A Sex-Specific Genome-Wide Association Study of Depression Phenotypes in UK Biobank =================================================================================== * Patrícia Pelufo Silveira * Irina Pokhvisneva * David M Howard * Michael J. Meaney ## Abstract **Background** There are marked sex differences in the prevalence, phenotypic presentation and treatment response for major depression. While genome-wide association studies (GWAS) adjust for sex differences, to date no studies seek to identify sex-specific markers and pathways. In this study we performed a sex-stratified genome-wide association analysis for broad depression. **Methods** A genome-wide association study for broad depression was performed in the UK Biobank total participants (N=274,141), including only non-related participants, as well as separately in males (N=127,867) and females (N=146,274). Bioinformatics analyses were performed to characterize common and sex-specific markers and associated processes/pathways. **Results** We identified 11 loci passing genome level significance (P < 5* 10−8) in females and one in males. In both males and females, genetic correlations were significant between the broad depression GWA and other psychopathologies, however, correlations with educational attainment and metabolic features including body fat, waist circumference, waist-to-hip ratio and triglycerides were significant only in females. Gene-based analysis showed 147 genes significantly associated with broad depression in the total sample, 64 in the females and 53 in males. Gene-based analysis revealed “Regulation of Gene Expression” as a common biological process, but suggested sex-specific molecular mechanisms. Finally, sex-specific PRSs for broad depression outperformed total and the opposite sex PRSs in the prediction of broad MDD. **Conclusions** These findings provide evidence for sex-dependent genetic pathways for clinical depression as well as for health conditions comorbid with depression. Keywords * Depression * sex * GWAS * biological pathways * co-morbidities * polygenic scores ## Introduction Major depressive disorder (MDD) is the leading contributor to the global burden of disease and disability worldwide (1–4). Depression shows marked sex differences with women significantly more affected in terms of prevalence, repeated occurrence, symptomatology and patterns of co-morbidity (5–12). The neural correlates of MDD differ in males and females at the level of brain structure (13,14) and potentially cell-type composition (15). There are strikingly sex-specific transcriptional signatures of depression in corticolimbic brain regions associated with mood disorders (15–18). These functional disparities are likely reflected in differences in the response to antidepressant treatment (19–25), which makes the understanding of sex-specific risk factors and vulnerabilities a pressing concern. An understanding of sex-specific pathways to psychopathology is also a fruitful approach to identifying novel mechanisms of pathophysiology (26). Twin studies reveal evidence for moderate overall heritability for depression (27–29) with some evidence for greater heritability in women and for sex-specific genetic pathways to MDD (e.g. (28,30). Despite the evidence for sex-specific molecular mechanisms for depression, genome-wide association studies (GWASs) for MDD are still performed aggregating males and females into a single sample using sex as a covariate. The justification is based on the assumption that large sample sizes are essential and that the analyses of GWA datasets are adjusted by sex. There are two concerns with this approach: 1) pronounced sex differences in transcription suggest that aggregating males and females masks signals that are apparent only when considering datasets from males and females independently, and the noise of combining data regardless of sex might offset any advantage of a larger sample size; 2) collapsing male and female MDD data only permits the identification of molecular signals shared across males and females and thus to an incomplete understanding of the genetic risk factors linked to depression. The risk is that of ignoring critical, sex-specific molecular pathways and targets for the development of new therapies. The existing science suggests sex-specific approaches to treatment and is consistent with the broader objectives of precision medicine. However, to date this approach advances in the absence of an understanding of sex-specific genetic pathways. Here we directly explored sex-dependency in the genetic architecture of MDD, by performing a follow-up analysis of a published GWAS for ‘broad depression’ (31) after stratifying the analysis by sex. Our findings are consistent with previous twin studies revealing sex-specific genetic architecture to clinical depression, and implicate sex-specific molecular pathways, aligned with genome-wide transcriptomic analyses (32). ## Methods and Materials ### Study population The UK Biobank cohort is a population-based cohort consisting of 502,543 individuals aged 37-73 with genotyping data from 487,409 subjects. After retaining only Caucasians and variants with minor allele frequency <0.01 there were 276,511 individuals and 7,351,435 variants in the data set. The broad depression phenotype was defined according to Howard et al (31), resulting in 274,141 unrelated subjects with both the broad depression phenotype and genodata (see **Table S1-A** for case/control demographics). From the 132,066 subsample of related subjects, we selected one participant resulting in a test sample with 65,285 subjects (for details see **Supplemental Methods**). This research was conducted using the UK Biobank Resource under Application Number 41975. Approval for the UK Biobank was obtained by the North West Multicentre Research 580 Ethics Committee (REC reference 11/NW/0382), the National Information Governance Board for Health and Social Care and the Community Health Index Advisory Group. The raw data were used under license from UK Biobank ([http://www.ukbiobank.ac.uk/](http://www.ukbiobank.ac.uk/)). ### Association analysis Linear regression analysis using BGENIE v1.132 was used to explore the effect of each SNP on the broad depression phenotype. Genetic correlations were calculated between the MDD phenotype for each sex and 235 other behavioral and disease related traits using LD Hub (33) (for details see **Supplemental Methods**). ### Gene-based analysis Gene- and region-based analyses of the significant genes (P<2.6*10−6) were conducted using MAGMA (Multi-marker Analysis of GenoMic Annotation) available on FUMA_GWAS (Functional Mapping and Annotation of Genome-Wide Association Studies) (34). We compared male and female enrichment analyses using MetaCore™ (Clarivate Analytics, version 21.4). GTEx portal was used to identify brain expression quantitative loci (eQTL). Drug Targetor (35) was utilized to establish the potential mechanisms by which antidepressants act in male vs. female specific MDD. For details, see **Supplemental Methods**. ### Validation of the sex specific GWAS through polygenic risk scores Aligned sample size GWAS were generated and used to calculate polygenic risk score (PRSs) at different p-value thresholds using PRSice software (36,37) for each subject of the test sample. Akaike information criterion (AIC) was utilized to identify the model that explains the greatest amount of variation using the fewest possible independent variables. Lower AIC scores associate with better-fitting models (for details see **Supplemental Methods**). ## Results ### Broad depression MDD GWAS in the total sample A total of 18 independent loci showed genome-wide significance associated with broad depression (P<5*10−8) (**Data S1** and **Figure 1A**). The correlation of the beta coefficients between the Howard et al. (31) broad depression GWAS and our broad depression GWAS was r=0.91, and the correlation between the p values was r=0.72. There were 2,819 variants with P<10−6 for an association with broad depression for the total sample (**Figure 1A** and **Figure S1**). The phenotype examined did not show evidence of inflation of the test statistics due to population stratification, with any inflation due to polygenic signal (see **Table S2**). Genetic correlations from our total broad depression GWAS also replicated the findings from Howard et al. (31) **(Table 1** and **Data S2**). There were 24 significant correlations for broad depression with other traits (PFDR<0.05; **Data S2**). Correlations previously described in Howard et al. (31) between UK Biobank depression-related