was associated with increases in DNA methylation of the Zinc Finger Protein 57 gene (*ZFP57*) in whole blood, whereas DNA methylation in this region decreased during the development of PTSD, and also decreased in patients who did not respond to therapy [20]. A similar picture emerged from an epigenome-wide study of anorexia nervosa. Here, longer duration of illness was associated with lower DNA methylation levels in whole blood, whereas DNA methylation increased at the same positions with a concomitant increase in BMI across therapy [21]. Taken together, DNA methylation changes have thus been suggested as a marker or epigenetic correlate of therapy outcome. Caution is needed in the interpretation of these findings though. With exception of the two epigenome-wide association studies, only a limited number of CpGs were investigated, the observed changes were small, and the applied methods mostly had insufficient sensitivity to reliably assess such small DNA methylation differences. Furthermore, it is unclear whether differences in DNA methylation, as observed after the intervention, do not rather reflect changes in the cellular composition of the investigated tissue. DNA methylation patterns are highly cell type-specific, and each cell type within a tissue contributes to DNA methylation variation [22]. Given that the cell composition of the circulating leukocyte pool is dynamic and influenced by various external factors, including infections or stress exposure [23], DNA methylation changes in blood collected at different time points from the same individual might primarily reflect differences in cell composition [24], unrelated to the effect of intervention. To address these limitations, we set out to investigate intervention-associated changes in DNA methylation in a homogenous cell population (CD14+ monocytes), in two independent female-only patient cohorts (PTSD, AN). Monocytes were chosen as previous studies have shown that among the heterogeneous leukocyte population, monocytes were the most sensitive subtype for traumatic experiences and variation of psychosocial conditions [25]. For DNA methylation analysis we used whole-genome bisulfite sequencing (WGBS) in a subsample to identify candidate regions, followed by targeted deep bisulfite sequencing (DBS) of promising targets and commonly assessed candidate genes. Our approach thus addressed essential confounders, namely sex, cell heterogeneity, genetic variation, coverage of the genome and coverage of CpGs within candidate genes. ## 2. Materials and Methods ### 2.1 Study design and sample characteristics The overall study design is shown in Figure 1. Sixty female in-patients of European descent seeking treatment for PTSD at the Department of Psychosomatic Medicine and Psychotherapy, LWL-University Hospital, Ruhr-University Bochum participated in the study. The patients were between 20 and 60 years old (mean age: 40.0 ± 11.9 (SD) years), and the mean treatment duration was 6.5 ± 1.4 (SD) weeks. Inclusion criteria were PTSD diagnosis and female sex. The second cohort comprised seven female adolescent patients with AN, receiving in-patients treatment at the University Hospital Essen, University of Duisburg-Essen. The median age was 16 (with first and third quartile: Q1, Q3; 15, 17) years, median height 162.5 (160.2, 167.7) cm, median weight at admission 40.2 (37.6, 46.7) kg, median BMI at admission 15.2 (14.8, 16.1) kg/m2. Length of treatment was 94 (69, 99) days. The median age after treatment was 16.4 (with first and third quartile: Q1, Q3; 15.6, 17.3) years, median height after treatment 162.5 (160.2, 167.7) cm, median weight after treatment 46.0 (44.0, 52.6) kg, median BMI after treatment 17.5 (17.5, 17.8) kg/m2. Inclusion criteria for the AN group were a diagnosis of AN, female sex, German ancestry and age from 12 to 18 years. More information about the cohorts is described in Supplementary Table 1, Additional File 1. ![Figure 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/11/13/2020.11.11.20229567/F1.medium.gif) [Figure 1:](http://medrxiv.org/content/early/2020/11/13/2020.11.11.20229567/F1) Figure 1: Overview of the study design. ### 2.2 Clinical judgement and symptom self-report All patients with PTSD were diagnosed with ICD-10 (F43.1; International Classification of Diseases, 10th revision, WHO, 1993) prior to in-patient admission via structured clinical interviews in the outpatient department of the hospital. Furthermore, symptoms of PTSD were recorded before and after in-patient treatment with the PTSD-Check List [PCL-5; 26]. Patients with AN were examined at the Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen according to Diagnostic and Statistical Manual of Mental Disorders, fourth edition, text revision (DSM-IV-TR) criteria [27]. AN diagnosis was confirmed via clinical examination and a semi structured interview (Kiddie Schedule for Affective Disorders and Schizophrenia (K-SADS [28]). ### 2.3 Treatment Participants of the PTSD cohort received standard in-patient PTSD treatment. This involved one session each week of individual cognitive-behavioral therapy, three sessions of trauma group therapy, two sessions of trauma stabilization group therapy, one session of a “skills group”, two sessions of kinesitherapy, two sessions of art therapy, physiotherapy, clinical rounds, and daily short sessions with a nurse. During individual therapy sessions patients received different trauma exposure methods. Adolescent patients with AN participated in a multimodal multidisciplinary treatment program based on dialectical behavioral therapy for adolescents that included weight restoration, individual