Next Article in Journal
RSK2-Mediated ELK3 Activation Enhances Cell Transformation and Breast Cancer Cell Growth by Regulation of c-fos Promoter Activity
Previous Article in Journal
Individual and Combined Impact of Oxygen and Oxygen Transporter Supplementation during Kidney Machine Preservation in a Porcine Preclinical Kidney Transplantation Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Systems Approach to Identify Common Genes and Pathways Associated with Response to Selective Serotonin Reuptake Inhibitors and Major Depression Risk

1
Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Delhi 110007, India
2
Department of Psychiatry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
3
Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
4
Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB) Campus, New Delhi 110007, India
5
Department of Bioinformatics, Delhi Technological University, Shahbad Daulatpur, Main Bawana Road, Delhi 110042, India
6
Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA
7
Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2019, 20(8), 1993; https://doi.org/10.3390/ijms20081993
Submission received: 9 March 2019 / Revised: 17 April 2019 / Accepted: 20 April 2019 / Published: 23 April 2019
(This article belongs to the Section Molecular Pharmacology)

Abstract

:
Despite numerous studies on major depressive disorder (MDD) susceptibility, the precise underlying molecular mechanism has not been elucidated which restricts the development of etiology-based disease-modifying drug. Major depressive disorder treatment is still symptomatic and is the leading cause of (~30%) failure of the current antidepressant therapy. Here we comprehended the probable genes and pathways commonly associated with antidepressant response and MDD. A systematic review was conducted, and candidate genes/pathways associated with antidepressant response and MDD were identified using an integrative genetics approach. Initially, single nucleotide polymorphisms (SNPs)/genes found to be significantly associated with antidepressant response were systematically reviewed and retrieved from the candidate studies and genome-wide association studies (GWAS). Also, significant variations concerning MDD susceptibility were extracted from GWAS only. We found 245 (Set A) and 800 (Set B) significantly associated genes with antidepressant response and MDD, respectively. Further, gene set enrichment analysis revealed the top five co-occurring molecular pathways (p ≤ 0.05) among the two sets of genes: Cushing syndrome, Axon guidance, cAMP signaling pathway, Insulin secretion, and Glutamatergic synapse, wherein all show a very close relation to synaptic plasticity. Integrative analyses of candidate gene and genome-wide association studies would enable us to investigate the putative targets for the development of disease etiology-based antidepressant that might be more promising than current ones.

1. Introduction

Major depressive disorder (MDD) is the third largest cause of burden of disease and it is responsible for almost 80% of psychiatric hospitalizations. According to recently conducted world mental health surveys, MDD is experienced by 10–15% people in their lifetime [1] and it can lead to high incidence of suicide. Over 800,000 lives are lost yearly due to suicide, which translates to 3000 suicide deaths every day [2]. It is almost more than half a century now since the first antidepressant drug was discovered, starting from non-specific monoamine oxidase inhibitors (MAO-I) and tricyclic antidepressants (TCA) to target specific selective serotonin reuptake inhibitors (SSRIs). Among them, SSRIs have proven to be the most effective drugs to date, yet approximately 30–40% of depressive patients do not or partially respond to the therapy whereas 60–75% fail to achieve complete remission [3]. This may be attributed to the poor understanding of MDD pathophysiology and lack of etiology-based drugs. Candidate gene-based studies and GWAS have shown that the clinical heterogeneity in therapeutic outcome is also influenced by a variety of genetic (single nucleotide polymorphisms, SNP; copy number variations, CNV; insertions, I; deletions, D etc.), pathophysiological and environmental factors [4,5].
With the advent of high throughput technologies it became possible to generate numerous genomic datasets to identify genetic markers associated with complex disorders and predict drug response. However, these datasets are not consistent enough to turn these findings into clinical practice. The inconsistency may be attributed to the difference in ethnicity of the studied population, underpowered study design, and various confounding factors like age of onset, the severity of disease, gender, etc. Therefore, there is an impending need for integrating these datasets so that we can have a more advance understanding of disease etiology as well as drug response. In pursuit of a better understanding of the problem, several integrated approaches have been put forward for studying the interactions between disease-associated genes and proteins [6]. Network and pathway analysis of candidate genes involved in MDD has provided important information about gene interaction and regulation in MDD [7]. A very recent study has explored the common link for pathogenesis between MDD and glioblastoma using the transcriptomics convergence method, thus providing a new approach to analyze the available huge genetic datasets [8].
With the genetic data on SSRI response and MDD available across diverse populations, a systematic review will help us to better interpret (or curate) the findings of these studies. Therefore, in this study, we first systematically reviewed the literature concerning the genetics of SSRI response (studies on responder versus non-responder patients on SSRIs) and MDD (studies on MDD cases versus healthy controls). Further, we used an integrative genetics approach and retrieved common genes and pathways involved in SSRI response and MDD manifestation to find out potential drug targets for etiology-specific antidepressants development. We identified 29 overlapping genes from a systematic literature search concerning SSRI response and MDD development. Finally, we performed functional enrichment analysis using the WEB-based GEne SeT AnaLysis Toolkit (WebGestalt) [9] (http://webgestalt.org/) to identify the top pathways that are inter-linked between antidepressants response outcome as well as MDD, on the molecular basis. Hence, by integrating genes associated with disease and drug response studies we found a list of genes and pathways which can be validated and might be novel molecular targets for etiology-based antidepressant development.

2. Results

A systematic review of candidate gene studies for SSRI response and systematic literature search of GWAS for SSRI response and MDD both revealed 245 and 800 genes to be significantly associated with SSRI response and MDD pathophysiology, respectively. Using these sets of genes, we retrieved commonly associated genes, pathways, and gene ontology (GO) terms, between SSRI response and MDD.

2.1. Systematic Search and Study Selection of Antidepressant Response Candidate Gene Studies

Systematic search strategy for identifying SSRI response associated genes extracted 8936 candidate gene studies (4268 from MEDLINE and 4668 from Web of Science), which were further reduced to 441 unique articles of relevance after the title and abstract screening. Among these excluded articles, 2529 were duplicate articles between MEDLINE and Web of Science. Further, 2144 non-human, 1800 co-morbid, 580 non-response, 1241 non-SSRIs studies, and 201 other articles including non-genetic studies, review articles, genome-wide studies, editorials, and letters were also removed. Remaining 441 articles were then searched for their full text and 351 articles were again excluded from the study, as they did not meet the inclusion criteria (Figure 1), leading to a final of 90 articles for data extraction and processing.

2.2. Data Extraction from Candidate Gene Studies of SSRI Response

All the 186 significantly associated SNPs (p-value ≤ 0.05) from the 90 selected articles are summarized in Table 1 (complete table in Table S1 online). Sample size along with their phenotype and drugs studied in each of the articles are also detailed in the same. In some research studies, authors had performed two types of analysis, one, responder versus non-responder and second, remitter versus non-remitter; here, we have tabulated all the significant SNPs reported in either kind of analysis.

2.3. Data Extraction from GWAS of SSRI Response and MDD Susceptibility

Genes related to SSRI response and MDD etiology were also retrieved from the genome-wide literature. We retrieved 144 articles in total, comprising GWAS with keywords “Depression” or “Depressive Disorder”. Among them, seven GWAS [67,68,69,70,71,72,73] were related to SSRI response (Table 2) and eight GWAS [74,75,76,77,78,79,80,81] were related to MDD etiology (Table 3). In total, 423 and 3884 SNPs with p-value ≤ 0.0001 were found to be associated with SSRI response and MDD, respectively (Tables S3 and S4 online).

2.4. Data Processing and Genetic Co-Occurrence of SSRI Response and MDD

A total of 186 significant SNPs from a systematic review of SSRI candidate gene studies and 423 SNPs from GWAS of SSRI response were merged to form the first data, Set A, representing SSRI response. Additionally, 3884 SNPs from GWAS of MDD formed the second data, Set B, relating to MDD pathophysiology. Beside this, both sets of SNPs were enriched with probable genetic variations found to be in LD (r2 = 1) using the HaploReg v4.1 online tool. Now enriched Set A and Set B comprise 718 and 4769 SNPs, respectively. Enriched Set A and B thus obtained were mapped to genes using the SCAN and Ensembl VEP online tools. After excluding unmapped genes, pseudogenes, orfs, miRNAs, and uncharacterized genes, these annotated genes were assigned their respective HGNC IDs. This obtained a pool of 245 and 800 genes associated with SSRI response and MDD etiology, respectively (Table S5 online). Furthermore, we identified the overlap between SSRI-response genes (Set A) with that of MDD genes (Set B) and found 29 common genes. Since all the MDD associated genes were extracted from GWAS articles, i.e., an unbiased source, therefore, all of the 29 overlapped genes can be considered as unbiased molecular players co-occurring between SSRI response and MDD susceptibility (Figure 2a).

2.5. Functional Enrichment of SSRI Response and MDD Gene Pool

The WebGestalt web-tool was used to further enrich the genes obtained from systematic literature mining. The GO term and pathway enrichment analysis were performed keeping default GO slim classification, 0.05 significance level, and the minimum number of genes in a category was set at 3. The significance value was adjusted by the false discovery rate (FDR) analysis using the Benjamini–Hochberg (BH) procedure (Tables S7 and S8 online). Set A of the SSRI-response genes were significantly enriched in 298 GO terms, involving 228 GO biological process terms, 15 GO molecular function terms, and 55 GO cellular component terms along with 24 KEGG pathway terms. Set B of the MDD etiology genes was significantly enriched in 94 GO terms, involving 7 GO biological process terms, 1 GO molecular function terms, and 86 GO cellular component terms along with 32 KEGG pathway terms. On overlapping, these results showed nine common KEGG pathways (Table S6 online) and 43 common GO terms as shown in Figure 2b,c. This overlap directs a common molecular ground between the mechanism behind antidepressant response and MDD pathophysiology. Since the biased data source (candidate study available literature) in Set A was overlapped with Set B which has an unbiased source (GWAS), common enriched biological pathways must be unbiased and can be used as putative future novel drug targets for MDD. Moreover, functional enrichment of overlapping gene set, i.e., Set C pinpoints the significant pathways involved in both antidepressant response and MDD etiology. Altogether, 5 out of 21 enriched pathways, namely, Cushing syndrome (p = 0.005), Axon guidance (p = 0.007), cAMP signaling pathway (p = 0.010), Insulin secretion (p = 0.016), and Glutamatergic synapse (p = 0.028) were coinciding when compared to nine commonly enriched pathways among Set A and B (Figure 3). Thus, these represents the most promising pathways, which on in- vitro or in vivo validation, may prove to be new drug targets for etiology-based antidepressant therapy. Thereafter, we evaluated genes commonly present in the five enriched pathways, for their possible roles or interactions in both antidepressant response and MDD pathophysiology, from both the data Sets A and B. Thus, the top recognized genes were: KCNK2, CACNA1C, CAMK2D, GSK3B, APC, CRHR2, CRHR1, PDE11A, ADCY9, CREB5, ADCY3, GNAI3, DVL3, ADCY2, TCF7L1, RAP1B, WNT1, CACNA1I (Cushing syndrome); SLIT3, CAMK2D, GSK3B, RND1, SEMA5A, SEMA6D, GNAI3, ABLIM1, SRGAP3, EFNA5, SEMA3E, NTNG1, BMPR2, EPHB6, ROBO1, BOC, ROBO2 (Axon guidance); GRIA4, CACNA1C, CAMK2D, HTR1A, ADRB1, DRD2, GRIA3, BDNF, HTR1B, TSHR, ATP1B2, ADCY9, ABCC4, CREB5, ADCY3, GNAI3, ADCY2, RAP1B, ACOX3, PLD2 (cAMP signaling pathway); CACNA1C, CAMK2D, CHRM3, ATP1B2, ADCY9, CREB5, ADCY3, ADCY2, PCLO (Insulin secretion); GRIA4, GRM7, CACNA1C, GRIK2, GRIA3, SLC17A7, ADCY9, ADCY3, KCNJ3, GNAI3, ADCY2, PLD2 (Glutamatergic synapse).