phenotypes and clinically defined MDD as well as schizophrenia (38) (rg=0.30) and bipolar disorder (39) (rg=0.34). View this table: [Table 1](http://medrxiv.org/content/early/2022/03/31/2022.03.30.22273201/T1) Table 1 Genetic correlations comparison between original Howard et al., 2018 Broad MDD GWAS, current study total sample GWAS, female specific and male specific Broad MDD GWAS. ![Figure 1](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/03/31/2022.03.30.22273201/F1.medium.gif) [Figure 1](http://medrxiv.org/content/early/2022/03/31/2022.03.30.22273201/F1) Figure 1 Manhattan plot of all the variants analyzed in UK Biobank for broad depression in the **(A)** total sample (N=274,141), **(B)** females (N=146,274), and **(C)** males (N=127,867). ### Sex-specific broad depression MDD GWAS We then stratified our total sample by sex to perform sex-specific GWASs separately for 127,867 non-related male and 146,274 non-related female participants (see **Table S1-B and C** for case/control demographics). Results for the associated variants with broad depression phenotype in males and females are provided in **Data S1, Figures 1B** and **S2** for females and **1C** and **S3** for males. Broad depression in females and males did not show evidence of inflation of the test statistics due to population stratification, with any inflation due to polygenic signal (**Table S2**). Eleven loci passed genome level significance (P<5*10−8) in females and one in males. Interestingly, among these loci was a SNP (rs10501696) that mapped to *GRM5* reported in Howard et al. (31), significant only in females. ### Sex-dependent genetic correlations Depression is comorbid with a range of other diseases including conditions considered to be not primarily of brain origin, especially cardio-metabolic conditions. Our findings (**Table 1**) demonstrated 25 and 13 significant (PFDR<0.05) correlations with other traits in females and males, respectively. Genetic correlations between the UK Biobank depression-related phenotypes and clinically defined MDD, schizophrenia (38), bipolar disorder (39), neuroticism (40), subjective well-being (40), PGC cross-disorder (41), insomnia (42,43) and smoking (44) were significant for both females and males. Two phenotypic categories showed striking, sex-dependent correlations, each with greater evidence for associations amongst females (**Table 1**). While analyses with both male and female participants showed associations with measures of academic achievement, the evidence for significant genetic correlations was stronger and more pervasive in females. Genetic significant (PFDR<0.05) correlations only in females included multiple analyses of years of schooling and college completion (rg=-0.21 and -0.26, respectively) (45). The most striking sex difference were those between broad MDD and GWASs for metabolic features, including body fat (46), waist circumference (47), waist-to-hip ratio (47) and triglycerides (48), significant at PFDR<0.05 only in females. ### Sex-specific broad MDD GWAS: Gene-based enrichment analysis SNPs with P<10−5 were selected to compare female and male GWAs. 147 genes were significantly associated with broad depression in the total sample, 64 in females and 53 in males (**Figures S4, S5 and S6, Data S3A-C**). In addition to *GRM5, ELAVL4*, implicated in depression and in epigenome-wide association studies of suicide (49), was significant in females, but not males. In contrast, 29 genes were significant only in males. Notably, *TCF4, NKAPL, ZKSCAN3, ZSCAN16, ZSCAN31* have been associated with depression in previous GWASs (50–54). Twenty-four common genes were found when comparing the list of total, males and females broad MDD GWAS, which were enriched for biological processes related to sensory perception and G protein-coupled receptor signaling pathways. There were several Pathway Maps in common between genes derived from the male and female broad MDD GWASs, the most significant of which is glutathione metabolism (PFDR<0.025) (**Data S4A**). Glutathione is involved in antioxidant defense and regulation of gene expression, cell proliferation and apoptosis, signal transduction, and immune response (55). Several common GO processes were found between males and females, the most significant of which were negative regulation of viral life cycle, regulation of viral release from host cell, regulation of transcription DNA-templated, regulation of nucleic acid-templated transcription, regulation of RNA biosynthetic process (all PFDR<0.002) (**Data S4B**). Common genes between males and females were enriched for diseases related to neurocognitive and neurodegenerative conditions (**Data S4C**). Enrichment analysis for the genes uniquely identified in male- or female-specific broad MDD GWAS’s is provided in **Data S5A-E**. Male-specific broad MDD GWAS genes were enriched for several Pathway Maps, GO processes and process networks (**Data S5A, B, D**) with epigenetic regulation of gene expression as the recurrently enriched pathway. Female-specific broad MDD GWAS genes did not show significant enrichment for Pathway Maps but, as in males, were enriched for regulation of gene expression (PFDR<0.01, **Data S5C**). It is noteworthy that “regulation of gene expression” was the most significant common GO process associated with genes from both male- and female-specific broad MDD GWAS, but this finding was due to sex-specific gene networks (**Figure 2**). In males, “regulation of gene expression” was mapped to genes including *TCF4* as well as an impressive number of genes coding for histone protein variants. *TCF4* is a known regulator of epigenetic states including DNA methylation (56). In females, “regulation of gene expression” was associated with a number of neurexin-related genes, *DRD2* and *GRM5* genes. ![Figure 2](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/03/31/2022.03.30.22273201/F2.medium.gif) [Figure 2](http://medrxiv.org/content/early/2022/03/31/2022.03.30.22273201/F2) Figure 2 Enrichment analysis of genes associated with male-specific or female specific broad MDD GWAS. Processes and diseases are commonly enriched in both sexes, but due to unique, sex-specific mechanisms. Several psychopathology and neurodegenerative disease terms were associated with genes from both male- and female-specific broad MDD GWAS (**Figure 2**), but this finding was due to sex-specific gene networks. In males, brain pathology was associated with genes involved in epigenetic processes and regulation of neurotransmitter release. In females, brain pathology was related to *DRD2* signaling, and an important number of genes related to adaptive immunity. Taken together, the findings depicted in **Figure 2** suggest that MDD-related alterations in gene expression regulation and brain pathology in males and females occur via unique, sex-specific mechanisms. ### Sex-specific broad MDD GWAS: eQTL’s, transcription factors and drug targets We sought eQTLs among the SNPs identified in male- and female-specific broad MDD GWAS (top 125 significant SNPs, P <10−7 in females and P <9.16*10−7 in males). There was a striking sex difference with almost twice the percentage of SNP/eQTLs in males [18% compared with females 10% (χ2 = 945.9, p<0.00001)] considering all brain regions together. The cerebellum showed a high number of eQTLs in both male- and female-specific GWASs (**Figure S7**). There was a highly sex-specific distribution of identified eQTLs across brain regions. eQTLs from the female-specific GWAs in the caudate-basal ganglia (χ2 = 38.8; p<.00001), putamen-basal ganglia (χ2 = 31.6; p<.00001), and hippocampus (χ2 = 19.7; p<.00001) significantly exceeded that derived from the male-specific GWAs. The most notable finding was that of increased eQTLs in the basal ganglia of females, including the ventral striatum/nucleus accumbens, which are prominent dopamine target regions. A conspicuous pattern apparent in both males and females was the enrichment for transcription factors linked to immune responses and NFK-ß signaling, including FOXP3, RBPJ kappa, RUNX1 and TAL1. BMAL1, a critical regulator of circadian rhythms (57), was highly enriched amongst the genes from both the male- and female-specific GWASs (**Data S6A-B**). This finding is interesting considering the highly significant genetic correlation between both the male- and female-specific GWASs for broad MDD and that for insomnia (see **Table 1**). Transcription factors uniquely associated with genes identified in the female broad MDD GWAS were linked to oxidative stress, apoptosis and type II diabetes (e.g. HNF3-beta, MafA, p63, RelA, VDR), which