nutritional counseling, individual therapy (twice a week), skills-groups and individual skill training (twice a week), family sessions (fortnightly), and a group psychoeducation program for parents (monthly). During weight restoration, patients received nutritional counselling and meal support training. Therapy goals were a weight gain of at least 500g per week and a minimum target weight in the range of the 10-20th age-adjusted BMI percentile. ### 2.4 Monocyte and DNA Isolation From PTSD and AN individuals 9 ml blood was drawn in the morning between 7-9 am (S-Monovette 9 ml K3E, Sarstedt). Monocytes were immunomagnetically purified from whole blood with the MACS System (Miltenyi Biotec, Bergisch Gladbach, Germany), shock frozen and stored at -80°C. Cell homogeneity is described in Supplementary Methods 1, Additional File 1. DNA from the PTSD cohort was isolated with the AllPrep RNA/DNA Mini Kit (Qiagen, Hilden, Germany). AN DNA was extracted with a standard salting-out procedure. DNA was stored at -80°C until further analysis. ### 2.5 Whole-genome bisulfite sequencing Pre- and post-intervention monocyte DNA from five patients (two PTSD, three AN) was subjected to WGBS. The patients for the WGBS were selected based on their responsiveness to the therapy, absence of co-morbidity, and non-smoking status (Supplementary Table 2, Additional File 1). For PTSD samples, WGBS libraries were prepared as previously described [29]. For the AN samples, 5 µg DNA at 50 ng/µl in water was used for library preparation and NGS sequencing on a HiSeq X platform (DKFZ Genomics & Proteomics Core Facility (GPCF) with 1 sample per lane. Read data from both datasets were processed as described previously [30, 31]. As a reference genome, we used hg19. For calling differentially methylation regions (DMR), we used camel [32] and metilene [33]. ### 2.7 Targeted bisulfite sequencing For DBS in both cohorts, DNA was bisulfite-modified with the EZ DNA Methylation Gold Kit (Zymo Research, Orange, CA, USA) and processed as described elsewhere [34, 35] using primers as indicated in Supplementary Table 3, Additional File 1. ### 2.8 Statistical Analyses Comparison of whole genome mean methylation values was performed by Mann-Whitney U test (PTSD versus AN samples) or Wilcoxon signed rank test (pre-versus post-intervention samples) using the R *stats* package (4.0.0). In the PTSD cohort, repeated measures analyses of variance (ANOVA) were performed to assess changes in DNA methylation. Therapy response was included as an additional between-subject factor to check for therapy outcome-dependent changes in DNA methylation. Moreover, Pearson correlations were computed between percent DNA methylation change and PCL-5 symptom change to test a continuous measure of therapy response. In addition to changes over time, linear regression analysis was performed with pre-treatment mean DNA methylation of DBS targets and severity of baseline PTSD symptoms. Generally, non-significant results do not provide evidence for the null hypothesis (i.e. absence of effect), especially when the statistical power is limited. Therefore, p-values derived from both approaches, categorical and continuous, were supplemented with Bayes factors to quantify the evidence for the null hypothesis using the BayesFactor package (v0.9.12-4.2) in R (3.6.1) and non-informative default priors [36, 37]. Complementary to the Bayesian approach, we conducted equivalence tests to assess whether effect sizes in DNA methylation change are significantly smaller than the smallest biologically meaningful effect size. It has been argued that DNA methylation below 5% should be interpreted with extreme caution [38, 39]. We used the two one-tailed t-test procedure [40] to check whether empirical effect sizes for methylation change are smaller than 5% or even a more conservative 1%. For a detailed account of statistical methods please see Supplementary Methods 2, Additional File 1. ## 3. Results ### 3.1 Clinical outcome Patients with PTSD had a clinically relevant reduction in PTSD symptoms, with a mean reduction of 15.6 ± 14.6 (SD) on the PCL-5. Thirty-six patients were classified as responder according to PCL-5 (drop by 10 points from pre- to post-treatment). The responders showed a mean difference between post- and pre-treatment in PCL-5 of -24.1 ± 10.3, while the non-responders (n=21, 35%) showed a mean difference of -1.0 ± 7.8 (PCL-5 data was missing for three patients). Patients with anorexia nervosa had a median BMI at admission of 15.2 (14.8, 16.1) kg/m2 and median BMI at end of treatment was 17.5 (17.5, 17.8) kg/m2. During the treatment period (i.e. hospital stay) of a median of 94 (69, 99) days, all seven patients gained considerable weight (median weight gain 5.8 (5.6, 6.7) kg; median BMI gain 2.2 (2.0, 2.4) kg/m2). ### 3.2 Explorative whole genome bisulfite sequencing Following recommendations by Ziller et al. [41], we sequenced at least two biological replicates at >5x coverage. We chose two PTSD and three AN patients. In each case, WGBS-libraries from monocytes isolated before and after therapeutic intervention were sequenced (Supplementary Table 4, Additional File 1). There was a slight difference in the genome-wide mean methylation levels between the PTSD and AN samples (0.72 vs. 0.75). DNA methylation did not differ between pre- and post-intervention samples of the same individuals (p-value = 1). A principal component analysis (PCA) of the PTSD and AN methylomes did not group samples according to pre- or post-intervention state (Supplementary Figure 1, Additional File 1). The same was observed in a cluster analysis of the 1,000 most variable