3. Discussion

Major depressive disorder is a progressive brain disease with one of the leading cause of disability-adjusted life years affecting approximately 10–15% of the population worldwide [82]. Various treatment regimens are present for MDD, but SSRI pharmacotherapy is being commonly used and often recommended as a first-line treatment option in moderate-to-severe depression because of their higher efficacy and lower side effects compared to other antidepressants. Majorly, all the antidepressants available up to date are aimed at symptomatic management rather than complete cure owing to poorly understood MDD disease etiology. Since the response to antidepressant treatment varies markedly between individuals due to considerable clinical heterogeneity, therefore, the role of genetic predictors of antidepressant response and MDD disease per se are of utmost importance for the development of better treatment regimen which would further improve clinical management of MDD.
In this study, we used an integrative genetics approach to unbiasedly elucidate the possible association between antidepressant therapy and MDD etiology. Initially, an extensive literature search was done for identification of genetic variants involved in antidepressant response and disease etiology, followed by functional gene set enrichment, and thereby distinguishing the GO terms and molecular pathways involved in both antidepressant response and MDD susceptibility. For the first time in this uniquely designed study, to the best of our knowledge, has incorporated the amalgam of two exhaustive datasets of disease, concerning its pathophysiology and drug response genes, in search of precise drug targets to accelerate future drug development with minimal side effects.
For quality assessment of the studies included in this systematic review, based on the modified criteria used by Guin D et al. [83], the present study has incorporated the most comprehensive quality assessment scoring of research articles for screening SSRI response articles. Our systematic review has retrieved a total of 55 genes from the 90 research articles which were found to be involved in various synaptic transmission and neuronal development pathways. For instance, rs1083801 (GRM7, a glutamate receptor gene), was found to be most significantly associated with the early response with SSRIs [54]. The TPH2 gene and its variant, rs4760815, has been reported as probable risk factors for the development of MDD and also associated with SSRI response [84]. In addition, other genetic variants from TPH2, (rs11179027 and rs17110532); glutamate receptor ionotropic, kainate 2 gene, GRIK2 (rs543196), glutamic acid decarboxylase gene, GAD1 (rs3828275) and SLC6A4 (rs2066713) were also reported to be strongly associated with SSRI response [15]. SERTPR often regarded as 5-HTTLPR is a serotonin transporter gene promoter polymorphism which is been exhaustively studied in both remission rate and response rate of antidepressants [85]. A review and meta-analysis study by Kato and Serretti [86] has extensively studied and reported a significant association between the 5-HTTLPR variant and better response to antidepressants. Zill et al. [87] have identified functional polymorphism in the β1 adrenergic receptor 1165G>C (rs1801253), which was found to be involved in conferring the faster response towards antidepressant treatment, but did not influences the depressive phenotype. Furthermore, HTR2A, a serotonergic receptor gene variant rs7997012 is found to be associated with SSRI response as well as remission [56,59,88,89]. However, Illi A et al. [90] and Kishi T et al. [28] have reported a negative association of rs7997012 with SSRI response or remission. Thus, to overcome such inconsistencies across several research studies, we need an innovative approach to eliminate the biases and limitation of candidate gene studies [91]. Hence in the present study, we have opted for “candidate gene studies and GWAS overlapping” approach to overcome the lack-of-harmony among candidate gene studies. As a result, the systematic review of candidate gene studies and GWAS was merged to ascertain the unbiased genes and molecular pathways involved in both antidepressant response and MDD etiology.
After merging findings from the systematic review and GWAS studies, all the genetic variants were grouped into Set A and Set B followed by their LD enrichment, gene annotation, and functional enrichment analysis, to reveal multiple relevant pathways among antidepressant response and MDD etiology (Figure 1). Furthermore, 56 KEGG pathways (24 from Set A and 32 from Set B) were identified with 9 commonly enriched pathways, namely Cushing syndrome, Retrograde endocannabinoid signaling, Axon guidance, cAMP signaling pathway, Glutamatergic synapse, Insulin secretion, Gap junction, Gastric acid secretion, and Salivary secretion, from Set A and Set B. In the view of MDD etiology, the most interesting molecular pathways are Axon guidance, Glutamatergic synapse, and Cushing syndrome. These 9 co-occurring pathways were further overlapped with the molecular pathways enriched from the 29 common genes implicated in antidepressant response and MDD etiology. This results in five overlapping molecular pathways, namely, Cushing syndrome, Axon guidance, cAMP signaling pathway, Insulin secretion, and Glutamatergic synapse. As MDD is a complex disorder, multiple pathways are expected to be involved in its etiology and our result is in concordance with the same showing the interplay of stress, neurodevelopmental, synaptic plasticity, and metabolic physiology in MDD etiology and antidepressant response.
Cushing syndrome, as being the top pathway of the analysis, is a metabolic disorder caused by overproduction of cortisol (glucocorticoid) produced by the adrenal cortex of the adrenal gland in response to glucocorticoids medication or in stressful condition. The adrenal gland is a part of stress-responsive hypothalamic pituitary adrenal (HPA) axis which consists of stimulating and feedback mechanism involving the hypothalamus, pituitary, and adrenal gland, and thus regulates the production of glucocorticoids. The HPA axis dysregulation has been implicated in the pathophysiology of many neuropsychiatry traits including MDD [92,93,94]. Genes associated with the homeostatic response to environmental stressors particularly lies in the HPA axis [95].
Corticotrophin-releasing hormone (CRH), released by the hypothalamus, stimulates the release of adrenocorticotropic hormone (ACTH) from pituitary which in turns regulate the levels of cortisol secreted from the adrenal gland. Hence CRH and its downstream effects are of prime importance which is controlled by cortisol level through feedback mechanism and two CRH receptors i.e., CRHR1 and CRHR2 [96]. Alteration in CRHR1 and CRHR2 activity may lead to HPA dysregulation and can be a major risk factor for depressive symptoms. Bradley et al. [97] demonstrated the role of genetic variants in CRHR1 as moderators of the effects of child abuse on adult depressive symptoms in two independent populations which was further confirmed by a replication study which reported the association of TAT (rs7209436, rs110402, and rs242924) haplotype in CRHR1 in predicting the adult depression [98,99]. Woody et al. [100] also supported the hypothesis of protective CRHR1 haplotype (TAT) as their result suggested the development of brooding among children without the protective CRHR1 haplotype. Moreover, animal studies also indicate the prenatal glucocorticoid exposure can cause epigenetic instability in CRHR1 promoter which further increases the risk for the affective disorder in offspring’s across two generations [101]. Epistatic interaction between AVPR1b, CRHR1, and BDNF genes has also been reported to be involved in susceptibility to MDD [102,103]. Ressler et al. [104] reported the involvement of interaction of 5-HTTLPR S allele with CRHR1 haplotype in predicting adult depression in individuals with child abuse. The SNPs rs110402, rs242924 rs3779250, rs7209436, and rs173365 from CRHR1 and CRHR2 genes were reported to be positively associated with MDD in the Japanese population [105]. However, a CRHR2-based 10 SNP study showed no significant allelic or genotypic differences among unipolar patients and matched healthy controls [106].
In addition, there are evidences for an antidepressant response via glucocorticoid receptors (GRs) where it has been demonstrated that sertraline increases human hippocampal neurogenesis via a GR-dependent mechanism [107]. A recent study has shown the significant association of rs41423247 polymorphism with fluoxetine response in depressed patients [10]. Another study of haplotype-tag SNPs (rs1876828, rs242939 and rs242941) in CRHR1 shown the significantly improved response to antidepressants among highly anxious patients homozygous for the GAG haplotype, suggesting the possible role of CRHR1 and other stress-inflammatory pathway genes in variable antidepressant response [108,109].
Increasing evidence suggests the dysfunction of glutamatergic neurotransmission impairs neuroplasticity in the brain and this leads to major depressive disorder [110,111]. Many novel targets in relation to this pathway are evidenced for causing the depression phenotype. In the past decade, NMDA has gained the utmost attention with respect to the biology of depression and also serves as a potential target for drug development and treatment. As under pathological condition, elevated levels of glutamate resulted in the impairment of synaptic plasticity and even excitotoxicity. Alternate depression hypothesis includes exposure to stress inculpates the release of the high level of stress hormone i.e., cortisol from the adrenal gland. Correspondingly, adequate literature demonstrates that stress and glucocorticoids are responsible for alteration in the expression and activity of vesicular proteins of the neurotransmission of glutamate [112]. Emerging evidences from post-mortem studies has reported the dysregulation of genes and increased glutamate levels, thus highlighting the role of altered glutamate signaling in MDD patients [113]. Decrease expression of presynaptic genes in MDD patients such as SYN3, SNAP25, essential for vesicular release of neurotransmitters have been reported. Likewise, a significant downregulation of postsynaptic genes was also reported in the dentate gyrus (DG) and CA1 of MDD patients such as AMPA receptors, specifically GLU1 and GLU3 [114]. In the same note, several NMDA gene variations have been reported to have an association with an abnormality in NMDA receptors. One such study had shown significant association of GRIN1 (rs4880213) with depression. Further studies revealed that variations in the GRIN2B were associated with schizophrenia, psychiatric disorders, and brain plasticity [115]. Similarly, downregulation of NMDA receptor subunits GRIN1A and GRIN2B, as well as PSD-95 have been demonstrated in the anterior prefrontal cortex of MDD subjects [116]. Likewise, metabotropic glutamate receptor 7 (GRM7) encodes the protein mGluR7 mediates the glutamate neurotransmission, found to be involved in the development of the major depressive disorder. Li W et al. [117] studied the association of genetic variation of the GRM7 gene with MDD and schizophrenia and reported the significant association of GRM7 gene variation (rs779706) with MDD and (rs2229902 and rs9870680) with schizophrenia in the Han Chinese population. Thus, it is hypothesized that dysfunction of ionotropic and metabotropic receptors are associated with the depressed phenotype. However, in a resequencing study, none of the GRM7 variants had shown the significance level with MDD in the Dutch cohort [118].
Also, a number of studies support the association between the antidepressant response and GRIK4 in MDD patients [89,119,120]. Genes encoded ionotropic glutamate receptors were studied with respect to citalopram treatment and demonstrated a significant association of two SNPs (rs4825476 and rs2518224) located within GRIA3 and GRIK2, respectively, with the treatment-emergent suicidal ideation in MDD patients [121]. Moreover, in order to explore the potential targets for treatment-resistant depressive patients, emerging evidence from clinical trials supported the use of glutamate receptor modulators for the treatment of depression and these include non-competitive NMDA receptor antagonists such as ketamine, subunit (NR2B)-specific NMDA receptor antagonists, NMDA receptor glycine-site partial agonists and metabotropic glutamate receptor (mGluR) modulators [122]. Therefore, glutamate pathways and its associated receptors are important and further insights and detailed understanding could help us to target the accurate site for future drug development.
Neuronal circuit formation involves a molecular cascade of events such as axon guidance, where axons move to their target cells in a complex, constantly changing the environment. It has been speculated that change in this gene-environment interaction may lead to alteration in axon guidance followed by its implication in neuropsychiatric disorders. Furthermore, a meta-analysis by S Jovanova O et al. [123], has reported three methylated sites associated with depressive symptoms and were also found to be involved in axon guidance as a pathway in major depression. Interestingly, research articles have also shown that miRNAs implicated in axon guidance are involved in differential antidepressant response where miRNAs namely miR-146a-5p, miR-146b-5p, miR-221-3p, miR-24-3p, miR-26a-5p are known to be involved in axon guidance and were also associated with antidepressant response in MDD patients [124,125]. Our current finding of Slit guidance ligand 3 gene, SLIT3 in antidepressant and MDD gene set is in consensus with findings from Glessner JT et al. [126], where they have performed genome-wide copy number variation scan of large cohort of MDD patients and controls and has observed 5q35.1 as the most significant locus harboring the SLIT3 gene which is integral to repulsive axon guidance. Thus synaptic plasticity mediated via axon guidance is a topic of new research and can further be studied for better antidepressant development.
Since the majority of intracellular messenger cascades are regulated either by G protein-coupled receptors (GPCRs) or protein tyrosine kinases (PKAs) and are major activator of various cellular and molecular signaling pathways such as cortisol secretion in the adrenal cortex contributing to Cushing’s syndrome and neurotransmitters such as serotonin and norepinephrine, which are known to mediate the effects of antidepressant treatments modulates secondary messenger cascades via interaction with GPCRs or PKAs [127,128]. Hence, receptor activation induced by ligands such as neurotransmitters confers to cAMP generation via stimulating adenylate cyclase (AC), and their binding to the G-protein subtype leads to activation of PKA, which is an important factor for driving several biological functions either by phosphorylation or dephosphorylation of specific target proteins, undermines the antidepressant actions. Furthermore, the cAMP response element binding protein (CREB), a transcription factor that mediates the actions of the cAMP cascade, is a substrate for PKA, is involved in regulating gene expression, and has the capability to modulate their transcriptional activity, which is important for cellular adaptions during antidepressant administration.
Dowlatshahi et al. [129] have reported low CREB levels in post-mortem temporal cortex of naive major depressive disorder patients as compared to MDD patients treated with antidepressants. Odagaki et al. [130] have even demonstrated the increased immunoreactivities of phosphorylated CREB as well as total CREB levels in the prefrontal cortex of depressed suicide victims and specifically in antidepressant drug-free subjects. Also, with a slight trend for increased levels of PKA-Cα in depressed suicide victims only as compared to healthy controls. As a downstream consequence, the expression of various target genes critical to the organization of neuronal networks and synaptic plasticity, like neurotrophin, brain-derived neurotrophic factor (BDNF), and neuropeptide Y (NPY) is also increased contributing towards antidepressant-mediated changes in structural remodeling, neuronal plasticity and synaptic restructuring [131,132]. Henceforth, evidence from such studies confer the ability of HTR receptors in either stimulating or inhibiting the AC-cAMP-PKA signaling transduction pathway but further invokes solicitation and functional validation of pathway genes in undermining the action of antidepressants, as this transduction pathway is highly regulated by several other factors such as stress, apoptosis, inflammation, and others.
Hence pathway enrichment analysis of “biased and unbiased” merged data is assured to facilitate our understanding of the underlying molecular mechanism of the complex trait of anti-depressant response and major depression as a disease, to circumvent the symptomatic respite and design a definite therapy. Application of this elaborated analytical tactics in translational research concerning complex disorders like MDD would be beneficial after in-vitro and in-vivo validation of the top promising pathways and genes involved in antidepressant response and MDD etiology.