aligns with the genetic correlation of the female-specific GWAS with cardio-metabolic conditions (see **Table 1**). Transcription factors linked specifically with genes identified in the male-specific broad MDD GWAS were related to tissue and neuron differentiation as well as epigenetic processes (e.g. E2F1, Esrrb, NRSF, ZFX, ZNF423), which is consistent with the results of the gene enrichment analyses (see **Figure 2**) that underscore the potential role for chromatin remodeling factors in males. The central biological processes associated with broad MDD and targeted by antidepressants were strongly associated with: (1) epigenetic processes such as chromatin assembly, cell cycle regulation as well as inflammation (mainly through IL-7), in males; (2) neuronal migration, regulation of neurotrophic factors and synaptic plasticity, and dopamine neurotransmission, in females (**Data S7A-B**). The implication of the dopamine neurotransmission is consistent with the pathways identified in females in the gene enrichment analyses (**Figure 2**), likewise, the finding of chromatin assembly in males, a process intimately linked to histone proteins. These findings suggest that the biological processes targeted by antidepressants and involved in genetic architecture of MDD differ in males and females, implying sex-dependent therapeutic pathways. ### Validation of the sex-specific broad MDD GWAS using polygenic risk scores We calculated PRSs using our total, male- and female-specific GWASs in a UK Biobank test sample of 65,285 non-related individuals (**Table S3**). In males, the male-specific PRS showed better predictive value for the broad MDD outcome (**Figure 3B**) than did the total PRS derived from a similar sized GWAS (**Figure 3C**) or the female-specific PRS (**Figure 3A**) (AICm=33962.86, AICf=34001.21, AICt=33965.37). Similarly, the PRS derived from the female-specific GWAS showed better predictive value for the broad depression outcome among females (**Figure 3E**) than did the total PRS from a GWAS of comparable size (**Figure 3G**) and male-specific PRS (**Figure 3F**) (AICf=48987.21, AICm=49100.48, AICt=49016.82). The well-established effect of GWAS sample size to best predict the outcomes was observed, as the combined, mixed GWAS sample had a similar predictive capacity to the sex-specific male or female-specific GWAS (**Figure 3D and 3H**). Although a larger GWAS does indeed have a similar predictive capacity than the sex-specific GWAS, it does not have the ability to disentangle sex-specific mechanisms and drug targets. ![Figure 3](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/03/31/2022.03.30.22273201/F3.medium.gif) [Figure 3](http://medrxiv.org/content/early/2022/03/31/2022.03.30.22273201/F3) Figure 3 Comparison between odds ratios (ORs) of associations between the polygenic risk scores (PRS) for Broad MDD in a test sample of males (A-D) and females (E-H) using the male-specific broad MDD GWAS (M129), female-specific broad MDD GWAS (F147), or a mixed sample MDD GWAS of similar size (T129 or T147, respectively). The ORs of associations using the total mixed sample MDD GWAS (T276) is shown in panels D for males participants and panel H for female participants. ## Discussion Sex differences in the prevalence and severity of depression are well established. Nevertheless, research to elucidate the genetic architecture of depression has not been designed to examine sex-specific pathways (58). The current study addresses this gap. To our knowledge our results are the first extensive molecular analysis of sex-specific genetic pathways for MDD. Our findings reveal evidence for sex-differences in the genetic pathways to MDD and are consistent with previous, genetically-informed epidemiological studies (27,28) suggesting that the influences of genetic variants on the risk for depression are sex-specific. We showed that a PRS derived from a female-specific, genome-wide analysis outperformed a version derived from a male-specific analysis in the prediction of broad MDD in females; the reverse was true for the prediction of broad MDD in males (**Figure 3**). While the gene enrichment analysis identified “regulation of gene expression” as a common biological process linking genetic variants to broad MDD, the underlying gene pathways were highly sex-dependent (**Figure 2**). Regulation of gene expression in males was associated with epigenetic mechanisms, notably variants in genes coding for the core histone proteins. In contrast, regulation of gene expression in females associated with neurexin as well as the DRD2 and mGluR5 receptors. The implication of the DRD2 receptor in the “regulation of gene expression” pathway is consistent with previous reports. In their MDD GWAS Howard et al. (31) explored drug – gene interactions and identified the DRD2 receptor as the primary target. Levey et al. (59) mapped genes from their GWAS meta-analysis of depression to expression QTL data in GTEx revealing a transcriptome-wide association with a predicted decrease in nucleus accumbens DRD2 expression. Our findings suggest that these results might be largely driven by data from women. Note the inclusion of the DRD2 receptor in the female-specific “Regulation of Gene Expression Pathway” (**Figure 2**). Our gene-based eQTL analysis of the female-specific GWAS revealed an enrichment for eQTL’s in the caudate, which includes the dorsal and ventral striatum/nucleus accumbens, brain regions rich in DRD2 receptors. Considerable evidence from both pre-clinical and clinical studies show the importance of dopaminergic projections from the ventral tegmental area (VTA) to nucleus accumbens for depression (60). Anhedonia is a core feature of depression and the mesolimbic dopamine pathway mediates activation of the reward system (61). Deep brain stimulation affecting the nucleus accumbens has sustained efficacy in treatment-resistant depression (62). The mesolimbic dopamine pathway is critical for chronic stress-induced depressive-like behaviors in rodents (63–65). Buproprion, a mixed norepinephrine/dopamine-reuptake inhibitor, is an accepted treatment for depression with efficacy comparable to that of SSRI’s (66,67). Pre-clinical studies reveal greater DRD2-mediated reward enhancing effects of buproprion in females than males (68). Rodent studies consistently show sex differences in dopaminergic systems in relation to reward processing and activation of behavioral responses to stress (69). Human PET imaging studies using [11C]raclopride, a DRD2/3-specific ligand, show in vivo evidence for stress-induced DRD2 signaling in the ventral striatum (70) and decreased ventral striatal D2 synaptic activity in depressed patients (71). Human neuroimaging studies reveal greater stress-induced activation of the ventral striatum in women compared to men (72,73). Eleven loci passed genome level significance (P<5*10−8) in females including a SNP (rs10501696) mapped to *GRM5* also reported in Howard et al. (31). Wray et al. (50) reported on a meta-analysis of seven independent cohorts and identified a significant association of *GRM5* with MDD. *GRM5* gene, which encodes the mGluR5 metabotropic glutamate receptor, was also featured in the female-specific “Regulation of Gene Expression Pathway” (**Figure 2**). These sex-specific *GRM5* findings are consistent with Gray et al. (74) reporting extensive sex-dependent differences in glutamate receptor gene expression in post-mortem samples from MDD and control subjects, and higher expression levels of *GRIN1, GRIN2A-D, GRIA2-4, GRIK1-2, GRM1, GRM4, GRM5* and *GRM7* in female MDD patients; amongst males only *GRM5* expression differed and in the opposite direction from females. The G-protein coupled mGluR5 receptor is located both pre- and post-synaptically, regulating synaptic plasticity (75). mGluR5-/- mice show increased stress-induced depressive-like behaviors (76). Reductions in mGluR5 protein levels are apparent in multiple rodent models of depression (77,78), in contrast to the increases in mGluR5 levels observed following anti-depressant treatments (79). *In vivo* PET imaging with [11C]ABP688 reveals lower mGluR5 receptor density in MDD in several regions (80), which seems to be associated with response to antidepressant treatment (81). Our gene level analysis identified “regulation of gene expression” as the primary biological process for both males and females, revealing the involvement of neurexin signaling in females (**Figure 2**), which is critical for the formation of neural circuits and implicated in a range of neuropsychiatric disorders (82,83). To our