CpGs for the PTSD and AN datasets, in which separate branches are observed for each individual (Figure 2). We observed large differences between individuals, most likely reflecting the different genetic backgrounds, but no recurrent differences between the pre- and post-intervention states. ![Figure 2:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/11/13/2020.11.11.20229567/F2.medium.gif) [Figure 2:](http://medrxiv.org/content/early/2020/11/13/2020.11.11.20229567/F2) Figure 2: Cluster analysis of 1,000 most variable CpGs. A) The PTSD monocyte methylomes were obtained from two individuals (47 and 43), pre-(.1) and post-intervention (.2). B) The AN monocyte methylomes were obtained pre- and post-treatment from three individuals (AN1, AN2 and AN3). CpG SNPs were excluded from the analysis. The difference between individuals is much greater than between pre- and post-intervention. To identify DMRs between pre- and post-treatment samples from each cohort, we used two different bioinformatic tools (camel and metilene). Owing to lower coverage in the PTSD samples compared to the AN samples, we set the threshold to a minimum coverage of five reads and ten reads, respectively. Defining a camel DMR as a region with at least four CpGs and a minimum methylation difference of 0.3, we detected 33 DMRs in the PTSD patients (Supplementary Table 5, Additional File 1). Using a filter of q < 0.05 for metilene DMRs, we detected four DMRs (Supplementary Table 6, Additional File 1). Using the same DMR filters, camel identified no DMRs in the AN data set, indicating that there are no major recurrent methylation changes. Since the samples of this cohort were sequenced at a higher depth, we decided to lower the minimum methylation difference between groups to 0.2 while at the same time increasing stringency by setting the minimum number of CpGs to 10. Using these filters, we identified 35 DMRs with camel (Supplementary Table 7, Additional File 1). At q<0.05, metilene reported 10 DMRs, with seven DMRs being detected by both tools (Supplementary Table 8, Additional File 1). There is one DMR common to the PTSD and AN datasets, which overlaps the binding sites for CTCF and other transcription factors within intron 1 of *EXD3*, a gene encoding an exoribonuclease required for 3’-end trimming of AGO1-bound miRNAs. ### 3.3 Candidate genes and WGBS targets In the entire PTSD cohort, DBS was performed for validation of four targets that emerged as potentially responsive to intervention from WGBS data: DMR-4 within Adenosine A1 Receptor (*ADORA1)*, DMR-21 within Tetraspanin 9 *(TSPAN9)*, DMR-10 within Ribosomal Protein S6 Kinase A2 *(RPS6KA2)*, and intergenic DMR-1 (Supplementary Table 4, Additional File 1). Furthermore, four candidate genes that have previously been investigated in therapy epigenetic studies were analyzed: *NR3C1, FKBP5, SLC6A4* and Oxytocin Receptor (*OXTR;* see Supplementary Figure 2, Additional File 1 for exact chromosomal locations). Linear regression analysis showed no significant association between pre-treatment mean DNA methylation of DBS targets and the severity of baseline PTSD symptoms. Likewise there were no statistically significant differences in DNA methylation between pre- and post-intervention for all investigated targets, even without correction for multiple comparisons. Bayes factors favored the null hypothesis of no methylation change for *NR3C1, FKBP5, RPS6KA2* and DMR-1. Furthermore, DNA methylation change was not conditional on therapy response (all p > 0.05; Table 1), with all Bayes factors larger than one and Bayes factors for *SLC6A4, FKBP5* and DMR-1 above the threshold of three. Moreover, we observed no statistically significant correlation between symptom changes and DNA methylation changes (all p > 0.05; Table 2 and Supplementary Figure 3, Additional File 1), with all Bayes factors larger than one and Bayes factors for *SLC6A4* and *FKBP5* above three. Distributions of DNA methylation changes are shown in Figure 3 by responder status. The largest mean differences between time points was found for *TSPAN9* (−2.78% ± 9.78%), and the smallest was -0.019% ± 0.19% for *NR3C1*. Mean DNA methylation levels pre- and post-intervention averaged across the analyzed genomic regions are shown in Supplementary Table 9, and Supplementary Figure 4, Additional File 1. ![Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2020/11/13/2020.11.11.20229567/F3.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2020/11/13/2020.11.11.20229567/F3) Figure 3. Density plots for the distribution of DNA methylation change (post minus pre-treatment) by therapy response and gene. View this table: [Table 1:](http://medrxiv.org/content/early/2020/11/13/2020.11.11.20229567/T1) Table 1: Effect of *intervention* and the effect of *intervention* by *responder* status interaction on DNA methylation in the PTSD cohort View this table: [Table 2:](http://medrxiv.org/content/early/2020/11/13/2020.11.11.20229567/T2) Table 2: Correlations between change in DNA methylation and change in symptom scores (PTSD cohort) In PTSD the four classical candidate genes (*NR3C1, SLC6A4, OXTR, FKBP5*) all had effect sizes significantly smaller than 1% DNA methylation change, even in the therapy-responder group and after Bonferroni correction (all *p**corrected* < .001). The effect sizes of *ADORA1, RPS6KA2*, and DMR-1 were all significantly smaller than 5% DNA methylation change in both the whole cohort and the responder cohort (all *p**corrected* < .001). The effect of *TSPAN9* was only significantly different from 5% in the whole cohort on an uncorrected *p* = .043, which did not survive Bonferroni