4. Methodology

The complete workflow of the study is represented in Figure 4. We initiated with integrating significantly associated SNPs from candidate genes studies and GWAS concerning SSRI response and from GWAS of MDD etiology. Therefore all the SNPs from candidate gene studies and GWAS of SSRI response were merged in one group and another group consist significant genetic variations from MDD GWAS. Further, they were annotated into genes followed by functional and pathway enrichment analysis.

4.1. A systematic Review of Antidepressant Response Candidate Gene Studies

Among currently available antidepressants, selective serotonin reuptake inhibitors (SSRIs) like escitalopram, sertraline, and fluoxetine are the most commonly prescribed drugs [133] considering their higher efficacy and lower side-effects. Hence in the present study, we have considered “SSRIs” and “antidepressants” interchangeably in this manuscript. A systematic literature search was performed in accordance with PRISMA guidelines [134]. The MEDLINE and Web of Science databases were searched using Medical Subject heading (MeSH) terms “selective serotonin reuptake inhibitor”, “SSRI”, “pharmacogenetics”, “pharmacogenomics”, “response”, “treatment outcome”, “SNP”, “variant”, “polymorphism” with AND/OR Boolean operators to extract all the studies evaluating association of SNPs with SSRI treatment outcome in patients with MDD. The search and study selection was carried out independently by three authors (AS, HG, and PS) covering the articles published till 28th February 2018.
The searches were confined to human and English language studies. All the articles that were review, meta-analysis, commentary, editorial, clinical trial, letter, randomized control trial, technical report were excluded. Articles were sorted for their relevance at two stages, first using the title and second using their abstract. At the first stage of title screening, duplicates, reviews, systematic reviews, meta-analysis, non-human, and co-morbid studies were removed. Secondly, the abstracts of all remaining articles were retrieved and screened based on the inclusion and exclusion criteria of the study. Inclusion criteria required genetic association of SSRI response in MDD patients, with age range of 18–75 years, and that response should be inferred based on gold standard severity rating scales like Hamilton Depression Rating Scale (HAM-D) or Montgomery Asberg Depression Rating Scale (MADRS). Whereas studies involving MDD as co-morbidity or MDD patients with other severe medical illness, psychiatric disorder or substance abuse were excluded. Detailed exclusion criteria are given in Figure 1.

4.2. Data Extraction and Quality Assessment of Antidepressant Response Candidate Gene Articles

Data, from selected full-text articles, were extracted by AS, HG, and PS and checked by DG and RK. Ethnicity, used response criteria, sample size, genes with information of genetic variant, related genotypic or allelic frequency and respective reported p-value, drug, dose, and follow-up period were included in the data collection table. Ethnicity was classified as reported in the respective article, else the country in which the study was conducted, assumed to be the individual’s ethnicity. A cut-off p-value ≤ 0.05 was used for extracting SNPs from the selected articles. Methodological quality of each article was assessed by two independent reviewers (AS and HG) using modified criteria for quality assessment, as used by Guin D et al. [83]. The quality assessment was scored on 8 parameters (Table S2 online), with a positive score awarded for each detail present in the study, the lack of detail was described as 0. Conflicting scores were reached to a consensus upon discussing with DG and RK. If the score obtained was 7 or higher, the study was considered as high quality.

4.3. A Systematic Literature Search of GWAS of SSRI Response and MDD Susceptibility

Data were also extracted from GWAS, using keywords “Depression” or “Depressive Disorder”, correlating genetic variability with SSRI response in patients with MDD, and distinguishing MDD patients from healthy controls. To maintain homogeneity in studies, only those articles were selected where researchers have opted peripheral blood as their source of DNA extraction. In the case of disease vs. control GWAS, the diagnosis has been made by a psychiatrist/clinical psychologist, otherwise, a study has been excluded from the pool. Similarly, in GWAS correlating SNPs with SSRI response, if the response were adjudicated using standard depression severity rating scales the study was included, else excluded. Studies considering meta-analysis were also excluded. A cut-off p-value ≤ 0.0001, was used for extracting SNPs reported in respective GWAS.

4.4. Data Processing of Candidate Gene Studies for SSRI Response and GWAS of SSRI Response and MDD Susceptibility

SNPs from candidate and GWAS studies were retrieved and categorized into two data sets, Set A containing SNPs associated with SSRI response (data from both systematic review and GWAS of SSRI response), and Set B comprising SNPs associated with MDD susceptibility (data only from GWAS of MDD disease). Here, we were investigating the targets for etiology based antidepressant development using genomic integration of disease and drug response, one dataset needed to be unbiased and hence we decided not to include candidate gene studies for MDD. Thus, Set A can be considered to be a biased data set as it contains SNPs from candidate gene studies as well as from GWAS, whereas Set B is genetically unbiased data set as it contains data from GWAS only. As interpatient genetic variability can modulate SSRI clinical response and MDD etiology, therefore, in order to widen the genetic region to find out the biological relevance of the associated SNPs, we have incorporated all the probable genetic variations found to be in linkage disequilibrium (LD) with previously extracted SNPs of Set A and Set B. Furthermore, pairwise LD among the extracted SNPs of set A and B were performed using HaploReg v4.1 which uses mammalian conservation algorithm from GERP and SiPhy-omega with LD threshold (r2) = 1, corresponds to 100% LD [135]. SNPs that are found to be in 100% LD in all the four major population i.e., African, American, Asian, and European were included in the respective SNP sets. In addition, SCAN (SNP and CNV Annotation Database) [136] (http://www.scandb.org/newinterface/about.html) and VEP (Variant Effect Predictor —Ensembl) [137] (https://asia.ensembl.org/info/docs/tools/vep/index.html) online tools were employed for gene annotation for all the identified SNPs, which were extracted and enriched from LD. The SCAN utilizes two ways for SNP annotation, i.e., relative position based and eQTLs (expression quantitative trait loci) method. Relative position-based method identifies a gene based on SNP position (intronic, inter-genic, etc.) or an intergenic variant can be annotated to a gene if it is in LD with any other variant present in the gene. An eQTLs-based method annotates a gene whose quantitative expression is altered by input SNP. Whereas VEP annotates an SNP to a gene if it has a functional effect on that gene, transcript, protein sequence or regulatory regions. The SNPs, which remained unmapped, were excluded from further analysis. Genes thus obtained were assigned HGNC (HUGO Gene Nomenclature Committee) (https://www.genenames.org/) IDs manually [138]. The pseudogenes, hypothetical loci, non-coding RNAs, non-protein coding genes, open reading frames (orfs), microRNA (miRNA), and uncharacterized genes were excluded from each dataset. Further, a third data set, Set C, was also framed which contained all the overlapping genes from Set A and Set B. Set C was used to distinguish the most significant pathways from the commonly enriched pathways among Set A and Set B.

4.5. Functional Enrichment Analysis

For each of the three datasets, functional enrichment analysis was conducted using a WEB-based GEne SeT AnaLysis Toolkit (WebGestalt) [9]. Gene Ontology (GO) term enrichment analysis (http://www.geneontology.org/) and pathway enrichment analysis with Kyoto Encyclopedia of Genes and Genomes (KEGG) engine (https://www.genome.jp/kegg/) was performed keeping default GO slim classification, 0.05 significance level and a minimum number of genes in a category was set at 3. For enrichment analysis of each dataset, the significance value was adjusted by the false discovery rate (FDR) analysis using the Benjamini–Hochberg (BH) procedure. The GO terms and molecular pathways overlapping between Set A and Set B were extracted, which further overlaid with the enriched pathways from Set C to identify pathophysiological grounds which can be targeted to develop etiology based antidepressants.

5. Conclusions

Burgeoning literature evidence has so far pointed out the clinical relevance of antidepressant response and MDD etiology individually, but such studies are not consistent enough to manifest these findings into clinical practice. In the present study, we have focused on highlighting the possible confounding factors responsible for antidepressant response and MDD pathophysiology altogether. Thus we have shortlisted significant genes and pathways implicated in the same which can be further utilized as novel molecular targets for the development of more efficacious antidepressant drugs. Application of this elaborated analytical approach in translational research concerning complex disorders like MDD would be beneficial after in-vitro and in-vivo validation.

Supplementary Materials

Supplementary materials can be found at https://www.mdpi.com/1422-0067/20/8/1993/s1.

Author Contributions

A.S. conceived and wrote the manuscript. A.S. and R.K. contributed to the study design. A.S., H.G., and P.S. extracted data from the literature. A.S. did the gene set enrichment analysis shown in the manuscript. In case of disagreement in systematic review or cumulative quality assessment, D.G. and H.K. helped in resolving. A.S., D.G., N.K., and P.S. contributed in writing manuscript. R.K., D.V., H.K., L.S., and J.Y. contributed for critical evaluation of the study and improving the manuscript. M.S. and R.K.C. provided valuable insights for selection of relevant articles from clinical perspectives. R.K. conceived, interpreted, and supervised the study design. All the authors read and approved the final manuscript.

Funding

This research was funded by Indian Council of Medical Research, grant number GAP-0136.