knowledge previous GWASs have not provided evidence for an association between polymorphisms in neurexin genes and MDD. However, alternative gene sequencing platforms do suggest a potential role for neurexin in MDD. Rucker et al. (84) found significant enrichment of genomic and exonic deletion CNVs in cases of recurrent depression. The analysis showed overlap with CNVs previously associated with schizophrenia, including neurexin 1. This same exonic *NRXN1* deletion CNV was also associated with a poor treatment response to antidepressants (85). An intronic SNP in neurexin 3 was significantly associated with symptom improvement following citalopram/escitalopram treatment (86). Transcriptomic analyses with post-mortem human brain samples reveal significant sex differences in both the expression and splicing of neurexin genes (87,88). Studies with model systems suggest a prominent role for neurexin in establishing and maintaining sexually-dimorphic neuronal circuits (89). The “Regulation of Gene Expression” process in males was associated with histone modifications. An analysis from the Psychiatric Genomics Consortium (90) examined common pathways across GWASs for schizophrenia, major depression and bipolar disorder, identifying histone methylation as the strongest emerging common process. Pre-clinical models of depression underscore the importance of histone methylation (91,92), with emerging evidence from human post-mortem analyses (93). Our analysis identified genes coding for actual histone protein variants rather than enzymes associated with methylation. Histone protein variants emerge from a histone gene cluster as key components of the transcriptional machinery (94). Histone variants affect nucleosome dynamics associated with activity-dependent transcription in brain (95,96) and differ in the capacity for post-translational modification. Hodes et al. (97) summarized the existing evidence for sex-dependent effects of epigenetic mechanisms in rodent models of depression. The extensive sex-dependent transcriptomic profiles of depression together with the sex differences in eQTLs described here points to an important area for future analyses. While we emphasize the sex-dependent features of our dataset, there were important points of convergence between male and female analyses. Our transcription factor enrichment analysis revealed common transcriptional signals, most notably factors involved in NFK-ß-signaling such as FOXP3, TAL1 and RUNX1, consistent with the proposed association between inflammation and MDD (98). Another common factor was BMAL1, a regulator of circadian rhythms and sleep. Genetic correlation between the broad MDD GWAS and insomnia was significant in both sexes (**Table 1**). Notably absent in the transcription factor enrichment analysis were sex-steroid receptors, which serve as ligand-gated regulators of gene expression. Genetic correlations for male- and female-specific GWASs revealed significant associations with previous GWASs for a range of psychiatric disorders (see **Table 1**). Nevertheless, a striking sex difference in the genetic correlations were those between broad MDD and GWASs for metabolic features, including body fat, waist circumference, waist-to-hip ratio and triglycerides, all of which associate with an increased risk for cardio-metabolic disease and were highly correlated in women, not men (**Table 1**). Co-morbidity between MDD and cardio-metabolic diseases is well established (99) being significantly more prevalent in women (100–103) and see (104) for a review]. Marcus et al. (105) reported that women with MDD were more likely to endorse changes in appetite and weight gain than were men in the STAR*D ([www.star-d.org](http://www.star-d.org)) trial. Our findings suggest that sex-specific genetic pathways may explain, in part, this increased co-morbidity in women. The sex-specific MDD pathway analyses highlighted dopamine signaling in females and analyses of the sex-specific GWAS’s showed greater eQTLs in the basal ganglia in women, including the caudate and nucleus accumbens. Dopamine signaling through DRD2 receptors in these regions regulates appetite and feeding behavior (106). Our study was an exploratory investigation into potential sex-dependent genetic pathways to depression. Our genome-wide analyses were not sufficiently powered to provide definitive identification of sex-dependent loci associated with MDD, requiring extension with sex-based stratification of larger sample sizes and diverse populations. Nevertheless, we note that despite the comparatively smaller sample size, the female-specific GWAS did yield 11 loci that passed genome-wide statistical significance. A comparably powered analysis failed to yield significant loci in the male-specific GWAS. Sex-stratified analyses with larger sample sizes are required to examine whether this reflects a greater genetic contribution to MDD in women. The reliance on a self-reported, “broad MDD” designation is a limitation of this exploratory study. However, Howard et al. (31) showed a highly significant genetic correlation (rG= 0.86) between broad depression and clinically diagnosed MDD [also see (107)]. The analyses are also limited by the focus on British Caucasians, with subjects generally exceeding the health and wealth of the overall British population. In summary, our exploratory study suggests that the genetic background linked to human major depression includes sex specific variants. There are both common and unique biological mechanisms mapped from male-specific and female-specific broad MDD GWAS. Common processes like regulation of gene expression and diseases like brain pathology emerged from sex-specific gene networks. Our findings may contribute for the development of tailored therapeutic options. The consideration of sex-specific molecular alterations related to major depression during disease management can lead to more effective response to antidepressants. Significantly larger samples across more diverse populations will be required to meet this objective. Our results are intended to add to the rationale for studies of sex-specific mechanisms. ## Supporting information Supplemental Methods [[supplements/273201_file02.docx]](pending:yes) Supplemental Tables [[supplements/273201_file03.docx]](pending:yes) Supplemental figures [[supplements/273201_file04.pptx]](pending:yes) Supplemental Data 1 [[supplements/273201_file05.xlsx]](pending:yes) Supplemental Data 2 [[supplements/273201_file06.xlsx]](pending:yes) Supplemental Data 3 [[supplements/273201_file07.xlsx]](pending:yes) Supplemental Data 4 [[supplements/273201_file08.xlsx]](pending:yes) Supplemental Data 5 [[supplements/273201_file09.xlsx]](pending:yes) Supplemental Data 6 [[supplements/273201_file10.xlsx]](pending:yes) Supplemental Data 7 [[supplements/273201_file11.xlsx]](pending:yes) ## Data Availability All data produced in the present study are available upon reasonable request to the authors and with permission of UK Biobank. ## Disclosures The authors report no biomedical financial interests or potential conflicts of interest. ## Acknowledgements We are grateful to UK Biobank participants. This research has been conducted using the UK Biobank Resource under application number 41975. This research was funded by the Hope for Depression Research Foundation (MJM). * Received March 30, 2022. * Revision received March 30, 2022. * Accepted March 31, 2022. * © 2022, Posted by Cold Spring Harbor Laboratory The copyright holder for this pre-print is the author. All rights reserved. The material may not be redistributed, re-used or adapted without the author's permission. ## References 1. 1.Vos T, Barber RM, Bell B, Bertozzi-Villa A, Biryukov S, Bolliger I, et al. (2015): Global, regional, and national incidence, prevalence, and years lived with disability for 301 acute and chronic diseases and injuries in 188 countries, 1990-2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet (London, England) 386: 743–800. 2. 2.James SL, Abate D, Abate KH, Abay SM, Abbafati C, Abbasi N, et al. (2018): Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet (London, England) 392: 1789–1858. 3. 3.Smith K (2014): Mental health: a world of depression. Nature 515: 181. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25391942&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 4. 4.Rehm J, Shield KD (2019): Global Burden of Disease and the Impact of Mental and Addictive Disorders. Curr Psychiatry Rep 21. [https://doi.org/10.1007/S11920-019-0997-0](https://doi.org/10.1007/S11920-019-0997-0) 5. 