correction. Hence, although we cannot rule out that there is a true effect size as large as 5% DNA methylation change for the *TSPAN9* locus, we can conclude with relative certainty that true effect sizes of all classic candidate genes are smaller than 1%, and true effect sizes of the WBGS nominated genes are smaller than 5%. In the AN cohort, DBS was performed for validation of targets that emerged from WGBS (methylation difference ≥ 0.2) in all three individuals as potentially responsive to weight gain in eight regions (four detected by both DMR-calling tools and four detected by camel: DMR-1 within Galactosidase Beta 1 Like (*GLB1L*), intergenic DMR-3, DMR-7 within Family With Sequence Similarity 50 Member B (*FAM50B*), DMR-11 within Mesoderm Specific Transcript (*MEST*), DMR-13 within ER Lipid Raft Associated 2 (*ERLIN2*), DMR-16 within Exonuclease 3’-5’ Domain Containing 3 (*EXD3*), DMR-22 within SNRPN Upstream Reading Frame/ Small Nuclear Ribonucleoprotein Polypeptide N (*SNURF*/*SNRPN*) and DMR-28 within Heat Shock Protein Family A (Hsp70) Member 12B (*HSPA12B*) (for DMR numbering see Supplementary Table 7, Additional File 1). All CpGs contained in the DMRs were included in the amplicons. Since for this cohort we had used a lower minimum methylation difference between groups (0.2), we first attempted technical confirmation using DBS in the same three individuals (pre- and post-treatment; same DNA preparation as for WGBS). As can be seen in Supplementary Figure 5, Additional File 1, the methylation differences identified by WBGS could not be confirmed in any of these regions. Therefore, the four independent samples were not analyzed. ## 4. Discussion Epigenetic processes have been proposed as a potential mechanism mediating the link between exposure to trauma or adversity and mental health problems. Expectedly, there is great interest in the question whether psychological interventions translate into a modification of disorder-related epigenetic signatures. The first few studies addressing this question have found divergent patterns of DNA methylation changes between patients who responded to therapy and those who did not. However, results of these pilot studies remain inconclusive, because differences in genetic background and changes in cell composition over time, which are the main confounders in epigenetic research, were not taken into account. In our study, we avoided these confounders by using isogenic pre- and post-treatment samples and a homogenous cell population (monocytes). Furthermore, we used DBS for targeted high resolution methylation analysis. With this approach, we could not identify any evidence of intervention-associated changes in DNA methylation. We investigated four candidate genes that were previously shown to be sensitive to variation in the social environment and/or seemed responsive to intervention. The prime candidate for epigenetic research in humans has been *NR3C1*, since DNA methylation of the promoter of alternative Exon 17 (1F in humans) had been shown to vary as a function of maternal care in the rodent model [7]. We found no change between pre- and post-intervention in PTSD patients and no differences between therapy responders and non-responders. For PTSD we also investigated DNA methylation of *SLC6A4*, the most widely studied candidate gene in psychiatry. One previous study found small increases in *SLC6A4* methylation in therapy responders in one of four assessed CpG sites [17]. Here, we investigated 82 CpGs sites across the entire CpG island in the gene’s promoter region and found no evidence for change over time - neither overall nor when stratified by responders and non-responders. This was also true for *FKBP5*, where we focused on 5 CpGs in intron 7, previously associated with PTSD risk following early life adversity [42], and the *OXTR* gene, where we investigated 16 CpGs in a putative enhancer region - characterized by H3K27 acetylation peaks - in intron 3 of the gene. In sum, we did not observe intervention-associated changes in DNA methylation of commonly investigated candidate genes when assessed in a homogenous cell population. In small samples with limited statistical power, non-significant results do not provide evidence for the absence of meaningful effects. We thus further conducted equivalence tests to ascertain whether observed effect sizes are significantly smaller than a minimal effect size of interest. These revealed that DNA methylation changes were significantly below physiologically meaningful levels for all genes. Only for *TSPAN9* the evidence was inconclusive (i.e. neither favoring the null nor the alternative hypothesis after Bonferroni correction). Hence, a true DNA methylation change of 5% cannot be ruled out for this particular gene. Candidate gene studies can only provide a very limited view on potential dynamics of the epigenome. True epigenome-wide studies covering the ∼28 million CpG sites across the human genome are still prohibitive in large cohorts because of the associated costs. Here, we used an explorative strategy to identify genomic regions whose DNA methylation pattern might potentially be responsive to intervention, by using WGBS in two PTSD and three AN patients before and after therapy. We identified a small number of pre-to-post intervention differentially methylated regions (36 for PTSD and 38 for AN). However, the validation of a subset of the identified PTSD DMRs in the remaining 58 patients could not confirm the potential differential methylation patterns found by WGBS. In addition, AN DMRs identified after reducing the stringency in DMR calling (by lowering to 0.2 the