Acknowledgments

We would like to acknowledge the Director, CSIR-IGIB, Anurag Agrawal, for his vision and support. A.S. and N.K. acknowledge CSIR, Government of India for providing their fellowships. P.S. acknowledge UGC, Government of India, and DG acknowledge ICMR and the Government of India for providing their fellowships. We thank the anonymous reviewers for their helpful suggestions in improving the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bromet, E.; Andrade, L.H.; Hwang, I.; Sampson, N.A.; Alonso, J.; de Girolamo, G.; de Graaf, R.; Demyttenaere, K.; Hu, C.; Iwata, N.; et al. Cross-national epidemiology of dsm-iv major depressive episode. BMC Med. 2011, 9, 90. [Google Scholar] [CrossRef] [PubMed]
  2. World Health Organization. Depression and Other Common Mental Disorders; World Health Organization: Geneva, Switzerland, 2017. [Google Scholar]
  3. Amsterdam, J.D.; Hornig-Rohan, M. Treatment algorithms in treatment-resistant depression. Psychiatr. Clin. North Am. 1996, 19, 371–386. [Google Scholar] [CrossRef]
  4. Peyrot, W.J.; Middeldorp, C.M.; Jansen, R.; Smit, J.H.; de Geus, E.J.; Hottenga, J.J.; Willemsen, G.; Vink, J.M.; Virding, S.; Barragan, I.; et al. Strong effects of environmental factors on prevalence and course of major depressive disorder are not moderated by 5-httlpr polymorphisms in a large dutch sample. J. Affect. Disord. 2013, 146, 91–99. [Google Scholar] [CrossRef]
  5. Nabeshima, T.; Kim, H.C. Involvement of genetic and environmental factors in the onset of depression. Exp. Neurobiol. 2013, 22, 235–243. [Google Scholar] [CrossRef] [PubMed]
  6. Lee, S.A.; Tsao, T.T.; Yang, K.C.; Lin, H.; Kuo, Y.L.; Hsu, C.H.; Lee, W.K.; Huang, K.C.; Kao, C.Y. Construction and analysis of the protein-protein interaction networks for schizophrenia, bipolar disorder, and major depression. BMC Bioinform. 2011, 12, S20. [Google Scholar] [CrossRef] [PubMed]
  7. Jia, P.; Kao, C.F.; Kuo, P.H.; Zhao, Z. A comprehensive network and pathway analysis of candidate genes in major depressive disorder. BMC Syst. Biol. 2011, 5, S12. [Google Scholar] [CrossRef] [PubMed]
  8. Xie, Y.; Wang, L.; Xie, Z.; Zeng, C.; Shu, K. Transcriptomics evidence for common pathways in human major depressive disorder and glioblastoma. Int. J. Mol. Sci. 2018, 19, 234. [Google Scholar] [CrossRef]
  9. Wang, J.; Vasaikar, S.; Shi, Z.; Greer, M.; Zhang, B. Webgestalt 2017: A more comprehensive, powerful, flexible and interactive gene set enrichment analysis toolkit. Nucleic Acids Res. 2017, 45, W130–W137. [Google Scholar] [CrossRef]
  10. Nouraei, H.; Firouzabadi, N.; Mandegary, A.; Zomorrodian, K.; Bahramali, E.; Shayesteh, M.R.H.; Ansari, S. Glucocorticoid receptor genetic variants and response to fluoxetine in major depressive disorder. J. Neuropsychiatry Clin. Neurosci. 2018, 30, 45–50. [Google Scholar] [CrossRef] [PubMed]
  11. Firouzabadi, N.; Raeesi, R.; Zomorrodian, K.; Bahramali, E.; Yavarian, I. Beta adrenoceptor polymorphism and clinical response to sertraline in major depressive patients. J. Pharm. Pharm. Sci. 2017, 20, 1–7. [Google Scholar] [CrossRef] [PubMed]
  12. Xu, Z.; Reynolds, G.P.; Yuan, Y.; Shi, Y.; Pu, M.; Zhang, Z. Tph-2 polymorphisms interact with early life stress to influence response to treatment with antidepressant drugs. Int. J. Neuropsychopharmacol. 2016, 19. [Google Scholar] [CrossRef] [PubMed]
  13. Manoharan, A.; Shewade, D.G.; Rajkumar, R.P.; Adithan, S. Serotonin transporter gene (slc6a4) polymorphisms are associated with response to fluoxetine in south indian major depressive disorder patients. Eur. J. Clin. Pharmacol. 2016, 72, 1215–1220. [Google Scholar] [CrossRef]
  14. Paroni, G.; Seripa, D.; Fontana, A.; D’Onofrio, G.; Gravina, C.; Urbano, M.; Addante, F.; Lozupone, M.; Copetti, M.; Pilotto, A.; et al. Klotho gene and selective serotonin reuptake inhibitors: Response to treatment in late-life major depressive disorder. Mol. Neurobiol. 2017, 54, 1340–1351. [Google Scholar] [CrossRef] [PubMed]
  15. Lim, S.W.; Won, H.H.; Kim, H.; Myung, W.; Kim, S.; Kim, K.K.; Carroll, B.J.; Kim, J.W.; Kim, D.K. Genetic prediction of antidepressant drug response and nonresponse in korean patients. PLoS ONE 2014, 9, e107098. [Google Scholar] [CrossRef] [PubMed]
  16. Fukui, N.; Suzuki, Y.; Sugai, T.; Watanabe, J.; Ono, S.; Tsuneyama, N.; Someya, T. Promoter variation in the catechol-o-methyltransferase gene is associated with remission of symptoms during fluvoxamine treatment for major depression. Psychiatry Res. 2014, 218, 353–355. [Google Scholar] [CrossRef]
  17. Li, X.; Yu, T.; Huang, X.; Cao, Y.; Li, X.; Liu, B.; Yang, F.; Li, W.; Zhao, X.; Feng, G.; et al. Slc17a7 gene may be the indicator of selective serotonin reuptake inhibitor treatment response in the chinese han population. J. Clin. Psychopharmacol. 2014, 34, 331–336. [Google Scholar] [CrossRef]
  18. Wang, X.C.; Xu, D.J.; Chen, G.H.; Xia, Q.; Liu, L.N. Association of 2 neurotrophic factor polymorphisms with efficacy of paroxetine in patients with major depressive disorder in a chinese population. Ther. Drug Monit. 2014, 36, 612–617. [Google Scholar] [CrossRef]
  19. Han, K.M.; Chang, H.S.; Choi, I.K.; Ham, B.J.; Lee, M.S. Cyp2d6 p34s polymorphism and outcomes of escitalopram treatment in koreans with major depression. Psychiatry Investig. 2013, 10, 286–293. [Google Scholar] [CrossRef]
  20. Sahraian, S.; Babashams, M.; Reza-Soltani, P.; Najmabadi, H.; Kahrizi, K.; Gorgani, S.H. Serotonin transporter polymorphism (5-httlpr) and citalopram effectiveness in iranian patients with major depressive disorder. Iran J. Psychiatry 2013, 8, 86–91. [Google Scholar]
  21. Liu, Z.; Zhu, F.; Yao, L.; Yang, C.; Xiao, L.; Zhu, J.; Wang, H.; Wang, G.; Liu, W.; Xiao, Z. Pdlim5 gene polymorphisms and short term antidepressant response in chinese major depressive disorders. Int. J. Clin. Exp. Med. 2013, 6, 677–682. [Google Scholar]
  22. Mitjans, M.; Serretti, A.; Fabbri, C.; Gasto, C.; Catalan, R.; Fananas, L.; Arias, B. Screening genetic variability at the cnr1 gene in both major depression etiology and clinical response to citalopram treatment. Psychopharmacology 2013, 227, 509–519. [Google Scholar] [CrossRef] [PubMed]
  23. Myung, W.; Lim, S.W.; Kim, S.; Kim, H.; Chung, J.W.; Seo, M.Y.; Kim, J.W.; Carroll, B.J.; Kim, D.K. Serotonin transporter genotype and function in relation to antidepressant response in koreans. Psychopharmacology 2013, 225, 283–290. [Google Scholar] [CrossRef]
  24. Wang, Y.; Liu, X.; Yu, Y.; Han, Y.; Wei, J.; Collier, D.; Li, T.; Ma, X. The role of single nucleotide polymorphism of d2 dopamine receptor gene on major depressive disorder and response to antidepressant treatment. Psychiatry Res. 2012, 200, 1047–1050. [Google Scholar] [CrossRef]
  25. Yang, Z.; Ma, X.; Wang, Y.; Wang, J.; Xiang, B.; Wu, J.; Deng, W.; Li, M.; Wang, Q.; Li, T. Association of apc and reep5 gene polymorphisms with major depression disorder and treatment response to antidepressants in a han chinese population. Gen. Hosp. Psychiatry 2012, 34, 571–577. [Google Scholar] [CrossRef]
  26. Xu, Z.; Zhang, Z.; Shi, Y.; Pu, M.; Yuan, Y.; Zhang, X.; Li, L.; Reynolds, G.P. Influence and interaction of genetic polymorphisms in the serotonin system and life stress on antidepressant drug response. J. Psychopharmacol. 2012, 26, 349–359. [Google Scholar] [CrossRef]
  27. Illi, A.; Poutanen, O.; Setala-Soikkeli, E.; Kampman, O.; Viikki, M.; Huhtala, H.; Mononen, N.; Haraldsson, S.; Koivisto, P.A.; Leinonen, E.; et al. Is 5-httlpr linked to the response of selective serotonin reuptake inhibitors in mdd? Eur. Arch. Psychiatry Clin. Neurosci. 2011, 261, 95–102. [Google Scholar] [CrossRef]
  28. Kishi, T.; Yoshimura, R.; Kitajima, T.; Okochi, T.; Okumura, T.; Tsunoka, T.; Yamanouchi, Y.; Kinoshita, Y.; Kawashima, K.; Naitoh, H.; et al. Htr2a is associated with ssri response in major depressive disorder in a japanese cohort. Neuromolecular Med. 2010, 12, 237–242. [Google Scholar] [CrossRef] [PubMed]
  29. Liou, Y.J.; Chen, T.J.; Tsai, S.J.; Yu, Y.W.; Cheng, C.Y.; Hong, C.J. Support for the involvement of the kcnk2 gene in major depressive disorder and response to antidepressant treatment. Pharmacogenet. Genom. 2009, 19, 735–741. [Google Scholar] [CrossRef]
  30. Min, W.; Li, T.; Ma, X.; Li, Z.; Yu, T.; Gao, D.; Zhang, B.; Yun, Y.; Sun, X. Monoamine transporter gene polymorphisms affect susceptibility to depression and predict antidepressant response. Psychopharmacology 2009, 205, 409–417. [Google Scholar] [CrossRef]
  31. Kishi, T.; Kitajima, T.; Ikeda, M.; Yamanouchi, Y.; Kinoshita, Y.; Kawashima, K.; Okochi, T.; Okumura, T.; Tsunoka, T.; Ozaki, N.; et al. Clock may predict the response to fluvoxamine treatment in japanese major depressive disorder patients. Neuromolecular Med. 2009, 11, 53–57. [Google Scholar] [CrossRef]
  32. Tsai, S.J.; Hong, C.J.; Liou, Y.J.; Yu, Y.W.; Chen, T.J.; Hou, S.J.; Yen, F.C. Tryptophan hydroxylase 2 gene is associated with major depression and antidepressant treatment response. Prog. Neuropsychopharmacol. Biol. Psychiatry 2009, 33, 637–641. [Google Scholar] [CrossRef] [PubMed]
  33. Arias, B.; Serretti, A.; Mandelli, L.; Gasto, C.; Catalan, R.; Ronchi, D.D.; Fananas, L. Dysbindin gene (dtnbp1) in major depression: Association with clinical response to selective serotonin reuptake inhibitors. Pharmacogenet. Genom. 2009, 19, 121–128. [Google Scholar] [CrossRef]
  34. Wong, M.L.; Dong, C.; Maestre-Mesa, J.; Licinio, J. Polymorphisms in inflammation-related genes are associated with susceptibility to major depression and antidepressant response. Mol. Psychiatry 2008, 13, 800–812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Tsai, S.J.; Liou, Y.J.; Hong, C.J.; Yu, Y.W.; Chen, T.J. Glycogen synthase kinase-3beta gene is associated with antidepressant treatment response in chinese major depressive disorder. Pharmacogenom. J. 2008, 8, 384–390. [Google Scholar] [CrossRef] [PubMed]
  36. Gau, Y.T.; Liou, Y.J.; Yu, Y.W.; Chen, T.J.; Lin, M.W.; Tsai, S.J.; Hong, C.J. Evidence for association between genetic variants of p75 neurotrophin receptor (p75ntr) gene and antidepressant treatment response in chinese major depressive disorder. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2008, 147b, 594–599. [Google Scholar] [CrossRef] [PubMed]
  37. Bozina, N.; Peles, A.M.; Sagud, M.; Bilusic, H.; Jakovljevic, M. Association study of paroxetine therapeutic response with sert gene polymorphisms in patients with major depressive disorder. World J. Biol. Psychiatry 2008, 9, 190–197. [Google Scholar] [CrossRef] [PubMed]
  38. Papiol, S.; Arias, B.; Gasto, C.; Gutierrez, B.; Catalan, R.; Fananas, L. Genetic variability at hpa axis in major depression and clinical response to antidepressant treatment. J. Affect. Disord. 2007, 104, 83–90. [Google Scholar] [CrossRef] [PubMed]
  39. Ham, B.J.; Lee, B.C.; Paik, J.W.; Kang, R.H.; Choi, M.J.; Choi, I.G.; Lee, M.S. Association between the tryptophan hydroxylase-1 gene a218c polymorphism and citalopram antidepressant response in a korean population. Prog. Neuropsychopharmacol. Biol. Psychiatry 2007, 31, 104–107. [Google Scholar] [CrossRef]
  40. Choi, M.J.; Kang, R.H.; Lim, S.W.; Oh, K.S.; Lee, M.S. Brain-derived neurotrophic factor gene polymorphism (val66met) and citalopram response in major depressive disorder. Brain Res. 2006, 1118, 176–182. [Google Scholar] [CrossRef]
  41. Hong, C.J.; Chen, T.J.; Yu, Y.W.; Tsai, S.J. Response to fluoxetine and serotonin 1a receptor (c-1019g) polymorphism in taiwan chinese major depressive disorder. Pharmacogenom. J. 2006, 6, 27–33. [Google Scholar] [CrossRef]
  42. Choi, M.J.; Kang, R.H.; Ham, B.J.; Jeong, H.Y.; Lee, M.S. Serotonin receptor 2a gene polymorphism (-1438a/g) and short-term treatment response to citalopram. Neuropsychobiology 2005, 52, 155–162. [Google Scholar] [CrossRef]
  43. Kraft, J.B.; Slager, S.L.; McGrath, P.J.; Hamilton, S.P. Sequence analysis of the serotonin transporter and associations with antidepressant response. Biol. Psychiatry 2005, 58, 374–381. [Google Scholar] [CrossRef]
  44. Yu, Y.W.; Chen, T.J.; Hong, C.J.; Chen, H.M.; Tsai, S.J. Association study of the interleukin-1 beta (c-511t) genetic polymorphism with major depressive disorder, associated symptomatology, and antidepressant response. Neuropsychopharmacology 2003, 28, 1182–1185. [Google Scholar] [CrossRef]
  45. Yoshida, K.; Ito, K.; Sato, K.; Takahashi, H.; Kamata, M.; Higuchi, H.; Shimizu, T.; Itoh, K.; Inoue, K.; Tezuka, T.; et al. Influence of the serotonin transporter gene-linked polymorphic region on the antidepressant response to fluvoxamine in japanese depressed patients. Prog. Neuropsychopharmacol. Biol. Psychiatry 2002, 26, 383–386. [Google Scholar] [CrossRef]