5.Weissman MM, Klerman GL (1977): Sex differences and the epidemiology of depression. Arch Gen Psychiatry 34: 98–111. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/archpsyc.1977.01770130100011&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=319772&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1977CT62100011&link_type=ISI) 6. 6.Frank E, Carpenter LL, Kupfer DJ (1988): Sex differences in recurrent depression: are there any that are significant? Am J Psychiatry 145: 41–45. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1176/ajp.145.1.41&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=3337291&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1988L631300007&link_type=ISI) 7. 7.Young MA, Fogg LF, Scheftner WA, Keller MB, Fawcett JA (1990): Sex differences in the lifetime prevalence of depression: does varying the diagnostic criteria reduce the female/male ratio? J Affect Disord 18: 187–192. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/0165-0327(90)90035-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=2139063&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1990CV88600005&link_type=ISI) 8. 8.Kessler RC, McGonagle KA, Swartz M, Blazer DG, Nelson CB (1993): Sex and depression in the National Comorbidity Survey. I: Lifetime prevalence, chronicity and recurrence. J Affect Disord 29: 85–96. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/0165-0327(93)90026-G&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=8300981&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1993MK59200003&link_type=ISI) 9. 9.Kessler RC, Berglund P, Demler O, Jin R, Merikangas KR, Walters EE (2005): Lifetime prevalence and age-of-onset distributions of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry 62: 593–602. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/archpsyc.62.6.593&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15939837&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000229628400003&link_type=ISI) 10. 10.Gater R, Tansella M, Korten A, Tiemens BG, Mavreas VG, Olatawura MO (1998): Sex differences in the prevalence and detection of depressive and anxiety disorders in general health care settings: report from the World Health Organization Collaborative Study on Psychological Problems in General Health Care. Arch Gen Psychiatry 55: 405–413. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/archpsyc.55.5.405&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9596043&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000073540300003&link_type=ISI) 11. 11.Marcus SM, Young EA, Kerber KB, Kornstein S, Farabaugh AH, Mitchell J, et al. (2005): Gender differences in depression: findings from the STAR*D study. J Affect Disord 87: 141–150. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jad.2004.09.008&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15982748&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000231321500001&link_type=ISI) 12. 12.MacKenzie CS, Reynolds K, Cairney J, Streiner DL, Sareen J (2012): Disorder-specific mental health service use for mood and anxiety disorders: associations with age, sex, and psychiatric comorbidity. Depress Anxiety 29: 234–242. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/da.20911&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22065571&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000302904700010&link_type=ISI) 13. 13.Hastings RS, Parsey R V., Oquendo MA, Arango V, Mann JJ (2004): Volumetric analysis of the prefrontal cortex, amygdala, and hippocampus in major depression. Neuropsychopharmacology 29: 952–959. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/sj.npp.1300371&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=14997169&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000220919600012&link_type=ISI) 14. 14.Kong L, Chen K, Womer F, Jiang W, Luo X, Driesen N, et al. (2013): Sex differences of gray matter morphology in cortico-limbic-striatal neural system in major depressive disorder. J Psychiatr Res 47: 733–739. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jpsychires.2013.02.003&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23453566&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 15. 15.Seney ML, Huo Z, Cahill K, French L, Puralewski R, Zhang J, et al. (2018): Opposite Molecular Signatures of Depression in Men and Women. Biol Psychiatry 84: 18–27. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.biopsych.2018.01.017&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29548746&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 16. 16.Labonté B, Engmann O, Purushothaman I, Menard C, Wang J, Tan C, et al. (2017): Sex-specific transcriptional signatures in human depression. Nat Med 23: 1102–1111. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nm.4386&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28825715&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 17. 17.Issler O, van der Zee YY, Ramakrishnan A, Wang J, Tan C, Loh YHE, et al. (2020): Sex-Specific Role for the Long Non-coding RNA LINC00473 in Depression. Neuron 106: 912-926.e5. 18. 18.Seney ML, Glausier J, Sibille E (2022): Large-Scale Transcriptomics Studies Provide Insight Into Sex Differences in Depression. Biol Psychiatry 91: 14–24. 19. 19.Kornstein SG, Schatzberg AF, Thase ME, Yonkers KA, McCullough JP, Keitner GI, et al. (2000): Gender differences in treatment response to sertraline versus imipramine in chronic depression. Am J Psychiatry 157: 1445–1452. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1176/appi.ajp.157.9.1445&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10964861&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000089056800014&link_type=ISI) 20. 20.Martényi F, Dossenbach M, Mraz K, Metcalfe S (2001): Gender differences in the efficacy of fluoxetine and maprotiline in depressed patients: a double-blind trial of antidepressants with serotonergic or norepinephrinergic reuptake inhibition profile. Eur Neuropsychopharmacol 11: 227–232. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0924-977X(01)00089-X&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11418283&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000169938400006&link_type=ISI) 21. 21.Khan A, Brodhead AE, Schwartz KA, Kolts RL, Brown WA (2005): Sex differences in antidepressant response in recent antidepressant clinical trials. J Clin Psychopharmacol 25: 318–324. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/01.jcp.0000168879.03169.ce&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16012273&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000230876800005&link_type=ISI) 22. 22.Berlanga C, Flores-Ramos M (2006): Different gender response to serotonergic and noradrenergic antidepressants. A comparative study of the efficacy of citalopram and reboxetine. J Affect Disord 95: 119–123. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jad.2006.04.029&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16782204&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000241477700016&link_type=ISI) 23. 23.Young EA, Kornstein SG, Marcus SM, Harvey AT, Warden D, Wisniewski SR, et al. (2009): Sex differences in response to citalopram: a STAR*D report. J Psychiatr Res 43: 503–511. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jpsychires.2008.07.002&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18752809&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 24. 24.Sramek JJ, Murphy MF, Cutler NR (2016): Sex differences in the psychopharmacological treatment of depression. Dialogues Clin Neurosci 18: 447–457. 25. 25.LeGates TA, Kvarta MD, Thompson SM (2019): Sex differences in antidepressant efficacy. Neuropsychopharmacology 44: 140–154. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41386-018-0156-z&link_type=DOI) 26. 26.Rutter M, Caspi A, Moffitt TE (2003): Using sex differences in psychopathology to study causal mechanisms: unifying issues and research strategies. J Child Psychol Psychiatry 44: 1092–1115. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/1469-7610.00194&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=14626453&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000186442100003&link_type=ISI) 27. 27.Kendler KS, Gardner CO, Neale MC, Prescott CA (2001): Genetic risk factors for major depression in men and women: similar or different heritabilities and same or partly distinct genes? Psychol Med 31: 605–616. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1017/S0033291701003907&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11352363&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000168776900005&link_type=ISI) 28. 