minimum methylation difference allowed) could not be validated by DBS in the three samples subjected to WGBS. It is remarkable that the change of physiological conditions for monocytes in AN due to adaption to prolonged starvation is not associated with significant changes in DNA methylation pattern. This suggests that cell type-specific methylation patterns are highly stable. This stability can probably be attributed to stem or progenitor cells, because monocytes have a very short half-life in blood. Pre- and post-treatment samples are not drawn from the same pool of monocytes, but are derived from the same stem or progenitor cells. We previously showed differences in metabolic profiles in patients with AN compared to controls and in patients with AN between acute stage of starvation and after short term weight recovery. A substantial number of metabolite levels did not return to normal after short term recovery [43]. It might be assumed that weight gain could exert an effect on the methylome at a later time point. It is important to reflect on the question whether it is plausible to expect changes of DNA methylation patterns in peripheral surrogate tissues that are associated with changes in thoughts, feelings or somatic alterations in a meaningful way. Three scenarios exist: first, DNA methylation dynamics observed in blood or buccal epithelial cells reflect the processes occurring in neuronal cells [18]. However, as buccal epithelium or leukocytes do not have a biologically realistic link to cellular processes occurring in neurons, these transient and activity-dependent alterations of chromatin and the specific patterns of DNA methylation in neurons are most likely not reflected in peripheral tissue. The second scenario is that changes in peripheral DNA methylation patterns are brought about by intervention-associated psychophysiological changes that parallel changes in thoughts and feelings. Such cross talk between the central nervous system and peripheral organs is arguably mediated through regulation of the autonomic nervous system and the hypothalamic–pituitary– adrenal axis, activating the two main stress effectors noradrenaline and cortisol. These mediate their effects by engaging cellular receptor systems, which ultimately regulate specific gene expression responses. A testable model therefore suggests that changes in DNA methylation are brought about by alterations of transcriptional activity associated with changes in upstream signaling of stress mediators such as cortisol or noradrenalin [44]. The third scenario, which is in our opinion the most plausible explanation for many previous findings, is that changes in DNA methylation levels merely reflect changes in cell composition, which might be stochastic in nature, reflect differences in health status independent of disorder of interest, or reflect intervention-associated physiological changes. As such, change in the composition of the leukocyte pool might be a marker of therapy response [45]. The following limitations deserve a mention. The costs associated with WGBS limited the number of patients to 2-3 for each cohort, so that only pre-to-post differences >30% methylation could be reliably identified. Moreover, the DMR calling true positive rate (TPR) and false discovery rat (FDR) are dependent on the number of replicates and the coverage. Given our cohort size and sequencing depth, we estimate the TPR to be >80% and the FDR to be <20% [41]. Furthermore, only one cell type was investigated. Monocytes have been shown as the most sensitive subtype for social conditions and traumatic experiences, at least in terms of transcriptional changes [25, 46-48]. However, any effects in other cell types (e.g., T cells, B cells, NK cells, etc.), or changes in the relative prevalence of cells in the circulating leukocyte pool are missed in this analysis. ## 5. Conclusion In conclusion, our results provide no evidence in support of intervention-associated changes in DNA methylation in monocytes in PTSD or AN, and equivalence tests and Bayesian statistics provided evidence against even subtle DNA methylation changes associated with therapy outcome. We argue that the previously reported changes in DNA methylation following therapy are most likely explained by changes in cell composition rather than by cellular reprogramming of DNA methylation. In itself, any shifts in cellular composition might reflect intervention-associated physiological changes and could therefore be used as biomarkers, but markers should not be confused with mechanisms. ## Supporting information Additional File 1 [[supplements/229567_file05.pdf]](pending:yes) ## Data Availability The WGBS datasets generated and analyzed during the current study are available in the European Nucleotide Archive (ENA) under the accession number PRJEB38906. The DBS datasets analyzed during the current study are available from the corresponding author on reasonable request. ## 7. Declarations ### Ethics approval and consent to participate The PTSD study was approved by the ethics committee of the Faculty of Psychology, Ruhr University Bochum (Nr. 155), and the AN study was approved by the ethics committee of the Faculty of Medicine, University Hospital Essen (Nr. 06-3212 amendment of November 2016). Patients and their parents (AN cohort only) gave written informed consent. ### Consent for publication All authors have their consent for publication. ### Availability of data and materials The WGBS datasets generated and analyzed during the current study are available in the European Nucleotide Archive (ENA) under the accession number PRJEB38906. The DBS datasets analyzed during the current study are available from the corresponding author on reasonable request. ## Competing interests All authors report no conflict of interest. ## Funding This work was supported by a project grant by the German Research Foundation (DFG) to RK (KU 2479/3-1, KU 2479/3-2). AH has been funded by the German Research Foundation (DFG; HI865/2-1), the Landesprogramm für Geschlechtergerechte Hochschulen – Programmstrang Förderung von Denominationen in der Genderforschung and the Medical Faculty of the University of Duisburg-Essen. ## Authors’ contributions RK, BH, AH, ME, HK, DM contributed to the study concept development; RK, BH, AH, SH, HK, ME contributed to project administration; SH, HK, AH, JG, DÖ, MF, JH contributed to the recruitment of the patients; EH, DM, SM, EL, JB were involved in method development, sample preparation and DNA sequencing; CS, EL, JB, MZ, MS, EH contributed to data preparation and statistical analysis; RK, BH, AH, ME, JB, EL, MS, EH, JH were involved in writing the manuscript. All authors read and approved the final manuscript. ## Acknowledgement We thank the Cologne Center for Genomics (CCG) and the DKFZ Genomics & Proteomics Core Facility (GPCF) for performing WGBS. ## 6. List of abbreviations AN : anorexia nervosa ANOVA : analyses of variance DBS : targeted deep bisulfite sequencing DMR : differentially methylation regions PBMCs : peripheral blood mononuclear cells PCA : principal component analysis PCL : PTSD-Check List PTSD : post-traumatic stress disorder WGBS : whole-genome bisulfite sequencing * Received November 11, 2020. * Revision received November 11, 2020. * Accepted November 13, 2020. * © 2020, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial-NoDerivs 4.0 International), CC BY-NC-ND 4.0, as described at [http://creativecommons.org/licenses/by-nc-nd/4.0/](http://creativecommons.org/licenses/by-nc-nd/4.0/) ## References 1. 1.Szyf M. The early life social environment and DNA methylation: DNA methylation mediating the long-term impact of social environments early in life. Epigenetics 2011; 6(8): 971–978. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.4161/epi.6.8.16793&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21772123&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F11%2F13%2F2020.11.11.20229567.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000293344500003&link_type=ISI) 2. 2.Hubel C, Marzi SJ, Breen G, Bulik CM. Epigenetics in eating disorders: a systematic review. Mol Psychiatry 2019; 24(6): 901–915. 3. 3.Allis CD, Jenuwein T. The molecular hallmarks of epigenetic control. Nat Rev Genet 2016; 17(8): 487–500. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nrg.2016.59&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27346641&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F11%2F13%2F2020.11.11.20229567.atom) 4. 4.Godfrey KM, Costello PM, Lillycrop KA. The developmental environment, epigenetic biomarkers and long-term health. J Dev Orig Health Dis 2015; 6(5): 399–406. 5. 5.McGowan PO, Szyf M. The epigenetics of social adversity in early life: implications for mental health outcomes. Neurobiol Dis 2010; 39(1): 66–72. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.nbd.2009.12.026&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20053376&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F11%2F13%2F2020.11.11.20229567.atom) 6. 6.Tobi EW, Slieker RC, Luijk R, Dekkers KF, Stein AD, Xu KM et al. DNA methylation as a mediator of the association between prenatal adversity and risk factors for metabolic disease in adulthood. Sci Adv 2018; 4(1): eaao4364. [FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6MzoiUERGIjtzOjExOiJqb3VybmFsQ29kZSI7czo4OiJhZHZhbmNlcyI7czo1OiJyZXNpZCI7czoxMjoiNC8xL2VhYW80MzY0IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMTEvMTMvMjAyMC4xMS4xMS4yMDIyOTU2Ny5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 7. 7.Weaver IC, Cervoni N, Champagne FA, D’Alessio AC, Sharma S, Seckl JR et al. Epigenetic programming by maternal behavior. Nat Neurosci 2004; 7(8): 847–854. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nn1276&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15220929&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F11%2F13%2F2020.11.11.20229567.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000222930800015&link_type=ISI) 8. 8.Dekkers KF, van Iterson M, Slieker RC, Moed MH, Bonder MJ, van Galen M et al. Blood lipids influence DNA methylation in circulating cells. Genome Biol 2016; 17(1): 138. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13059-016-1000-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F11%2F13%2F2020.11.11.20229567.atom) 9. 9.Emeny RT, Baumert J, Zannas AS, Kunze S, Wahl S, Iurato S et al. Anxiety Associated Increased CpG Methylation in the Promoter of Asb1: A Translational Approach Evidenced by Epidemiological and Clinical Studies and a Murine Model. Neuropsychopharmacology 2018; 43(2): 342–353. 10. 10.Joehanes R, Just AC, Marioni RE, Pilling LC, Reynolds LM, Mandaviya PR et al. Epigenetic Signatures of Cigarette Smoking. Circ Cardiovasc Genet 2016; 9(5): 436–447. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiY2lyY2N2ZyI7czo1OiJyZXNpZCI7czo3OiI5LzUvNDM2IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMTEvMTMvMjAyMC4xMS4xMS4yMDIyOTU2Ny5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 11. 11.Saunderson EA, Spiers H, Mifsud KR, Gutierrez-Mecinas M, Trollope AF, Shaikh A et al. Stress-induced gene expression and behavior are controlled by DNA methylation and methyl donor availability in the dentate gyrus. Proc Natl Acad Sci U S A 2016; 113(17): 4830–4835. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMToiMTEzLzE3LzQ4MzAiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMC8xMS8xMy8yMDIwLjExLjExLjIwMjI5NTY3LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 12. 12.Wong CC, Caspi A, Williams B, Craig IW, Houts R, Ambler A et al. A longitudinal study of epigenetic variation in twins. Epigenetics 2010; 5(6): 516–526. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.4161/epi.5.6.12226&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20505345&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F11%2F13%2F2020.11.11.20229567.atom) 13. 13.Kumsta R. The role of epigenetics for understanding mental health difficulties and its implications for psychotherapy research. Psychol Psychother 2019; 92(2): 190–207. 14. 14.Roberts S, Keers R, Lester KJ, Coleman JR, Breen G, Arendt K et al. Hpa Axis Related Genes and Response to Psychological Therapies: Genetics and Epigenetics. Depress Anxiety 2015; 32(12): 861–870. 15. 15.Roberts S, Keers R, Breen G, Coleman JRI, Johren P, Kepa A et al. DNA methylation of FKBP5 and response to exposure-based psychological therapy. Am J Med Genet B Neuropsychiatr Genet 2019; 180(2): 150–158. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F11%2F13%2F2020.11.11.20229567.atom) 16. 16.Yehuda R, Daskalakis NP, Desarnaud F, Makotkine I, Lehrner AL, Koch E et al. Epigenetic Biomarkers as Predictors and Correlates of Symptom Improvement Following Psychotherapy in Combat Veterans with PTSD. Front Psychiatry 2013; 4: 118. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24098286&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F11%2F13%2F2020.11.11.20229567.atom) 17. 17.Roberts S, Lester KJ, Hudson JL, Rapee RM, Creswell C, Cooper PJ et al. Serotonin transporter [corrected] methylation and response to cognitive behaviour therapy in children with anxiety disorders. Transl Psychiatry 2014; 4: e444. 18. 18.Ziegler C, Richter J, Mahr M, Gajewska A, Schiele MA, Gehrmann A et al. MAOA gene hypomethylation in panic disorder-reversibility of an epigenetic risk pattern by psychotherapy. Transl Psychiatry 2016; 6: e773. 19. 19.Perroud N, Salzmann A, Prada P, Nicastro R, Hoeppli ME, Furrer S et al. Response to psychotherapy in borderline personality disorder and methylation status of the BDNF gene. Transl Psychiatry 2013; 3: e207. 20. 20.Vinkers CH, Geuze E, van Rooij SJH, Kennis M, Schur RR, Nispeling DM et al. Successful treatment of post-traumatic stress disorder reverses DNA methylation marks. Mol Psychiatry 2019. 21. 21.Steiger H, Booij L, Kahan, McGregor K, Thaler L, Fletcher E et al. A longitudinal, epigenome-wide study of DNA methylation in anorexia nervosa: results in actively ill, partially weight-restored, long-term remitted and non-eating-disordered women. J Psychiatry Neurosci 2019; 44(3): 205–213. 22. 22.Farre P, Jones MJ, Meaney MJ, Emberly E, Turecki G, Kobor MS. Concordant and discordant DNA methylation signatures of aging in human blood and brain. Epigenetics Chromatin 2015; 8: 19. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13072-015-0011-y&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25977707&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F11%2F13%2F2020.11.11.20229567.atom) 23. 23.Cole SW. Elevating the perspective on human stress genomics. Psychoneuroendocrinology 2010; 35(7): 955–962. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.psyneuen.2010.06.008&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20630660&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F11%2F13%2F2020.11.11.20229567.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000280898200001&link_type=ISI) 24. 24.Jones MJ, Moore SR, Kobor MS. Principles and Challenges of Applying Epigenetic Epidemiology to Psychology. Annu Rev Psychol 2018; 69: 459–485. 25. 25.Cole SW, Hawkley LC, Arevalo JM, Cacioppo JT. Transcript origin analysis identifies antigen-presenting cells as primary targets of socially regulated gene expression in leukocytes. Proc Natl Acad Sci U S A 2011; 108(7): 3080–3085. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMDoiMTA4LzcvMzA4MCI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIwLzExLzEzLzIwMjAuMTEuMTEuMjAyMjk1NjcuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 26. 26.Weathers FW, Litz BT, Keane TM, Palmieri PA, Marx BP, Schnurr PP. The PTSD Checklist for DSM-5 (PCL-5). Scale available from the National Center for PTSD at wwwptsdvagov 2013. 27. 27.(2000). APA. Diagnostic and statistical manual of mental disorders (4th ed., Text Revision): Washington, 2000. 28. 28.Kaufman J, Birmaher B, Brent D, Rao U, Flynn C, Moreci P et al. Schedule for Affective Disorders and Schizophrenia for School-Age Children-Present and Lifetime Version (K-SADS-PL): initial reliability and validity data. J Am Acad Child Adolesc Psychiatry 1997; 36(7): 980–988. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/00004583-199707000-00021&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9204677&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F11%2F13%2F2020.11.11.20229567.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1997XF81700021&link_type=ISI) 29. 29.Leitão E, Di Persio S, Laurentino S, Wöste M, Dugas M, Kliesch S et al. The sperm epigenome does not display recurrent epimutations in patients with severely impaired spermatogenesis. Clinical Epigenetics 2020; 12(1). 30. 30.Rademacher K, Schroder C, Kanber D, Klein-Hitpass L, Wallner S, Zeschnigk M et al. Evolutionary origin and methylation status of human intronic CpG islands that are not present in mouse. Genome Biol Evol 2014; 6(7): 1579–1588. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/gbe/evu125&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24923327&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F11%2F13%2F2020.11.11.20229567.atom) 31. 31.Wallner S, Schroder C, Leitao E, Berulava T, Haak C, Beisser D et al. Epigenetic dynamics of monocyte-to-macrophage differentiation. Epigenetics Chromatin 2016; 9: 33. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13072-016-0079-z&link_type=DOI) 32. 32.Schröder C. Bioinformatics from genetic variants to methylation. 2018. 33. 33.Juhling F, Kretzmer H, Bernhart SH, Otto C, Stadler PF, Hoffmann S. metilene: fast and sensitive calling of differentially methylated regions from bisulfite sequencing data. Genome Res 2016; 26(2): 256–262. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZ2Vub21lIjtzOjU6InJlc2lkIjtzOjg6IjI2LzIvMjU2IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMTEvMTMvMjAyMC4xMS4xMS4yMDIyOTU2Ny5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 34. 34.Rahmann S, Beygo J, Kanber D, Martin M, Horsthemke B, Buiting K. Amplikyzer: Automated methylation analysis of amplicons from bisulfite flowgram sequencing. PeerJ Preprints 2013. 35. 35.Moser DA, Müller S, Hummel EM, Limberg AS, Dieckmann L, Frach L et al. Targeted bisulfite sequencing: A novel tool for the assessment of DNA methylation with high sensitivity and increased coverage. Psychoneuroendocrinology 2020. 36. 36.Rouder JN, Morey RD, Speckman PL, Province JM. Default Bayes factors for ANOVA designs. Journal of Mathematical Psychology 2012; 56(5): 356–374. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jmp.2012.08.001&link_type=DOI) 37. 37.Jarosz AF, Wiley J. What Are the Odds? A Practical Guide to Computing and Reporting Bayes Factors. The Journal of Problem Solving 2014; 7(1): 1037–1040. 38. 38.Michels KB, Binder AM, Dedeurwaerder S, Epstein CB, Greally JM, Gut I et al. Recommendations for the design and analysis of epigenome-wide association studies. Nat Methods 2013; 10(10): 949–955. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nmeth.2632&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24076989&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F11%2F13%2F2020.11.11.20229567.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000325073800014&link_type=ISI) 39. 39.Leenen FA, Muller CP, Turner JD. DNA methylation: conducting the orchestra from exposure to phenotype? Clin Epigenetics 2016; 8: 92. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13148-016-0256-8&link_type=DOI) 40. 40.Lakens D. Equivalence Tests: A Practical Primer for t Tests, Correlations, and Meta-Analyses. Soc Psychol Personal Sci 2017; 8(4): 355–362. 41. 41.Ziller MJ, Hansen KD, Meissner A, Aryee MJ. Coverage recommendations for methylation analysis by whole-genome bisulfite sequencing. Nat Methods 2015; 12(3): 230-232, 231 p following 232. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nmeth.3152&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25362363&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F11%2F13%2F2020.11.11.20229567.atom) 42. 42.Klengel T, Mehta D, Anacker C, Rex-Haffner M, Pruessner JC, Pariante CM et al. Allele-specific FKBP5 DNA demethylation mediates gene-childhood trauma interactions. Nat Neurosci 2013; 16(1): 33–41. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nn.3275&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23201972&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F11%2F13%2F2020.11.11.20229567.atom) 43. 43.Föcker M, Timmesfeld N, Scherag S, Knoll N, Singmann P, Wang-Sattler R et al. Comparison of metabolic profiles of acutely ill and short-term weight recovered patients with anorexia nervosa reveals alterations of 33 out of 163 metabolites. J Psychiatr Res 2012; 46(12): 1600–1609. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22981704&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F11%2F13%2F2020.11.11.20229567.atom) 44. 44.Zannas AS, Chrousos GP. Epigenetic programming by stress and glucocorticoids along the human lifespan. Mol Psychiatry 2017; 22(5): 640–646. 45. 45.Holbrook JD, Huang RC, Barton SJ, Saffery R, Lillycrop KA. Is cellular heterogeneity merely a confounder to be removed from epigenome-wide association studies? Epigenomics 2017; 9(8): 1143–1150. 46. 46.Cole SW, Conti G, Arevalo JM, Ruggiero AM, Heckman JJ, Suomi SJ. Transcriptional modulation of the developing immune system by early life social adversity. Proc Natl Acad Sci U S A 2012; 109(50): 20578–20583. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMjoiMTA5LzUwLzIwNTc4IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMTEvMTMvMjAyMC4xMS4xMS4yMDIyOTU2Ny5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 47. 47.O’Donovan A, Sun B, Cole S, Rempel H, Lenoci M, Pulliam L et al. Transcriptional control of monocyte gene expression in post-traumatic stress disorder. Disease markers 2011; 30(2-3): 123–132. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1155/2011/560572&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21508516&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2020%2F11%2F13%2F2020.11.11.20229567.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000291348100007&link_type=ISI) 48. 48.Powell ND, Sloan EK, Bailey MT, Arevalo JM, Miller GE, Chen E et al. Social stress up-regulates inflammatory gene expression in the leukocyte transcriptome via beta-adrenergic induction of myelopoiesis. Proc Natl Acad Sci U S A 2013; 110(41): 16574–16579. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMjoiMTEwLzQxLzE2NTc0IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjAvMTEvMTMvMjAyMC4xMS4xMS4yMDIyOTU2Ny5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=)