  46. Yin, L.; Zhang, X.; Huang, Y.; Sun, X. Catecholamine pathway polymorphisms and antidepressant response. Asia Pac. Psychiatry 2016, 8, 109–117. [Google Scholar] [CrossRef]
  47. Chang, H.S.; Lee, H.-Y.; Ham, B.-J.; Park, Y.C.; Hahn, S.-W.; Jeong, Y.-J.; Kim, B.; Lee, M.-S. Association between brain-derived neurotrophic factor v66m and treatment responses to escitalopram in patients with major depressive disorder. Asia Pac. Psychiatry 2012, 4, 241–249. [Google Scholar]
  48. Lin, K.M.; Tsou, H.H.; Tsai, I.J.; Hsiao, M.C.; Hsiao, C.F.; Liu, C.Y.; Shen, W.W.; Tang, H.S.; Fang, C.K.; Wu, C.S.; et al. Cyp1a2 genetic polymorphisms are associated with treatment response to the antidepressant paroxetine. Pharmacogenomics 2010, 11, 1535–1543. [Google Scholar] [CrossRef]
  49. Lee, S.H.; Lee, M.S.; Lee, J.H.; Kim, S.W.; Kang, R.H.; Choi, M.J.; Park, S.J.; Kim, S.J.; Lee, J.M.; Cole, S.P.; et al. Mrp1 polymorphisms associated with citalopram response in patients with major depression. J. Clin. Psychopharmacol. 2010, 30, 116–125. [Google Scholar] [CrossRef]
  50. Tsai, S.J.; Gau, Y.T.; Hong, C.J.; Liou, Y.J.; Yu, Y.W.; Chen, T.J. Sexually dimorphic effect of catechol-o-methyltransferase val158met polymorphism on clinical response to fluoxetine in major depressive patients. J. Affect. Disord. 2009, 113, 183–187. [Google Scholar] [CrossRef]
  51. Yu, Y.W.; Tsai, S.J.; Liou, Y.J.; Hong, C.J.; Chen, T.J. Association study of two serotonin 1a receptor gene polymorphisms and fluoxetine treatment response in chinese major depressive disorders. Eur. Neuropsychopharmacol. 2006, 16, 498–503. [Google Scholar] [CrossRef]
  52. Suzuki, Y.; Sawamura, K.; Someya, T. The effects of a 5-hydroxytryptamine 1a receptor gene polymorphism on the clinical response to fluvoxamine in depressed patients. Pharmacogenom. J. 2004, 4, 283–286. [Google Scholar] [CrossRef] [PubMed]
  53. Jamerson, B.D.; Payne, M.E.; Garrett, M.E.; Ashley-Koch, A.E.; Speer, M.C.; Steffens, D.C. Folate metabolism genes, dietary folate and response to antidepressant medications in late-life depression. Int. J. Geriatr Psychiatry. 2013, 28, 925–932. [Google Scholar] [CrossRef]
  54. Fabbri, C.; Drago, A.; Serretti, A. Early antidepressant efficacy modulation by glutamatergic gene variants in the star*d. Eur. Neuropsychopharmacol. 2013, 23, 612–621. [Google Scholar] [CrossRef]
  55. Glubb, D.M.; McHugh, P.C.; Deng, X.; Joyce, P.R.; Kennedy, M.A. Association of a functional polymorphism in the adrenomedullin gene (adm) with response to paroxetine. Pharmacogenom. J. 2010, 10, 126–133. [Google Scholar] [CrossRef]
  56. Peters, E.J.; Slager, S.L.; Jenkins, G.D.; Reinalda, M.S.; Garriock, H.A.; Shyn, S.I.; Kraft, J.B.; McGrath, P.J.; Hamilton, S.P. Resequencing of serotonin-related genes and association of tagging snps to citalopram response. Pharmacogenet. Genom. 2009, 19, 1–10. [Google Scholar] [CrossRef]
  57. Mrazek, D.A.; Rush, A.J.; Biernacka, J.M.; O’Kane, D.J.; Cunningham, J.M.; Wieben, E.D.; Schaid, D.J.; Drews, M.S.; Courson, V.L.; Snyder, K.A.; et al. Slc6a4 variation and citalopram response. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2009, 150b, 341–351. [Google Scholar] [CrossRef]
  58. Kato, M.; Fukuda, T.; Wakeno, M.; Fukuda, K.; Okugawa, G.; Ikenaga, Y.; Yamashita, M.; Takekita, Y.; Nobuhara, K.; Azuma, J.; et al. Effects of the serotonin type 2a, 3a and 3b receptor and the serotonin transporter genes on paroxetine and fluvoxamine efficacy and adverse drug reactions in depressed japanese patients. Neuropsychobiology 2006, 53, 186–195. [Google Scholar] [CrossRef]
  59. McMahon, F.J.; Buervenich, S.; Charney, D.; Lipsky, R.; Rush, A.J.; Wilson, A.F.; Sorant, A.J.M.; Papanicolaou, G.J.; Laje, G.; Fava, M.; et al. Variation in the gene encoding the serotonin 2a receptor is associated with outcome of antidepressant treatment. Am. J. Hum. Genet. 2006, 78, 804–814. [Google Scholar] [CrossRef]
  60. Peters, E.J.; Slager, S.L.; McGrath, P.J.; Knowles, J.A.; Hamilton, S.P. Investigation of serotonin-related genes in antidepressant response. Mol. Psychiatry 2004, 9, 879–889. [Google Scholar] [CrossRef] [Green Version]
  61. Ji, Y.; Biernacka, J.; Snyder, K.; Drews, M.; Pelleymounter, L.L.; Colby, C.; Wang, L.; Mrazek, D.A.; Weinshilboum, R.M. Catechol o-methyltransferase pharmacogenomics and selective serotonin reuptake inhibitor response. Pharmacogenom. J. 2012, 12, 78–85. [Google Scholar] [CrossRef]
  62. Lekman, M.; Laje, G.; Charney, D.; Rush, A.J.; Wilson, A.F.; Sorant, A.J.; Lipsky, R.; Wisniewski, S.R.; Manji, H.; McMahon, F.J.; et al. The fkbp5-gene in depression and treatment response--an association study in the sequenced treatment alternatives to relieve depression (star*d) cohort. Biol. Psychiatry 2008, 63, 1103–1110. [Google Scholar] [CrossRef]
  63. Kraft, J.B.; Peters, E.J.; Slager, S.L.; Jenkins, G.D.; Reinalda, M.S.; McGrath, P.J.; Hamilton, S.P. Analysis of association between the serotonin transporter and antidepressant response in a large clinical sample. Biol. Psychiatry 2007, 61, 734–742. [Google Scholar] [CrossRef]
  64. Binder, E.B.; Owens, M.J.; Liu, W.; Deveau, T.C.; Rush, A.J.; Trivedi, M.H.; Fava, M.; Bradley, B.; Ressler, K.J.; Nemeroff, C.B. Association of polymorphisms in genes regulating the corticotropin-releasing factor system with antidepressant treatment response. Arch. Gen. Psychiatry 2010, 67, 369–379. [Google Scholar] [CrossRef] [PubMed]
  65. Garriock, H.A.; Tanowitz, M.; Kraft, J.B.; Dang, V.C.; Peters, E.J.; Jenkins, G.D.; Reinalda, M.S.; McGrath, P.J.; von Zastrow, M.; Slager, S.L.; et al. Association of mu-opioid receptor variants and response to citalopram treatment in major depressive disorder. Am. J. Psychiatry 2010, 167, 565–573. [Google Scholar] [CrossRef]
  66. Lin, K.M.; Chiu, Y.F.; Tsai, I.J.; Chen, C.H.; Shen, W.W.; Liu, S.C.; Lu, S.C.; Liu, C.Y.; Hsiao, M.C.; Tang, H.S.; et al. Abcb1 gene polymorphisms are associated with the severity of major depressive disorder and its response to escitalopram treatment. Pharmacogenet. Genom. 2011, 21, 163–170. [Google Scholar] [CrossRef] [PubMed]
  67. Myung, W.; Kim, J.; Lim, S.W.; Shim, S.; Won, H.H.; Kim, S.; Kim, S.; Lee, M.S.; Chang, H.S.; Kim, J.W.; et al. A genome-wide association study of antidepressant response in koreans. Transl. Psychiatry 2015, 5, e633. [Google Scholar] [CrossRef] [PubMed]
  68. Biernacka, J.M.; Sangkuhl, K.; Jenkins, G.; Whaley, R.M.; Barman, P.; Batzler, A.; Altman, R.B.; Arolt, V.; Brockmoller, J.; Chen, C.H.; et al. The international ssri pharmacogenomics consortium (ispc): A genome-wide association study of antidepressant treatment response. Transl. Psychiatry 2015, 5, e553. [Google Scholar] [CrossRef]
  69. Hunter, A.M.; Leuchter, A.F.; Power, R.A.; Muthen, B.; McGrath, P.J.; Lewis, C.M.; Cook, I.A.; Garriock, H.A.; McGuffin, P.; Uher, R.; et al. A genome-wide association study of a sustained pattern of antidepressant response. J. Psychiatr. Res. 2013, 47, 1157–1165. [Google Scholar] [CrossRef] [PubMed]
  70. Tansey, K.E.; Guipponi, M.; Perroud, N.; Bondolfi, G.; Domenici, E.; Evans, D.; Hall, S.K.; Hauser, J.; Henigsberg, N.; Hu, X.; et al. Genetic predictors of response to serotonergic and noradrenergic antidepressants in major depressive disorder: A genome-wide analysis of individual-level data and a meta-analysis. PLoS Med. 2012, 9, e1001326. [Google Scholar] [CrossRef] [PubMed]
  71. Ji, Y.; Biernacka, J.M.; Hebbring, S.; Chai, Y.; Jenkins, G.D.; Batzler, A.; Snyder, K.A.; Drews, M.S.; Desta, Z.; Flockhart, D.; et al. Pharmacogenomics of selective serotonin reuptake inhibitor treatment for major depressive disorder: Genome-wide associations and functional genomics. Pharmacogenom. J. 2013, 13, 456–463. [Google Scholar] [CrossRef] [PubMed]
  72. Sasayama, D.; Hiraishi, A.; Tatsumi, M.; Kamijima, K.; Ikeda, M.; Umene-Nakano, W.; Yoshimura, R.; Nakamura, J.; Iwata, N.; Kunugi, H. Possible association of cux1 gene polymorphisms with antidepressant response in major depressive disorder. Pharmacogenom. J. 2013, 13, 354–358. [Google Scholar] [CrossRef]
  73. Uher, R.; Perroud, N.; Ng, M.Y.; Hauser, J.; Henigsberg, N.; Maier, W.; Mors, O.; Placentino, A.; Rietschel, M.; Souery, D.; et al. Genome-wide pharmacogenetics of antidepressant response in the gendep project. Am. J. Psychiatry 2010, 167, 555–564. [Google Scholar] [CrossRef]
  74. Power, R.A.; Cohen-Woods, S.; Ng, M.Y.; Butler, A.W.; Craddock, N.; Korszun, A.; Jones, L.; Jones, I.; Gill, M.; Rice, J.P.; et al. Genome-wide association analysis accounting for environmental factors through propensity-score matching: Application to stressful live events in major depressive disorder. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2013, 162b, 521–529. [Google Scholar] [CrossRef] [PubMed]
  75. Ripke, S.; Wray, N.R.; Lewis, C.M.; Hamilton, S.P.; Weissman, M.M.; Breen, G.; Byrne, E.M.; Blackwood, D.H.; Boomsma, D.I.; Cichon, S.; et al. A mega-analysis of genome-wide association studies for major depressive disorder. Mol. Psychiatry 2013, 18, 497–511. [Google Scholar]
  76. Wray, N.R.; Pergadia, M.L.; Blackwood, D.H.; Penninx, B.W.; Gordon, S.D.; Nyholt, D.R.; Ripke, S.; MacIntyre, D.J.; McGhee, K.A.; Maclean, A.W.; et al. Genome-wide association study of major depressive disorder: New results, meta-analysis, and lessons learned. Mol. Psychiatry 2012, 17, 36–48. [Google Scholar] [CrossRef]
  77. Shi, J.; Potash, J.B.; Knowles, J.A.; Weissman, M.M.; Coryell, W.; Scheftner, W.A.; Lawson, W.B.; DePaulo, J.R., Jr.; Gejman, P.V.; Sanders, A.R.; et al. Genome-wide association study of recurrent early-onset major depressive disorder. Mol. Psychiatry 2011, 16, 193–201. [Google Scholar] [CrossRef]
  78. Shyn, S.I.; Shi, J.; Kraft, J.B.; Potash, J.B.; Knowles, J.A.; Weissman, M.M.; Garriock, H.A.; Yokoyama, J.S.; McGrath, P.J.; Peters, E.J.; et al. Novel loci for major depression identified by genome-wide association study of sequenced treatment alternatives to relieve depression and meta-analysis of three studies. Mol. Psychiatry 2011, 16, 202–215. [Google Scholar] [CrossRef] [PubMed]
  79. Muglia, P.; Tozzi, F.; Galwey, N.W.; Francks, C.; Upmanyu, R.; Kong, X.Q.; Antoniades, A.; Domenici, E.; Perry, J.; Rothen, S.; et al. Genome-wide association study of recurrent major depressive disorder in two european case-control cohorts. Mol. Psychiatry 2010, 15, 589–601. [Google Scholar] [CrossRef] [PubMed]
  80. Sullivan, P.F.; de Geus, E.J.; Willemsen, G.; James, M.R.; Smit, J.H.; Zandbelt, T.; Arolt, V.; Baune, B.T.; Blackwood, D.; Cichon, S.; et al. Genome-wide association for major depressive disorder: A possible role for the presynaptic protein piccolo. Mol. Psychiatry 2009, 14, 359–375. [Google Scholar] [CrossRef]
  81. Rietschel, M.; Mattheisen, M.; Frank, J.; Treutlein, J.; Degenhardt, F.; Breuer, R.; Steffens, M.; Mier, D.; Esslinger, C.; Walter, H.; et al. Genome-wide association-, replication-, and neuroimaging study implicates homer1 in the etiology of major depression. Biol. Psychiatry 2010, 68, 578–585. [Google Scholar] [CrossRef] [PubMed]
  82. Murray, C.J.L.; Lopez, A.D. The Global Burden of Disease: A Comprehensive Assessment of Mortality and Disability from Diseases, Injuries, and Risk Factors in 1990 and Projected to 2020; Harvard School of Public Health: Boston, MA, USA, 1996. [Google Scholar]
  83. Guin, D.; Mishra, M.K.; Talwar, P.; Rawat, C.; Kushwaha, S.S.; Kukreti, S.; Kukreti, R. A systematic review and integrative approach to decode the common molecular link between levodopa response and parkinson’s disease. BMC Med. Genom. 2017, 10, 56. [Google Scholar] [CrossRef] [PubMed]
  84. Zill, P.; Baghai, T.C.; Zwanzger, P.; Schule, C.; Eser, D.; Rupprecht, R.; Moller, H.J.; Bondy, B.; Ackenheil, M. Snp and haplotype analysis of a novel tryptophan hydroxylase isoform (tph2) gene provide evidence for association with major depression. Mol. Psychiatry 2004, 9, 1030–1036. [Google Scholar] [CrossRef] [PubMed]
  85. Kim, H.; Lim, S.W.; Kim, S.; Kim, J.W.; Chang, Y.H.; Carroll, B.J.; Kim, D.K. Monoamine transporter gene polymorphisms and antidepressant response in koreans with late-life depression. JAMA 2006, 296, 1609–1618. [Google Scholar] [CrossRef]
  86. Kato, M.; Serretti, A. Review and meta-analysis of antidepressant pharmacogenetic findings in major depressive disorder. Mol. Psychiatry 2010, 15, 473–500. [Google Scholar] [CrossRef] [PubMed]
  87. Zill, P.; Baghai, T.C.; Engel, R.; Zwanzger, P.; Schule, C.; Minov, C.; Behrens, S.; Bottlender, R.; Jager, M.; Rupprecht, R.; et al. Beta-1-adrenergic receptor gene in major depression: Influence on antidepressant treatment response. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2003, 120b, 85–89. [Google Scholar] [CrossRef] [PubMed]