28.Kendler KS, Gatz M, Gardner CO, Pedersen NL (2006): A Swedish national twin study of lifetime major depression. Am J Psychiatry 163: 109–114. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1176/appi.ajp.163.1.109&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16390897&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000234367100021&link_type=ISI) 29. 29.Flint J, Kendler KS (2014): The Genetics of Major Depression. Neuron 81: 1214. 30. 30.Kendler KS, Prescott CA, Myers J, Neale MC (2003): The structure of genetic and environmental risk factors for common psychiatric and substance use disorders in men and women. Arch Gen Psychiatry 60: 929–937. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/archpsyc.60.9.929&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12963675&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000185155800010&link_type=ISI) 31. 31.Howard DM, Adams MJ, Shirali M, Clarke TK, Marioni RE, Davies G, et al. (2018): Genome-wide association study of depression phenotypes in UK Biobank identifies variants in excitatory synaptic pathways. Nat Commun 9. [https://doi.org/10.1038/S41467-018-03819-3](https://doi.org/10.1038/S41467-018-03819-3) 32. 32.Wu W, Howard D, Sibille E, French L (2021): Differential and spatial expression meta-analysis of genes identified in genome-wide association studies of depression. Transl Psychiatry 11. [https://doi.org/10.1038/S41398-020-01127-3](https://doi.org/10.1038/S41398-020-01127-3) 33. 33.Zheng J, Erzurumluoglu AM, Elsworth BL, Kemp JP, Howe L, Haycock PC, et al. (2017): LD Hub: a centralized database and web interface to perform LD score regression that maximizes the potential of summary level GWAS data for SNP heritability and genetic correlation analysis. Bioinformatics 33: 272–279. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bioinformatics/btw613&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27663502&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 34. 34.Watanabe K, Taskesen E, Van Bochoven A, Posthuma D (2017): Functional mapping and annotation of genetic associations with FUMA. Nat Commun 8. [https://doi.org/10.1038/S41467-017-01261-5](https://doi.org/10.1038/S41467-017-01261-5) 35. 35.Gaspar HA, Hübel C, Breen G (2019): Drug Targetor: a web interface to investigate the human druggome for over 500 phenotypes. Bioinformatics 35: 2515–2517. 36. 36.Choi SW, O’Reilly PF (2019): PRSice-2: Polygenic Risk Score software for biobank-scale data. Gigascience 8. [https://doi.org/10.1093/GIGASCIENCE/GIZ082](https://doi.org/10.1093/GIGASCIENCE/GIZ082) 37. 37.Chen LM, Yao N, Garg E, Zhu Y, Nguyen TTT, Pokhvisneva I, et al. (2018): PRS-on-Spark (PRSoS): a novel, efficient and flexible approach for generating polygenic risk scores. BMC Bioinformatics 19. [https://doi.org/10.1186/S12859-018-2289-9](https://doi.org/10.1186/S12859-018-2289-9) 38. 38.Ripke S, Neale BM, Corvin A, Walters JTR, Farh KH, Holmans PA, et al. (2014): Biological insights from 108 schizophrenia-associated genetic loci. Nature 511: 421–427. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature13595&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25056061&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000339335700037&link_type=ISI) 39. 39.Sklar P, Ripke S, Scott LJ, Andreassen OA, Cichon S, Craddock N, et al. (2011): Large-scale genome-wide association analysis of bipolar disorder identifies a new susceptibility locus near ODZ4. Nat Genet 43: 977–985. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.943&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21926972&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 40. 40.Okbay A, Baselmans BML, De Neve JE, Turley P, Nivard MG, Fontana MA, et al. (2016): Genetic variants associated with subjective well-being, depressive symptoms, and neuroticism identified through genome-wide analyses. Nat Genet 48: 624–633. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.3552&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27089181&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 41. 41.Smoller JW, Kendler K, Craddock N, Lee PH, Neale BM, Nurnberger JN, et al. (2013): Identification of risk loci with shared effects on five major psychiatric disorders: a genome-wide analysis. Lancet (London, England) 381: 1371–1379. 42. 42.Hammerschlag AR, Stringer S, De Leeuw CA, Sniekers S, Taskesen E, Watanabe K, et al. (2017): Genome-wide association analysis of insomnia complaints identifies risk genes and genetic overlap with psychiatric and metabolic traits. Nat Genet 49: 1584–1592. 43. 43.Lane JM, Liang J, Vlasac I, Anderson SG, Bechtold DA, Bowden J, et al. (2017): Genome-wide association analyses of sleep disturbance traits identify new loci and highlight shared genetics with neuropsychiatric and metabolic traits. Nat Genet 49: 274–281. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.3749&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27992416&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 44. 44.Furberg H, Kim Y, Dackor J, Boerwinkle E, Franceschini N, Ardissino D, et al. (2010): Genome-wide meta-analyses identify multiple loci associated with smoking behavior. Nat Genet 42: 441–447. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.571&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20418890&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000277179500017&link_type=ISI) 45. 45.Rietveld CA, Medland SE, Derringer J, Yang J, Esko T, Martin NW, et al. (2013): GWAS of 126,559 individuals identifies genetic variants associated with educational attainment. Science 340: 1467–1471. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIzNDAvNjEzOS8xNDY3IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDMvMzEvMjAyMi4wMy4zMC4yMjI3MzIwMS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 46. 46.Lu Y, Day FR, Gustafsson S, Buchkovich ML, Na J, Bataille V, et al. (2016): New loci for body fat percentage reveal link between adiposity and cardiometabolic disease risk. Nat Commun 7. [https://doi.org/10.1038/NCOMMS10495](https://doi.org/10.1038/NCOMMS10495) 47. 47.Shungin D, Winkler T, Croteau-Chonka DC, Ferreira T, Locke AE, Mägi R, et al. (2015): New genetic loci link adipose and insulin biology to body fat distribution. Nature 518: 187– 196. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature14132&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25673412&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000349190300030&link_type=ISI) 48. 48.Teslovich TM, Musunuru K, Smith A V., Edmondson AC, Stylianou IM, Koseki M, et al. (2010): Biological, clinical and population relevance of 95 loci for blood lipids. Nature 466: 707–713. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature09270&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20686565&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000280562500029&link_type=ISI) 49. 49.Policicchio S, Washer S, Viana J, Iatrou A, Burrage J, Hannon E, et al. (2020): Genome-wide DNA methylation meta-analysis in the brains of suicide completers. Transl Psychiatry 10. [https://doi.org/10.1038/S41398-020-0752-7](https://doi.org/10.1038/S41398-020-0752-7) 50. 50.Wray NR, Ripke S, Mattheisen M, Trzaskowski M, Byrne EM, Abdellaoui A, et al. (2018): Genome-wide association analyses identify 44 risk variants and refine the genetic architecture of major depression. Nat Genet 50: 668–681. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41588-018-0090-3&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29700475&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 51. 51.Nagel M, Jansen PR, Stringer S, Watanabe K, De Leeuw CA, Bryois J, et al. (2018): Meta-analysis of genome-wide association studies for neuroticism in 449,484 individuals identifies novel genetic loci and pathways. Nat Genet 50: 920–927. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 52. 52.Howard DM, Adams MJ, Clarke TK, Hafferty JD, Gibson J, Shirali M, et al. (2019): Genome-wide meta-analysis of depression identifies 102 independent variants and highlights the importance of the prefrontal brain regions. Nat Neurosci 22: 343–352. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 53. 53.Amare AT, Vaez A, Hsu YH, Direk N, Kamali Z, Howard DM, et al. (2020): Bivariate genome-wide association analyses of the broad depression phenotype combined with major depressive disorder, bipolar disorder or schizophrenia reveal eight novel genetic loci for depression. Mol Psychiatry 25: 1420–1429. 54. 54.Dall’Aglio L, Lewis CM, Pain O (2021): Delineating the Genetic Component of Gene Expression in Major Depression. Biol Psychiatry 89: 627–636. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.biopsych.2020.09.010&link_type=DOI) 55. 55.Wu G, Fang YZ, Yang S, Lupton JR, Turner ND (2004): Glutathione metabolism and its implications for health. J Nutr 134: 489–492. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToibnV0cml0aW9uIjtzOjU6InJlc2lkIjtzOjk6IjEzNC8zLzQ4OSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIyLzAzLzMxLzIwMjIuMDMuMzAuMjIyNzMyMDEuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 56. 56.Kennedy AJ, Rahn EJ, Paulukaitis BS, Savell KE, Kordasiewicz HB, Wang J, et al. (2016): Tcf4 Regulates Synaptic Plasticity, DNA Methylation, and Memory Function. Cell Rep 16: 2666–2685. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.celrep.2016.08.004&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27568567&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 57. 57.Bunger MK, Wilsbacher LD, Moran SM, Clendenin C, Radcliffe LA, Hogenesch JB, et al. (2000): Mop3 is an essential component of the master circadian pacemaker in mammals. Cell 103: 1009–1017. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0092-8674(00)00205-1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11163178&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000166040600005&link_type=ISI) 58. 58.Goldstein JM, Hale T, Foster SL, Tobet SA, Handa RJ (2019): Sex differences in major depression and comorbidity of cardiometabolic disorders: impact of prenatal stress and immune exposures. Neuropsychopharmacology 44: 59–70. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41386-018-0146-1&link_type=DOI) 59. 59.Levey DF, Stein MB, Wendt FR, Pathak GA, Zhou H, Aslan M, et al. (2021): Bi-ancestral depression GWAS in the Million Veteran Program and meta-analysis in >1.2 million individuals highlight new therapeutic directions. Nat Neurosci 24. [https://doi.org/10.1038/s41593-021-00860-2](https://doi.org/10.1038/s41593-021-00860-2) 60. 60.Nestler EJ, Carlezon WA (2006): The mesolimbic dopamine reward circuit in depression. Biol Psychiatry 59: 1151–1159. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.biopsych.2005.09.018&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16566899&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000238692300007&link_type=ISI) 61. 61.Pizzagalli DA (2014): Depression, stress, and anhedonia: toward a synthesis and integrated model. Annu Rev Clin Psychol 10: 393–423. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1146/annurev-clinpsy-050212-185606&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24471371&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 62. 62.Bewernick BH, Kayser S, Sturm V, Schlaepfer TE (2012): Long-term effects of nucleus accumbens deep brain stimulation in treatment-resistant depression: evidence for sustained efficacy. Neuropsychopharmacology 37: 1975–1985. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/npp.2012.44&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22473055&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000306494600001&link_type=ISI) 63. 63.Krishnan V, Han MH, Graham DL, Berton O, Renthal W, Russo SJ, et al. (2007): Molecular adaptations underlying susceptibility and resistance to social defeat in brain reward regions. Cell 131: 391–404. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2007.09.018&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17956738&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000250507700027&link_type=ISI) 64. 64.Chaudhury D, Walsh JJ, Friedman AK, Juarez B, Ku SM, Koo JW, et al. (2013): Rapid regulation of depression-related behaviours by control of midbrain dopamine neurons. Nature 493: 532–536. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature11713&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23235832&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000313871400038&link_type=ISI) 65. 65.Tye KM, Mirzabekov JJ, Warden MR, Ferenczi EA, Tsai HC, Finkelstein J, et al. (2013): Dopamine neurons modulate neural encoding and expression of depression-related behaviour. Nature 493: 537–541. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature11740&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23235822&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000313871400039&link_type=ISI) 66. 66.Gelenberg AJ, Marlene Freeman CP, Markowitz JC, Rosenbaum JF, Thase ME, Trivedi MH, et al. (2010): PRACTICE GUIDELINE FOR THE Treatment of Patients With Major Depressive Disorder Third Edition WORK GROUP ON MAJOR DEPRESSIVE DISORDER. Retrieved March 20, 2022, from [http://www.psychiatryonline.com/pracGuide/pracGuideTopic_7.aspx](http://www.psychiatryonline.com/pracGuide/pracGuideTopic_7.aspx). 67. 67.Ang YS, Kaiser R, Deckersbach T, Almeida J, Phillips ML, Chase HW, et al. (2020): Pretreatment Reward Sensitivity and Frontostriatal Resting-State Functional Connectivity Are Associated With Response to Bupropion After Sertraline Nonresponse. Biol Psychiatry 88: 657–667. 68. 68.Barrett ST, Geary TN, Steiner AN, Bevins RA (2017): Sex differences and the role of dopamine receptors in the reward-enhancing effects of nicotine and bupropion. Psychopharmacology (Berl) 234: 187–198. 69. 69.Becker JB, Chartoff E (2019): Sex differences in neural mechanisms mediating reward and addiction. Neuropsychopharmacology 44: 166–183. 70. 70.Pruessner JC, Champagne F, Meaney MJ, Dagher A (2004): Dopamine release in response to a psychological stress in humans and its relationship to early life maternal care: a positron emission tomography study using [11C]raclopride. J Neurosci 24: 2825–2831. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Njoiam5ldXJvIjtzOjU6InJlc2lkIjtzOjEwOiIyNC8xMS8yODI1IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDMvMzEvMjAyMi4wMy4zMC4yMjI3MzIwMS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 71. 71.Peciña M, Sikora M, Avery ET, Heffernan J, Peciña S, Mickey BJ, Zubieta JK (2017): Striatal dopamine D2/3 receptor-mediated neurotransmission in major depression: Implications for anhedonia, anxiety and treatment response. Eur Neuropsychopharmacol 27: 977–986. 72. 72.Wang J, Korczykowski M, Rao H, Fan Y, Pluta J, Gur RC, et al. (2007): Gender difference in neural response to psychological stress. Soc Cogn Affect Neurosci 2: 227–239. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/scan/nsm018&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17873968&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000253813600008&link_type=ISI) 73. 73.Diekhof EK, Richter A, Brodmann K, Gruber O (2021): Dopamine multilocus genetic profiles predict sex differences in reactivity of the human reward system. Brain Struct Funct 226: 1099–1114. 74. 74.Gray AL, Hyde TM, Deep-Soboslay A, Kleinman JE, Sodhi MS (2015): Sex differences in glutamate receptor gene expression in major depression and suicide. Mol Psychiatry 20: 1057–1068. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/mp.2015.91&link_type=DOI) 75. 75.Purgert CA, Izumi Y, Jong YJI, Kumar V, Zorumski CF, O’Malley KL (2014): Intracellular mGluR5 can mediate synaptic plasticity in the hippocampus. J Neurosci 34: 4589–4598. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Njoiam5ldXJvIjtzOjU6InJlc2lkIjtzOjEwOiIzNC8xMy80NTg5IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDMvMzEvMjAyMi4wMy4zMC4yMjI3MzIwMS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 76. 76.Shin S, Kwon O, Kang JI, Kwon S, Oh S, Choi J, et al. (2015): mGluR5 in the nucleus accumbens is critical for promoting resilience to chronic stress. Nat Neurosci 18: 1017– 1024. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nn.4028&link_type=DOI) 77. 77.Wieroñska JM, Brañski P, Szewczyk B, Paucha A, Papp M, Gruca P, et al. (2001): PRELIMINARY COMMUNICATION CHANGES IN THE EXPRESSION OF METABOTROPIC GLUTAMATE RECEPTOR 5 (mGluR5) IN THE RAT HIPPOCAMPUS IN AN ANIMAL MODEL OF DEPRESSION. Polish J Pharmacol Pol J Pharmacol 53: 659–662. 78. 78.Kovacevic T, Skelin I, Minuzzi L, Rosa-Neto P, Diksic M (2012): Reduced metabotropic glutamate receptor 5 in the Flinders Sensitive Line of rats, an animal model of depression: an autoradiographic study. Brain Res Bull 87: 406–412. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.brainresbull.2012.01.010&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22310150&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000302585500006&link_type=ISI) 79. 79.Smialowska M, Szewczyk B, Branski P, Wieronska JM, Palucha A, Bajkowska M, Pilc A (2002): Effect of chronic imipramine or electroconvulsive shock on the expression of mGluR1a and mGluR5a immunoreactivity in rat brain hippocampus. Neuropharmacology 42: 1016–1023. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0028-3908(02)00062-X&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12128002&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 80. 80.Deschwanden A, Karolewicz B, Feyissa AM, Treyer V, Ametamey SM, Johayem A, et al. (2011): Reduced metabotropic glutamate receptor 5 density in major depression determined by [(11)C]ABP688 PET and postmortem study. Am J Psychiatry 168: 727–734. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1176/appi.ajp.2011.09111607&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21498461&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000292296200013&link_type=ISI) 81. 81.Esterlis I, DellaGioia N, Pietrzak RH, Matuskey D, Nabulsi N, Abdallah CG, et al. (2018): Ketamine-induced reduction in mGluR5 availability is associated with an antidepressant response: an [11 C]ABP688 and PET imaging study in depression. Mol Psychiatry 23: 824–832. 82. 82.Südhof TC (2017): Synaptic Neurexin Complexes: A Molecular Code for the Logic of Neural Circuits. Cell 171: 745–769. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2017.10.024&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29100073&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 83. 83.Hu Z, Xiao X, Zhang Z, Li M (2019): Genetic insights and neurobiological implications from NRXN1 in neuropsychiatric disorders. Mol Psychiatry 24: 1400–1414. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41380-019-0438-9&link_type=DOI) 84. 84.Rucker JJH, Breen G, Pinto D, Pedroso I, Lewis CM, Cohen-Woods S, et al. (2013): Genome-wide association analysis of copy number variation in recurrent depressive disorder. Mol Psychiatry 18: 183–189. 85. 85.Tansey KE, Guipponi M, Hu X, Domenici E, Lewis G, Malafosse A, et al. (2013): Contribution of common genetic variants to antidepressant response. Biol Psychiatry 73: 679–682. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.biopsych.2012.10.030&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23237317&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000316685400014&link_type=ISI) 86. 86.Fabbri C, Tansey KE, Perlis RH, Hauser J, Henigsberg N, Maier W, et al. (2018): Effect of cytochrome CYP2C19 metabolizing activity on antidepressant response and side effects: Meta-analysis of data from genome-wide association studies. Eur Neuropsychopharmacol 28: 945–954. 87. 87.Trabzuni D, Ramasamy A, Imran S, Walker R, Smith C, Weale ME, et al. (2013): Widespread sex differences in gene expression and splicing in the adult human brain. Nat Commun 4. [https://doi.org/10.1038/NCOMMS3771](https://doi.org/10.1038/NCOMMS3771) 88. 88.Hoffman GE, Ma Y, Montgomery KS, Bendl J, Jaiswal MK, Kozlenkov A, et al. (2022): Sex Differences in the Human Brain Transcriptome of Cases With Schizophrenia. Biol Psychiatry 91: 92–101. 89. 89.Hart MP, Hobert O (2018): Neurexin controls plasticity of a mature, sexually dimorphic neuron. Nature 553: 165–170. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature25192&link_type=DOI) 90. 90.O’dushlaine C, Rossin L, Lee PH, Duncan L, Parikshak NN, Newhouse S, et al. (2015): Psychiatric genome-wide association study analyses implicate neuronal, immune and histone pathways. Nat Neurosci 18: 199–209. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nn.3922&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25599223&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 91. 91.Bagot RC, Labonté B, Peña CJ, Nestler EJ (2014): Epigenetic signaling in psychiatric disorders: stress and depression. Dialogues Clin Neurosci 16: 281–295. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25364280&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 92. 92.Peña CJ, Nestler EJ (2018): Progress in Epigenetics of Depression. Prog Mol Biol Transl Sci 157: 41–66. 93. 93.Penner-Goeke S, Binder EB (2019): Epigenetics and depression. Dialogues Clin Neurosci 21: 397–405. 94. 94.Maze I, Noh KM, Soshnev AA, Allis CD (2014): Every amino acid matters: essential contributions of histone variants to mammalian development and disease. Nat Rev Genet 15: 259–271. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nrg3673&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24614311&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 95. 95.Michod D, Bartesaghi S, Khelifi A, Bellodi C, Berliocchi L, Nicotera P, Salomoni P (2012): Calcium-dependent dephosphorylation of the histone chaperone DAXX regulates H3.3 loading and transcription upon neuronal activation. Neuron 74: 122–135. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.neuron.2012.02.021&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22500635&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000302893800014&link_type=ISI) 96. 96.Maze I, Wenderski W, Noh KM, Bagot RC, Tzavaras N, Purushothaman I, et al. (2015): Critical Role of Histone Turnover in Neuronal Transcription and Plasticity. Neuron 87: 77– 94. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.neuron.2015.06.014&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26139371&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 97. 97.Hodes GE, Walker DM, Labonté B, Nestler EJ, Russo SJ (2017): Understanding the epigenetic basis of sex differences in depression. J Neurosci Res 95: 692–702. 98. 98.Miller AH, Raison CL (2016): The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat Rev Immunol 16: 22–34. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nri.2015.5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26711676&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 99. 99.Musselman DL, Evans DL, Nemeroff CB (1998): The relationship of depression to cardiovascular disease: epidemiology, biology, and treatment. Arch Gen Psychiatry 55: 580–592. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/archpsyc.55.7.580&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9672048&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000074737000001&link_type=ISI) 100.100.Anderson RJ, Freedland KE, Clouse RE, Lustman PJ (2001): The prevalence of comorbid depression in adults with diabetes: a meta-analysis. Diabetes Care 24: 1069–1078. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NzoiZGlhY2FyZSI7czo1OiJyZXNpZCI7czo5OiIyNC82LzEwNjkiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMi8wMy8zMS8yMDIyLjAzLjMwLjIyMjczMjAxLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 101.101.Naqvi TZ, Naqvi SSA, Merz CNB (2005, May): Gender differences in the link between depression and cardiovascular disease. Psychosomatic Medicine, vol. 67. Psychosom Med. [https://doi.org/10.1097/01.psy.0000164013.55453.05](https://doi.org/10.1097/01.psy.0000164013.55453.05) 102.102.Heo M, Pietrobelli A, Fontaine KR, Sirey JA, Faith MS (2006): Depressive mood and obesity in US adults: Comparison and moderation by sex, age, and race. Int J Obes 30: 513– 519. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/sj.ijo.0803122&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16302017&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000235645900015&link_type=ISI) 103.103.Möller-Leimkühler AM (2007): Gender differences in cardiovascular disease and comorbid depression. Dialogues Clin Neurosci 9: 71–83. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17506227&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) 104.104.Goldstein JM, Holsen L, Huang G, Hammond BD, James-Todd T, Cherkerzian S, et al. (2016): Prenatal stress-immune programming of sex differences in comorbidity of depression and obesity/metabolic syndrome. Dialogues Clin Neurosci 18: 425–436. 105.105.Marcus SM, Kerber KB, Rush AJ, Wisniewski SR, Nierenberg A, Balasubramani GK, et al. (2008): Sex differences in depression symptoms in treatment-seeking adults: confirmatory analyses from the Sequenced Treatment Alternatives to Relieve Depression study. Compr Psychiatry 49: 238–246. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.comppsych.2007.06.012&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18396182&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000255168600002&link_type=ISI) 106.106.Wang GJ, Volkow ND, Logan J, Pappas NR, Wong CT, Zhu W, et al. (2001): Brain dopamine and obesity. Lancet (London, England) 357: 354–357. 107.107.Hyde CL, Nagle MW, Tian C, Chen X, Paciga SA, Wendland JR, et al. (2016): Identification of 15 genetic loci associated with risk of major depression in individuals of European descent. Nat Genet 48: 1031–1036. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.3623&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27479909&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F03%2F31%2F2022.03.30.22273201.atom)