  88. Lucae, S.; Ising, M.; Horstmann, S.; Baune, B.T.; Arolt, V.; Muller-Myhsok, B.; Holsboer, F.; Domschke, K. Htr2a gene variation is involved in antidepressant treatment response. Eur. Neuropsychopharmacol. 2010, 20, 65–68. [Google Scholar] [CrossRef] [PubMed]
  89. Horstmann, S.; Lucae, S.; Menke, A.; Hennings, J.M.; Ising, M.; Roeske, D.; Muller-Myhsok, B.; Holsboer, F.; Binder, E.B. Polymorphisms in grik4, htr2a, and fkbp5 show interactive effects in predicting remission to antidepressant treatment. Neuropsychopharmacology 2010, 35, 727–740. [Google Scholar] [CrossRef]
  90. Illi, A.; Setala-Soikkeli, E.; Viikki, M.; Poutanen, O.; Huhtala, H.; Mononen, N.; Lehtimaki, T.; Leinonen, E.; Kampman, O. 5-htr1a, 5-htr2a, 5-htr6, tph1 and tph2 polymorphisms and major depression. Neuroreport 2009, 20, 1125–1128. [Google Scholar] [CrossRef] [PubMed]
  91. Fabbri, C.; Marsano, A.; Serretti, A. Genetics of serotonin receptors and depression: State of the art. Curr. Drug Targets 2013, 14, 531–548. [Google Scholar] [CrossRef]
  92. Keller, J.; Gomez, R.; Williams, G.; Lembke, A.; Lazzeroni, L.; Murphy, G.M., Jr.; Schatzberg, A.F. Hpa axis in major depression: Cortisol, clinical symptomatology and genetic variation predict cognition. Mol. Psychiatry 2017, 22, 527–536. [Google Scholar] [CrossRef] [PubMed]
  93. Young, E.A.; Lopez, J.F.; Murphy-Weinberg, V.; Watson, S.J.; Akil, H. Mineralocorticoid receptor function in major depression. Arch. Gen. Psychiatry 2003, 60, 24–28. [Google Scholar] [CrossRef] [PubMed]
  94. Holsboer, F. The corticosteroid receptor hypothesis of depression. Neuropsychopharmacology 2000, 23, 477–501. [Google Scholar] [CrossRef]
  95. Mello, A.F.; Juruena, M.F.; Pariante, C.M.; Tyrka, A.R.; Price, L.H.; Carpenter, L.L.; Del Porto, J.A. Depression and stress: Is there an endophenotype? Braz. J. Psychiatry 2007, 29 (Suppl. 1), S13–S18. [Google Scholar] [CrossRef]
  96. Arnett, M.G.; Muglia, L.M.; Laryea, G.; Muglia, L.J. Genetic approaches to hypothalamic-pituitary-adrenal axis regulation. Neuropsychopharmacology 2016, 41, 245–260. [Google Scholar] [CrossRef] [PubMed]
  97. Bradley, R.G.; Binder, E.B.; Epstein, M.P.; Tang, Y.; Nair, H.P.; Liu, W.; Gillespie, C.F.; Berg, T.; Evces, M.; Newport, D.J.; et al. Influence of child abuse on adult depression: Moderation by the corticotropin-releasing hormone receptor gene. Arch. Gen. Psychiatry 2008, 65, 190–200. [Google Scholar] [CrossRef] [PubMed]
  98. Polanczyk, G.; Caspi, A.; Williams, B.; Price, T.S.; Danese, A.; Sugden, K.; Uher, R.; Poulton, R.; Moffitt, T.E. Protective effect of crhr1 gene variants on the development of adult depression following childhood maltreatment: Replication and extension. Arch. Gen. Psychiatry 2009, 66, 978–985. [Google Scholar] [CrossRef] [PubMed]
  99. Kranzler, H.R.; Feinn, R.; Nelson, E.C.; Covault, J.; Anton, R.F.; Farrer, L.; Gelernter, J. A crhr1 haplotype moderates the effect of adverse childhood experiences on lifetime risk of major depressive episode in african-american women. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2011, 156B, 960–968. [Google Scholar] [CrossRef]
  100. Woody, M.L.; Kudinova, A.Y.; McGeary, J.E.; Knopik, V.S.; Palmer, R.H.; Gibb, B.E. Influence of maternal depression on children’s brooding rumination: Moderation by crhr1 tat haplotype. Cogn. Emot. 2016, 30, 302–314. [Google Scholar] [CrossRef] [PubMed]
  101. Xu, Y.J.; Sheng, H.; Wu, T.W.; Bao, Q.Y.; Zheng, Y.; Zhang, Y.M.; Gong, Y.X.; Lu, J.Q.; You, Z.D.; Xia, Y.; et al. Crh/crhr1 mediates prenatal synthetic glucocorticoid programming of depression-like behavior across 2 generations. FASEB J. 2018, 32, 4258–4269. [Google Scholar] [CrossRef] [PubMed]
  102. Szczepankiewicz, A.; Leszczynska-Rodziewicz, A.; Pawlak, J.; Rajewska-Rager, A.; Wilkosc, M.; Zaremba, D.; Dmitrzak-Weglarz, M.; Skibinska, M.; Hauser, J. Epistatic interaction between crhr1 and avpr1b variants as a predictor of major depressive disorder. Psychiatr. Genet. 2013, 23, 239–246. [Google Scholar] [CrossRef]
  103. Xiao, Z.; Liu, W.; Gao, K.; Wan, Q.; Yang, C.; Wang, H.; Wang, X.; Wang, G.; Liu, Z. Interaction between crhr1 and bdnf genes increases the risk of recurrent major depressive disorder in chinese population. PLoS ONE 2011, 6, e28733. [Google Scholar] [CrossRef]
  104. Ressler, K.J.; Bradley, B.; Mercer, K.B.; Deveau, T.C.; Smith, A.K.; Gillespie, C.F.; Nemeroff, C.B.; Cubells, J.F.; Binder, E.B. Polymorphisms in crhr1 and the serotonin transporter loci: Gene x gene x environment interactions on depressive symptoms. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2010, 153B, 812–824. [Google Scholar]
  105. Ishitobi, Y.; Nakayama, S.; Yamaguchi, K.; Kanehisa, M.; Higuma, H.; Maruyama, Y.; Ninomiya, T.; Okamoto, S.; Tanaka, Y.; Tsuru, J.; et al. Association of crhr1 and crhr2 with major depressive disorder and panic disorder in a japanese population. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2012, 159B, 429–436. [Google Scholar] [CrossRef] [PubMed]
  106. Villafuerte, S.M.; Del-Favero, J.; Adolfsson, R.; Souery, D.; Massat, I.; Mendlewicz, J.; Van Broeckhoven, C.; Claes, S. Gene-based snp genetic association study of the corticotropin-releasing hormone receptor-2 (crhr2) in major depression. Am. J. Med. Genet. 2002, 114, 222–226. [Google Scholar] [CrossRef] [PubMed]
  107. Anacker, C.; Zunszain, P.A.; Cattaneo, A.; Carvalho, L.A.; Garabedian, M.J.; Thuret, S.; Price, J.; Pariante, C.M. Antidepressants increase human hippocampal neurogenesis by activating the glucocorticoid receptor. Mol. Psychiatry 2011, 16, 738–750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Licinio, J.; O’Kirwan, F.; Irizarry, K.; Merriman, B.; Thakur, S.; Jepson, R.; Lake, S.; Tantisira, K.G.; Weiss, S.T.; Wong, M.L. Association of a corticotropin-releasing hormone receptor 1 haplotype and antidepressant treatment response in mexican-americans. Mol. Psychiatry 2004, 9, 1075–1082. [Google Scholar] [CrossRef]
  109. Liu, Z.; Zhu, F.; Wang, G.; Xiao, Z.; Tang, J.; Liu, W.; Wang, H.; Liu, H.; Wang, X.; Wu, Y.; et al. Association study of corticotropin-releasing hormone receptor1 gene polymorphisms and antidepressant response in major depressive disorders. Neurosci. Lett. 2007, 414, 155–158. [Google Scholar] [CrossRef]
  110. Niciu, M.J.; Ionescu, D.F.; Richards, E.M.; Zarate, C.A., Jr. Glutamate and its receptors in the pathophysiology and treatment of major depressive disorder. J. Neural. Transm. 2014, 121, 907–924. [Google Scholar] [CrossRef] [PubMed]
  111. Murrough, J.W.; Abdallah, C.G.; Mathew, S.J. Targeting glutamate signalling in depression: Progress and prospects. Nat. Rev. Drug Discov. 2017, 16, 472–486. [Google Scholar] [CrossRef] [PubMed]
  112. Popoli, M.; Yan, Z.; McEwen, B.S.; Sanacora, G. The stressed synapse: The impact of stress and glucocorticoids on glutamate transmission. Nat. Rev. Neurosci. 2011, 13, 22–37. [Google Scholar] [CrossRef] [PubMed]
  113. Hashimoto, K.; Sawa, A.; Iyo, M. Increased levels of glutamate in brains from patients with mood disorders. Biol. Psychiatry 2007, 62, 1310–1316. [Google Scholar] [CrossRef]
  114. Duric, V.; Banasr, M.; Stockmeier, C.A.; Simen, A.A.; Newton, S.S.; Overholser, J.C.; Jurjus, G.J.; Dieter, L.; Duman, R.S. Altered expression of synapse and glutamate related genes in post-mortem hippocampus of depressed subjects. Int. J. Neuropsychopharmacol. 2013, 16, 69–82. [Google Scholar] [CrossRef]
  115. Lee, L.C.; Cho, Y.C.; Lin, P.J.; Yeh, T.C.; Chang, C.Y.; Yeh, T.K. Influence of genetic variants of the n-methyl-d-aspartate receptor on emotion and social behavior in adolescents. Neural Plast 2016, 2016, 6851592. [Google Scholar] [CrossRef]
  116. Feyissa, A.M.; Chandran, A.; Stockmeier, C.A.; Karolewicz, B. Reduced levels of nr2a and nr2b subunits of nmda receptor and psd-95 in the prefrontal cortex in major depression. Prog. Neuropsychopharmacol. Biol. Psychiatry 2009, 33, 70–75. [Google Scholar] [CrossRef] [PubMed]
  117. Li, W.; Ju, K.; Li, Z.; He, K.; Chen, J.; Wang, Q.; Yang, B.; An, L.; Feng, G.; Sun, W.; et al. Significant association of grm7 and grm8 genes with schizophrenia and major depressive disorder in the han chinese population. Eur. Neuropsychopharmacol. 2016, 26, 136–146. [Google Scholar] [CrossRef] [PubMed]
  118. Verbeek, E.C.; Bevova, M.R.; Bochdanovits, Z.; Rizzu, P.; Bakker, I.M.; Uithuisje, T.; De Geus, E.J.; Smit, J.H.; Penninx, B.W.; Boomsma, D.I.; et al. Resequencing three candidate genes for major depressive disorder in a dutch cohort. PLoS ONE 2013, 8, e79921. [Google Scholar] [CrossRef]
  119. Paddock, S.; Laje, G.; Charney, D.; Rush, A.J.; Wilson, A.F.; Sorant, A.J.; Lipsky, R.; Wisniewski, S.R.; Manji, H.; McMahon, F.J. Association of grik4 with outcome of antidepressant treatment in the star*d cohort. Am. J. Psychiatry 2007, 164, 1181–1188. [Google Scholar] [CrossRef] [PubMed]
  120. Pu, M.; Zhang, Z.; Xu, Z.; Shi, Y.; Geng, L.; Yuan, Y.; Zhang, X.; Reynolds, G.P. Influence of genetic polymorphisms in the glutamatergic and gabaergic systems and their interactions with environmental stressors on antidepressant response. Pharmacogenomics 2013, 14, 277–288. [Google Scholar] [CrossRef]
  121. Laje, G.; Paddock, S.; Manji, H.; Rush, A.J.; Wilson, A.F.; Charney, D.; McMahon, F.J. Genetic markers of suicidal ideation emerging during citalopram treatment of major depression. Am. J. Psychiatry 2007, 164, 1530–1538. [Google Scholar] [CrossRef]
  122. Jaso, B.A.; Niciu, M.J.; Iadarola, N.D.; Lally, N.; Richards, E.M.; Park, M.; Ballard, E.D.; Nugent, A.C.; Machado-Vieira, R.; Zarate, C.A. Therapeutic modulation of glutamate receptors in major depressive disorder. Curr. Neuropharmacol. 2017, 15, 57–70. [Google Scholar] [CrossRef]
  123. Story Jovanova, O.; Nedeljkovic, I.; Derek, S.; Walker, R.M.; Liu, C.; Luciano, M.; Bressler, J.; Brody, J.; Drake, A.J.; Evans, K.L.; et al. DNA methylation signatures of depressive symptoms in middle-aged and elderly persons: Meta-analysis of multiethnic epigenome-wide studies. JAMA Psychiatry 2018. [Google Scholar] [CrossRef]
  124. Enatescu, V.R.; Papava, I.; Enatescu, I.; Antonescu, M.; Anghel, A.; Seclaman, E.; Sirbu, I.O.; Marian, C. Circulating plasma micro rnas in patients with major depressive disorder treated with antidepressants: A pilot study. Psychiatry Investig. 2016, 13, 549–557. [Google Scholar] [CrossRef]
  125. Bocchio-Chiavetto, L.; Maffioletti, E.; Bettinsoli, P.; Giovannini, C.; Bignotti, S.; Tardito, D.; Corrada, D.; Milanesi, L.; Gennarelli, M. Blood microrna changes in depressed patients during antidepressant treatment. Eur. Neuropsychopharmacol. 2013, 23, 602–611. [Google Scholar] [CrossRef]
  126. Glessner, J.T.; Wang, K.; Sleiman, P.M.; Zhang, H.; Kim, C.E.; Flory, J.H.; Bradfield, J.P.; Imielinski, M.; Frackelton, E.C.; Qiu, H.; et al. Duplication of the slit3 locus on 5q35.1 predisposes to major depressive disorder. PLoS ONE 2010, 5, e15463. [Google Scholar] [CrossRef] [PubMed]
  127. Sangkuhl, K.; Klein, T.E.; Altman, R.B. Selective serotonin reuptake inhibitors pathway. Pharmacogenet. Genom. 2009, 19, 907–909. [Google Scholar] [CrossRef]
  128. Lodish, M.; Stratakis, C.A. A genetic and molecular update on adrenocortical causes of cushing syndrome. Nat. Rev. Endocrinol. 2016, 12, 255–262. [Google Scholar] [CrossRef] [PubMed]
  129. Dowlatshahi, D.; MacQueen, G.M.; Wang, J.F.; Young, L.T. Increased temporal cortex creb concentrations and antidepressant treatment in major depression. Lancet 1998, 352, 1754–1755. [Google Scholar]
  130. Odagaki, Y.; Garcia-Sevilla, J.A.; Huguelet, P.; La Harpe, R.; Koyama, T.; Guimon, J. Cyclic amp-mediated signaling components are upregulated in the prefrontal cortex of depressed suicide victims. Brain Res. 2001, 898, 224–231. [Google Scholar] [CrossRef]
  131. Balu, D.T.; Hoshaw, B.A.; Malberg, J.E.; Rosenzweig-Lipson, S.; Schechter, L.E.; Lucki, I. Differential regulation of central bdnf protein levels by antidepressant and non-antidepressant drug treatments. Brain Res. 2008, 1211, 37–43. [Google Scholar] [CrossRef]
  132. Drago, A.; De Ronchi, D.; Serretti, A. Pharmacogenetics of antidepressant response: An update. Hum. Genomics 2009, 3, 257–274. [Google Scholar] [CrossRef]
  133. Mamdani, F.; Berlim, M.T.; Beaulieu, M.M.; Turecki, G. Pharmacogenomic predictors of citalopram treatment outcome in major depressive disorder. World J. Biol. Psychiatry 2014, 15, 135–144. [Google Scholar] [CrossRef]
  134. Hutton, B.; Salanti, G.; Caldwell, D.M.; Chaimani, A.; Schmid, C.H.; Cameron, C.; Ioannidis, J.P.; Straus, S.; Thorlund, K.; Jansen, J.P.; et al. The prisma extension statement for reporting of systematic reviews incorporating network meta-analyses of health care interventions: Checklist and explanations. Ann. Intern. Med. 2015, 162, 777–784. [Google Scholar] [CrossRef]
  135. Ward, L.D.; Kellis, M. Haploreg: A resource for exploring chromatin states, conservation, and regulatory motif alterations within sets of genetically linked variants. Nucleic Acids Res. 2012, 40, D930–D934. [Google Scholar] [CrossRef]
  136. Gamazon, E.R.; Zhang, W.; Konkashbaev, A.; Duan, S.; Kistner, E.O.; Nicolae, D.L.; Dolan, M.E.; Cox, N.J. Scan: Snp and copy number annotation. Bioinformatics 2010, 26, 259–262. [Google Scholar] [CrossRef]
  137. McLaren, W.; Gil, L.; Hunt, S.E.; Riat, H.S.; Ritchie, G.R.; Thormann, A.; Flicek, P.; Cunningham, F. The ensembl variant effect predictor. Genome Biol. 2016, 17, 122. [Google Scholar] [CrossRef]
  138. Gray, K.A.; Daugherty, L.C.; Gordon, S.M.; Seal, R.L.; Wright, M.W.; Bruford, E.A. Genenames.Org: The hgnc resources in 2013. Nucleic Acids Res. 2013, 41, D545–D552. [Google Scholar] [CrossRef]
Figure 1. PRISMA flowchart of the study selection process. Reasons for articles that were excluded given in the diagram.
Figure 1. PRISMA flowchart of the study selection process. Reasons for articles that were excluded given in the diagram.
Ijms 20 01993 g001
Figure 2. Common Genes (a), KEGG biological pathways (b), and GO terms (c) among SSRI response and MDD.
Figure 2. Common Genes (a), KEGG biological pathways (b), and GO terms (c) among SSRI response and MDD.
Ijms 20 01993 g002
Figure 3. Top five pathways associated with an antidepressant response as well as MDD etiology.
Figure 3. Top five pathways associated with an antidepressant response as well as MDD etiology.
Ijms 20 01993 g003
Figure 4. Flow chart of the study.
Figure 4. Flow chart of the study.
Ijms 20 01993 g004
Table 1. Characteristics of included studies for assessment of the association between genetic variants and SSRIs response in major depressive disorder.
Table 1. Characteristics of included studies for assessment of the association between genetic variants and SSRIs response in major depressive disorder.
No.Study NameSample SizeGenesStudied Variantsp-ValueOR (95% CI)
Genotypic
OR (95% CI)
Allelic
DrugsFPScore
MFTotal (n)Responder/Remitter (n)Non Responder/Non Remitter (n)GenotypicAllelic
1Nouraei H et al. (2018) [10]33671007030GCRrs414232470.0080.0323.3 (1.35–8.09)2.2 (1.09–4.44)Fluoxetine69
2Firouzabadi N et al. (2017) [11]25751003367ADRB1rs18012530.0030.00025.7 (1.4–23.9)3.3 (1.72–6.50)Sertraline69
3Xu Z et al. (2016) [12]11616528111450TPH2rs111789980.0209N.A.2.3 (1.14–4.50)N.A.SSRI69
rs79637170.0239N.A.2.2 (1.09–4.35)
4Manoharan A et al. (2016) [13]35671025646SLC6A45-HTTLPR0.0066N.A.4.0 (1.45–11.03)N.A.Fluoxetine69
5Paroni G et al. (2017) [14]95234329176153KLrs95363140.011N.A.N.A.N.A.Escitalopram, Sertraline, Paroxetine, Citalopram228
6Lim SW et al. (2014) [15]5918023915485TPH2rs47608150.00001N.A.N.A.N.A.SSRI69
rs111790270.00002N.A.
rs171105320.00009N.A.
rs171107470.0002N.A.
GRIK2rs5431960.00005N.A.
rs5724870.0001N.A.
GAD1rs38282750.00007N.A.
rs121856920.0002N.A.
SLC6A4rs20667130.0001N.A.
rs20209420.0003N.A.
7Fukui N et al. (2014) [16]65581232435COMTrs2075507N.A.0.0036N.A.N.A.Fluvoxamine127
rs1544325N.A.0.0036
rs5993883N.A.0.015
8Li X et al. (2014) [17]14114929022070SLC17A7rs741742840.0140.0080.57 (0.38–0.87)N.A.SSRI69
9Wang XC et al. (2014) [18]10918929821979BDNFrs62650.001N.A.N.A.N.A.Paroxetine69
GDNFrs29730490.005
rs22167110.005
10Han KM et al. (2013) [19]1381943719CYP2D6rs10658520.0010.001N.A.N.A.Escitalopram129
11Shima Sahraian et al. (2013) [20]26781046539SLC6A45-HTTLPR0.023N.A.N.A.N.A.Citalopram148
12Liu Z et al. (2013) [21]721131859887PDLIM5rs24333200.0145N.A.N.A.N.A.Fluoxetine69
13Mitjans M et al. (2013) [22]351201559651CNR1rs8063680.0290.021N.A.N.A.Citalopram129
rs8063710.0450.016
rs8063770.1880.043
14Myung W et al. (2013) [23]3454884642SLC6A45-HTTLPR0.004N.A.N.A.N.A.Sertraline, Fluoxetine69
15Wang Y et al. (2012) [24]18222140328778DRD2rs27348330.0445N.A.N.A.N.A.SSRI68
16Yang Z et al. (2012) [25]18222140313035APCrs2229992N.A.0.05N.A.N.A.SSRI68
SRP19rs4957940.0011
REEP5rs1535490.0015
rs1535600.0009
17Xu Z et al. (2012) [26]12118730811452HTR1Brs62980.023N.A.N.A.0.39 (0.17–0.91)SSRI69
18Illi A et al. (2011) [27]3649852956SLC6A45-HTTLPR0.03N.A.N.A.N.A.Citalopram, Fluoxetine, Paroxetine69
19Kishi T et al. (2010) [28]121144265150115HTR2Ars19280400.0540.0252N.A.N.A.Fluvoxamine, Sertraline, Paroxetine87
20Kishi T et al. (2010) * [28]121144265150115HTR2Ars19280400.09100.0418N.A.N.A.Fluvoxamine, Sertraline, Paroxetine87
21Liou YJ et al. (2009) [29]18626344942117KCNK2rs66677640.0460.360N.A.N.A.Citalopram, Fluoxetine89
rs104949940.050.082
rs66865290.0190.0008
22Min W et al. (2009) [30]272307579243119SLC6A45-HTTLPR0.0320.617N.A.N.A.SSRI69
23Kishi T et al. (2009) [31]60611216061CLOCKrs37365440.00430.0026N.A.N.A.Fluvoxamine89
24Kishi T et al. (2009) * [31]60611216061CLOCKrs37365440.00650.0026N.A.N.A.Fluvoxamine89
rs37494740.0730.025
25Tsai SJ et al. (2009) [32]20830050812661TPH2rs21713630.0090.512N.A.N.A.Fluoxetine, citalopram87
rs42902700.0190.459
26Arias B et al. (2009) [33]114331479652DTNBP1rs7607610.030.007N.A.N.A.Citalopram48
27Wong ML et al. (2008) [34]3771108--CYP3A4rs2242480N.A.0.02N.A.N.A.Fluoxetine87
PSMD13rs38176290.04
CD3Ers22314490.002
PRKCSHrs1608410.02
PSMA7rs20571690.004
rs20571680.003
rs22817400.002
rs37466510.01
28Tsai SJ et al. (2008) [35]1011292307492GSK3Brs3345580.0020.02N.A.N.A.Fluoxetine, Citalopram48
rs133217830.0020.002
rs23193980.0110.011
29Gau YT et al. (2008) [36]1001282287443NGFRrs20724460.0390.012N.A.N.A.Fluoxetine, Citalopram89
30Bozina N et al. (2008) [37]69611306565SLC6A45-HTTLPR0.0050.0004N.A.N.A.Paroxetine69
31Papiol S et al. (2007) [38]351241599551CRHR2rs22700070.0180.002N.A.N.A.Citalopram48
32Ham BJ et al. (2007) * [39]29761054263TPH1rs18005320.0470.017N.A.N.A.Citalopram89
33Choi MJ et al. (2006) [40]2459835726COMTrs62650.0120.009N.A.N.A.Citalopram87
34Hong CJ et al. (2006) [41]9313122481143HTR1Ars62950.009N.A.N.A.N.A.Fluoxetine48
SLC6A45-HTTLPR0.001
35Choi MJ et al. (2005) * [42]5120712249HTR2Ars63110.0180.034N.A.N.A.Citalopram48
36Kraft JB et al. (2005) [43]4947967719SLC6A4rs25531N.A.0.03N.A.N.A.Fluoxetine129
37Yu YW et al. (2003) * [44]67901574115IL-1Brs1939224900.028N.A.N.A.N.A.Fluoxetine47
38Yoshida K et al. (2002) [45]2232543519SLC6A45-HTTLPR0.0590.01N.A.N.A.Fluvoxamine69
39Yin L et al. (2016) [46]14115129022070DRD4rs18005440.030.41N.A.N.A.Fluoxetine, paroxetine, sertraline, citalopram69
40Hun Soo Chang et al. (2011) [47]16991154925BDNFrs62650.0010.006N.A.N.A.Escitalopram89
41Lin KM et al. (2010) * [48]3620524169102CYP1A2rs46464250.0020.032.3 (1.12–4.73)N.A.Paroxetine89
rs24723040.0240.010.39 (0.19–0.82)
rs24708900.0150.0040.34 (0.16–0.74)
42Lee SH et al. (2010) * [49]1747643529MRP1rs22393300.0380.005N.A.N.A.Citalopram89
rs2120870.1940.052
rs2120900.1330.035
43Tsai SJ et al. (2009) [50]13819633410152COMTrs46800.020.006N.A.N.A.Fluoxetine, citalopram89
44Yu YW et al. (2006) [51]9412822283139HTR1Ars62950.007N.A.N.A.N.A.Fluoxetine48
45Suzuki Y et al. (2004) [52]2923523517HTR1Ars18000420.042N.A.N.A.N.A.Fluvoxamine129
46Jamerson BD et al. (2013) [53]44601045549MTRRrs18013940.0077N.A.N.A.N.A.SSRI128
MTHFRrs18011310.0313
47Fabbri C et al. (2013) [54]59894315412601281GRM7rs10838010.0000005N.A.N.A.N.A.Citalopram28
GRIK2rs5995450.0003
rs27862470.0008
rs28525840.0002
rs25183130.0003
rs27862390.0006
GRIA4rs4954980.0008
rs107917730.0009
rs9945750.0009
rs112268560.0002
PRKCErs5053100.0005
CAMK2Drs125085660.0009
48Glubb DM et al. (2010) [55]N.A.N.A.2854719ADMrs110427250.001N.A.N.A.N.A.Paroxetine66
49Peters EJ et al. (2009) [56]74612071953N.A.N.A.HTR2Ars1923884N.A.0.02N.A.0.75 (0.58–0.97)Citalopram69
rs7997012N.A.0.0002N.A.1.43 (1.13–1.81)
50Peters EJ et al. (2009) * [56]74612071953N.A.N.A.HTR2Ars1923884N.A.0.01N.A.0.72 (0.55–0.95)Citalopram69
rs7997012N.A.3.0 × 10−5N.A.1.52 (1.20–1.95)
51Mrazek DA et al. (2009) * [57]4436311074104232SLC6A4SERTin20.041N.A.N.A.N.A.Citalopram69
5-HTTLPR0.039
52Kato M et al. (2006) [58]44561005723SLC6A45-HTTLPR0.043N.A.N.A.N.A.Paroxetine, Fluvoxamine68
53McMahon FJ et al. (2006) * [59]74812051953N.A.N.A.HTR2Ars79970120.000042.0 × 10−5N.A.N.A.Citalopram69
rs19280400.07010.0446
54McMahon FJ et al. (2006) [59]74812051953N.A.N.A.HTR2Ars79970122.0 × 10−64.0 × 10−5N.A.N.A.Citalopram69
rs19280400.01490.0709
55Peters EJ et al. (2004) [60]4749967719SLC614rs255330.037N.A.0.33 (0.08–1.35)N.A.Fluoxetine127
56Ji Y et al. (2012) [61]N.A.N.A.1232541691COMTrs13306278N.A.0.04N.A.0.78 (0.62–0.99)SSRI67
rs9332381N.A.0.0061.71 (1.16–2.51)
57Lekman M et al. (2008) [62]74812051953954415FKBP5rs47139160.00270.0007N.A.N.A.Citalopram67
58Lekman M et al. (2008) * [62]74812051953723466FKBP5rs47139160.0420.042N.A.N.A.Citalopram67
59Kraft JB et al. (2007) [63]73511791914991669SLC6A4rs255330.05N.A.1.81 (0.92–3.56) Citalopram69
60Binder EB et al. (2010) [64]74612071953982726CRHBPrs104739840.00680.0044N.A.1.42 (1.11–1.81)Citalopram69
rs104744850.0180.00651.25 (1.06–1.46)
rs100552550.0200.0171.19 (1.04–1.39)
CRHR2rs22677160.0240.0131.20 (1.04–1.38)
rs2551050.0430.00861.20 (1.05–1.38)
CRHR1rs129423000.0380.00861.31 (1.07–1.60)
61Binder EB et al. (2010) * [64]74612071953740649CRHBPrs104739840.00040.0006N.A.N.A.Citalopram69
rs104744850.0180.0062
rs100552550.0050.0023
CRHR1rs129423000.00870.0015
CRHR2rs22677160.0240.0098
AVPR1Ars73079970.0470.038
62Garriock HA et al. (2010) [65]74612071953531790OPRM1rs562859N.A.0.002N.A.1.33 (1.05–1.69)Citalopram69
rs1323044N.A.0.0031.46 (1.15–1.85)
rs540825N.A.0.0031.37 (1.07–1.75)
rs658156N.A.0.0031.54 (1.18–2.00)
rs13195018N.A.0.0021.60 (1.23–2.08)
rs538174N.A.0.0021.57 (1.20–2.05)
rs583664N.A.0.0011.61 (1.23–2.10)
rs618207N.A.0.0011.47 (1.13–1.91)
63Garriock HA et al. (2010) * [65]74612071953531669OPRM1rs562859N.A.0.002N.A.1.36 (1.06–1.74)Citalopram69
rs1323044N.A.0.0051.44 (1.12–1.85)
rs540825N.A.0.0021.44 (1.12–1.86)
64Lin KM et al. (2011) [66] * 19811004826ABCB1rs1882478N.A.0.037N.A.0.35 (0.17–0.71)Escitalopram88
rs10456420.045N.A.0.34 (0.16–0.72)
rs102568360.021N.A.3.82 (1.58–9.22)
N.A., data not available; M, male; F, female; FP, Follow-up period in weeks; OR, odds ratio; CI, confidence interval; Score, cumulative score for methodological quality assessment (see Tables S1 and S2 for detailed scoring) * Study name shows remission data. OR calculated using reported frequencies from the respective article. All the studies represent significant polymorphisms (p ≤ 0.05) with their corresponding genes.
Table 2. Characteristics of included genome-wide association studies concerning SSRI response in major depression patients.
Table 2. Characteristics of included genome-wide association studies concerning SSRI response in major depression patients.
No.Study NameStudy Population Responders (n)Remitters (n)Non-Remitters (n)Non-Responders (n)Total (n)Genotyping PlatformSNPs Studied (n)
1Myung W et al. (2015) [67]Korean497312558373870Affymetrix Genome-Wide Human Single-Nucleotide Polymorphism (SNP) Array Chip 6.0905,431
2Biernacka JM et al. (2015) [68]Asian, European416226190449865Illumina Human—Omni Express Exome Bead Chips631,765
3Hunter AM et al. (2013) [69]European869N.A.N.A.2471116Affymetrix 500 K and 5.0 Human SNP Arrays430,198
Replication data set706Illumina Human 610 Quad Bead Chip550,337
4Tansey KE et al. (2012) [70]EuropeanN.A.N.A.N.A.N.A.2283Illumina Human 610 Quad Bead Chips; Illumina Human 660 W-Quad Bead Chips520,978
5Ji Y et al. (2013) [71]European28720681212499Illumina Human 610-Quad Bead Chips550,337
6Sasayama D et al. (2013) [72]Japanese61N.A.N.A.3192Illumina Human CNV370 Quad Bead Chips356,075
Replication data set136
7Uher R et al. (2010) [73]EuropeanN.A.N.A.N.A.N.A.706Illumina Human 610 Quad Bead Chips550,337
M, male; F, female. All the GWAS were performed on MDD patients having SSRIs as an antidepressant.
Table 3. Characteristics of genome-wide association studies of major depressive disorder.
Table 3. Characteristics of genome-wide association studies of major depressive disorder.
No.Study NameStudy Population Cases (n)Controls (n)Total (n)PlatformSNPs Studied (n)
1Power RA et al. (2013) [74]European8058051610Illumina 610 K bead array457,670
2Ripke S et al. (2013) [75]European9240951918,759Illumina 610 K, 317 K, 370 K, 550 K, Perlegen 600 K, Affymetrix 6.0>200,000
678350,69557,478N.A.593
3Wray NR et al. (2012) [76]European243136736104Illumina 317 K, Illumina 370 K, Illumina 610 K, Affymetrix 600 K657,366
4Shi J et al. (2012) [77] European102016362656Affymetrix 6.0671,424
5Shyn SI et al. (2011) [78]European122116362857Affymetrix 6.0, 5.0 and 500 K, and Perlegen500,568
6Muglia P et al. (2010) [79]European102210002022Illumina HumanHap550551,101
49210521544Affymetrix 5.0370,697
7Sullivan PF et al. (2009) [80]European173818023540Perlegen435,291
8Rietschel M et al. (2010) [81]European60413641968Illumina HumanHap 550v3, and Illumina Human 610 W Quad Bead Chips491,238
409541950
All the GWAS were performed on MDD patients vs. healthy controls.

Share and Cite

MDPI and ACS Style

Srivastava, A.; Singh, P.; Gupta, H.; Kaur, H.; Kanojia, N.; Guin, D.; Sood, M.; Chadda, R.K.; Yadav, J.; Vohora, D.; et al. Systems Approach to Identify Common Genes and Pathways Associated with Response to Selective Serotonin Reuptake Inhibitors and Major Depression Risk. Int. J. Mol. Sci. 2019, 20, 1993. https://doi.org/10.3390/ijms20081993

AMA Style

Srivastava A, Singh P, Gupta H, Kaur H, Kanojia N, Guin D, Sood M, Chadda RK, Yadav J, Vohora D, et al. Systems Approach to Identify Common Genes and Pathways Associated with Response to Selective Serotonin Reuptake Inhibitors and Major Depression Risk. International Journal of Molecular Sciences. 2019; 20(8):1993. https://doi.org/10.3390/ijms20081993

Chicago/Turabian Style

Srivastava, Ankit, Priyanka Singh, Hitesh Gupta, Harpreet Kaur, Neha Kanojia, Debleena Guin, Mamta Sood, Rakesh Kumar Chadda, Jyoti Yadav, Divya Vohora, and et al. 2019. "Systems Approach to Identify Common Genes and Pathways Associated with Response to Selective Serotonin Reuptake Inhibitors and Major Depression Risk" International Journal of Molecular Sciences 20, no. 8: 1993. https://doi.org/10.3390/ijms20081993

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop