Open Access

Association of HHV‑6 reactivation and SLC6A3 (C>T, rs40184), BDNF (C>T, rs6265), and JARID2 (G>A, rs9383046) single nucleotide polymorphisms in depression

  • Authors:
    • Sureewan Bumrungthai
    • Surachat Buddhisa
    • Sureewan Duangjit
    • Supaporn Passorn
    • Sasiwimon Sumala
    • Nattaphol Prakobkaew
  • View Affiliations

  • Published online on: October 3, 2024     https://doi.org/10.3892/br.2024.1869
  • Article Number: 181
  • Copyright : © Bumrungthai et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Major depressive disorder (MDD) is a global health concern with a complex etiology involving genetic, environmental and infectious factors. The exact cause of MDD remains unknown. The present study explored the association between genetic factors, human herpesvirus 6 (HHV‑6) and MDD. The present study analyzed single nucleotide polymorphisms (SNPs) and HHV‑6 viral load in oral buccal samples from patients with MDD (with and without blood relatives with MDD) and healthy controls. The study used high‑resolution melt analysis to examine rs40184 (C>T) in the solute carrier family 6 member 3 (SLC6A31) gene, rs6265 (C>T) in the brain‑derived neurotrophic factor (BDNF) gene and rs9383046 (G>A) in the jumonji and AT‑rich interaction domain‑containing 2 (JARID2) gene. HHV‑6 infection and viral load was assessed using the quantitative PCR. Whole‑exome sequencing was used to examine SNPs. The variant alleles of SNPs rs40184 [18/40 (45.00) vs. 29/238 (12.55%)] and rs6265 [30/54 (55.46) vs. 117/292 (40.06%)] were significantly more common in patients with MDD than in healthy controls, indicating they may be probable hereditary risk factors for MDD. HHV‑6 positivity was significantly more common in carriers of the G/A genotype (12/15, 80%) than carriers of the G/G genotype (75/363, 20.7%) for rs9383046, implying that genetic variations may affect HHV‑6 risk and MDD onset. Similarly, HHV‑6 viral loads were significantly higher in carriers of the G/A genotype (99,990.85±118,392.64 copies/ng DNA) than carriers of the G/G genotype (48,249.30±101,216.28 copies/ng DNA) for rs9383046. Whole‑exome sequencing identified two SNPs in JARID2 (rs11757092 and rs9383050) associated with MDD, highlighting its genetic complexity. The present study helps explain the complex interactions between HHV‑6 infection, genetics and MDD onset, improving understanding of how SNPs in JARID2 contribute to HHV‑6 infection and MDD onset; these findings may impact future approaches to diagnosing and treating MDD.

Introduction

Major depressive disorder (MDD) is a global health concern that can lead to suicide. The World Health Organization estimates that 350 million individuals worldwide have MDD (1). MDD is common in Thailand and its incidence varies by age group; studies have reported rates of 7.0-21.4% in individuals aged 19-22 years, 12.0% in individuals >30 years, 39.10% in those >40 years, 9.8-29.2% in those >45 years and 6.5-18.5% in those >47 years (2-7). Its etiology is not entirely known. Many factors are associated with MDD, including viral infection that causes inflammation in the brain, abnormal hormone, neurotransmitter and proinflammatory cytokine levels and genetic factors (8-10).

Acute and latent infection can affect behavior and mental health. Increased proinflammatory cytokine levels and decreased serotonin and norepinephrine levels cause an inflammatory reaction in response to viral infection of microglia, oligodendrocytes and astrocytes (11,12). Previous research has revealed that elevated proinflammatory cytokine levels are associated with MDD and infection by the varicella zoster, Epstein-Barr, cytomegalovirus, Borna disease and herpes simplex viruses (12-16). Human herpes virus 6 (HHV-6) is a β-HV subfamily of HV family that can lead to chromatin modification and DNA methylation in target cells (17). Several studies have identified HHV-6 as a risk factor for MDD (16,18-21).

MDD is a complex mental disorder influenced by environmental, genetic and epigenetic factors. Genome-wide association studies (GWASs) have identified 178 genetic risk loci and >200 candidate genes (22-24). There is an association between epigenetic/genetic changes and aberrant neuromediators (serotonin, norepinephrine and dopamine) that can lead to MDD (1,25-27). These effects are hypothesized to be mediated by the metabolic disturbance of brain-derived neurotrophic factor (BDNF) in nerve tissue. The single nucleotide polymorphism (SNP) rs6265 in BDNF changes a valine to a methionine at codon 66 (Val66Met), which decreases activity-dependent release of BDNF and is associated with low BDNF levels and MDD pathogenesis (28-30). The dopaminergic system, including dopamine-producing cells, receptors and transporters, may serve a key role in MDD. Solute carrier family 6 member 3 (SLC6A3) regulates dopamine levels in the brain by inhibiting synaptic activity and inducing dopamine reuptake into presynaptic neurons (31-35). In addition, jumonji and AT-rich interaction domain containing 2 (JARID2) is a DNA-binding protein that regulates gene expression by modifying chromatin. SNP rs9383046 in JARID2 has been identified as a potential candidate gene for schizophrenia (36-38).

The interaction between several variables, including viral infection and genetic characteristics, may serve an important role in MDD onset. The present study investigated the association between HHV-6 infection and specific genetic factors [SLC6A3 (g.1394961C>T, rs40184), BDNF (g.27658368C>T, rs6265) and JARID2 (g.15281336G>A, rs9383046)] to elucidate their role in MDD onset. High-resolution melt analysis (HRM) for genetic analysis was performed, along with HHV-6 infection data and viral load assessment from a previous study (39). SNPs were assessed using whole-exome sequencing.

Materials and methods

Specimens

The present study, approved by the Committee on Human Research Ethics (approval nos. UP-HEC 1.3/013/65 and UBU-REC-68/2567), analyzed 471 buccal cell samples from a previous study (39). The MDD samples were collected using mail-in submissions following social media advertising in Thailand. A total of 376 female and 95 male participants were enrolled between July 2022 and June 2023. The participants included 360 healthy individuals, 59 MDD patients, 36 blood relatives of MDD patients, and 16 non-blood relatives of MDD patients. All participants' HHV-6 status was confirmed with quantitative PCR. The study specifically targeted patients aged 18-45 years, diagnosed based on the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition (40). Both recurrent MDD and first-episode cases were included. Certified hospital psychiatrists confirmed diagnoses using the Patient Health Questionnaire-9 (PHQ-9), with a score of ≥9 indicating MDD (39). Patients with MDD exhibited various forms of MDD, including recurrent episodes, acute phase and persistent MDD. Notably, patients with MDD did not present with comorbid conditions such as diabetes, hypertension, chronic obstructive pulmonary disease or coronary artery disease. Family members of patients with MDD were recruited to determine potential risk factors for the condition. Healthy controls were individuals with no present or lifetime psychiatric problems, PHQ-9 score <9 and no congenital disease (39). First-degree relatives of individuals with suicidal ideation or MDD diagnosis were excluded from the control group.

DNA extraction

DNA was extracted from buccal cells using the Genomic DNA Isolation kit (cat. no. PDC11-0100; Bio-Helix Co.) following the manufacturer's protocol as previously described (39).

HHV-6 DNA detection by quantitative (q)PCR and viral load

qPCR-based HHV-6 DNA (U97 gene) status and viral load data were taken from a previous study (39).

SNP detection by HRM

SNPs in SLC6A3 (g.1394961C>T, rs40184), JARID2 (g.15281336G>A, rs9383046) and BDNF (g.27658368C>T, rs6265) were detected by HRM analysis with amplicon sizes of 100, 88 and 146 bp, respectively. Table I shows primer sequences used for HRM analysis. PCR was performed in duplicate using the 5X FiREPOL Eva Green HRM Mix Plus (Solis Bio Dyne) as previously described (39,41,42). Positive controls for SLC6A3 (C/T, rs40184), JARID2 (G/A, rs9383046), and BDNF (C/T, rs6265) were utilized following sequencing confirmation. The primers used for HRM analysis are listed in Table I.

Table I

Primers used for HRM analysis and sequencing.

Table I

Primers used for HRM analysis and sequencing.

GeneSequence, 5'→3'Size, bp(Refs.)
BDNF (rs6265)Forward: CTTGACATCATTGGCTGACACT146(41)
 Reverse: GCTCCAAAGGCACTTGACTACT  
DAT1 (rs40184)Forward: CACAGTCTCGCGGCTTTT100(41)
 Reverse: TGGACCAACACACCCTTGA  
JARID2 (rs9383046)Forward: ACTGGCTGTGTCTCACTCTT88(42)
 Reverse: TATTCACGTTCTTTTGCTCTTGGA  
JARID2 (rs9383046)aForward: ATTTGACCCACACTGGCTGT96NA
 Reverse: TCACGTTCTTTTGCTCTTGGAC  

[i] aUsed for sequencing; the same DAT1 and BDNF primers were used for both HRM analysis and sequencing. BDNF, brain-derived neurotrophic factor; DAT1, dopamine transporter 1; JARID2, jumonji and AT-rich interaction domain containing 2; HRM, high resolution melt; NA, not applicable.

DNA sequencing

Sanger sequencing was performed to confirm the SNPs in SLC6A3 (g.1394961C>T, rs40184), JARID2 (g.15281336G>A, rs9383046), and BDNF (g.27658368C>T, rs6265). A total of 10 samples were analyzed for BDNF, 45 for SLC6A3, and 15 for JARID2, randomly selected from different patterns of HRM. The primers used for sequencing are listed in Table I. The sequences were analyzed by comparison with GenBank reference sequences, BDNF (accession no. NC_000011.10:27658340-27658399) on Homo sapiens chromosome 11, DAT1 (accession no. NC_000005.10:1394924-1394997) on chromosome 5 and JARID (accession no. NC_000006.12:15281287-15281391) on chromosome 6] using BioEdit version 7.2; bioedit.software.informer.com/7.2/).

Whole-exome sequencing

Whole-exome sequencing data to identify SNPs in samples taken from patients with MDD, blood relatives and the healthy controls was obtained from a previous study (39).

Statistical analysis

Data were analyzed using IBM SPSS software (Version 16.0, SPSS Inc.). Categorical variables were compared using Pearson's χ2 test. Continuous variables, reported as the mean ± standard deviation of ≥2 independent experimental repeats were compared between two groups using independent Student's or unpaired t-test and between more >2 groups using a one-way analysis of variance followed by LSD test or Median or Mann-Whitney U test. P<0.05 was considered to indicate a statistically significant difference.

Results

Association between SLC6A3 (g.1394961C>T, rs40184), BDNF (g.27658368C>T, rs6265), and JARID2 (g.15281336G>A, rs9383046) SNPs with MDD

SNPs were detected in healthy controls, patients with MDD and their blood and non-blood relatives using HRM analysis (Tables II and III). The variant T allele of rs40184 in SLC6A3 was significantly more common in patients with MDD (18/40, 45%) than in healthy controls [29/231, 12.6%; odds ratio (OR), 5.699, 95% confidence interval (CI), 2.734-11.879. Similarly, the variant T allele of rs6265 in BDNF was significantly more common in patients with MDD (30/54, 55.56%) than in healthy controls (117/292, 40.06%; OR, 1.870, 95% CI, 1.041-3.358). However, the frequency of variant A allele of rs9383046 in JARID2 did not differ significantly between patients with MDD and healthy controls.

Table II

Allele distribution.

Table II

Allele distribution.

GeneAllelenHealthy controls (%)Patients with MDD (%)Blood relatives (%)Non-blood relatives (%)P-value
DAT1T5229 (12.6)18 (45.0)4 (15.4)1 (7.7)<0.001
 C258202 (87.4)22 (55.0)22 (84.6)12 (92.3) 
BDNFT161117 (40.1)30 (55.6)8 (40.0)6 (50.0)0.034
 C217175 (59.9)24 (44.4)12 (60.0)6 (50.0) 
JARID2A1514 (4.7)0 (0.0)1 (4.2)0 (0)0.417
 G368283 (95.3)47 (100.0)23 (95.8)11(100) 

[i] DAT1, dopamine transporter 1; BDNF, brain-derived neurotrophic factor; JARID2, jumonji and AT-rich interaction domain containing 2; MDD, major depressive disorder; N, numbers.

Table III

Allele distribution in patients with MDD and the healthy controls.

Table III

Allele distribution in patients with MDD and the healthy controls.

GeneGroupnT (%)C (%)P-valueOR (95% CI)
DAT1MDD4018(45)22(55)<0.0015.699
 Healthy23129 (12.55)202 (87.45) (2.734-11.879)
BDNFMDD5430 (55.56)24 (44.44)0.0341.870
 Healthy292117 (40.06)175 (59.93) (1.041-3.358)
   AG  
JARID2MDD470 (0)47(100)0.129NA
 Healthy29714 (4.71)283 (95.29)  

[i] DAT1, dopamine transporter 1; BDNF, brain-derived neurotrophic factor; JARID2, jumonji and AT-rich interaction domain containing 2; MDD, major depressive disorder; NA, not applicable.

HHV-6 infection status was associated with SNP rs9383046 (G>A) in JARID2 but not with SNP rs40184 (C>T) in SLC6A3 or SNP rs6265 (C>T) in BDNF (Fig. 1; Table IV). The A allele of SNP rs9383046 in JARID2 was significantly more common in individuals with HHV-6 infection (80.0%, or 12 out of 15) than in those with the wild-type G allele (75/363, 20.7%; OR, 15.360, 95% CI, 4.226-55.823). This suggests that SNP rs9383046 (G>A) in JARID2 may be associated with HHV-6 infection.

Table IV

Association between HHV-6 status and single nucleotide polymorphism alleles.

Table IV

Association between HHV-6 status and single nucleotide polymorphism alleles.

  HHV-6 status 
GeneAllelenPositive (%)Negative (%)P-value
DAT1T5115 (29.4)36 (70.6)0.916
 C25874 (28.7)184 (71.3) 
BDNFT16136 (22.4)125 (77.6)0.578
 C21543 (20.0)172 (80.0) 
JARID2A1512 (80.0)3 (20.0)<0.001
 G36375 (20.7)288 (79.3) 

[i] DAT1, dopamine transporter 1; BDNF, brain-derived neurotrophic factor; JARID2, jumonji and AT-rich interaction domain containing 2.

High HHV-6 viral load associated with JARID2 SNP (g.15281336G>A)

Our previous study used qPCR to determine viral load in HHV-6-positive cases (39). HHV-6 loads were significantly higher in individuals with the G>A genotype (99,990.85±118,392.64) than in those with the G genotype (48,249.30±101,216.28 copies/ng DNA) for SNP rs9383046 in JARID2 (Fig. 2).

JARID2 SNP in MDD

Whole-exome sequencing was used to analyze the point mutation status of samples from four participants: Patient with MDD, their first-degree relative and a healthy female and male, all aged between 21 and 30 years. The sample was randomly selected from patients with MDD who had a first-degree (healthy) relative in the same age group. The whole-exome sequencing results are shown in Fig. 3, revealing 484 SNPs in JARID2 in MDD. Previous literature reviews have reported that the following genes are associated with JARID2 and its associated pathways: AE Binding Protein 2 (AEBP2), AT-rich interaction domain (ARID), AT-rich sequences (AT-rich), ataxin 1 (ATXN1), brain-derived neurotrophic factor (BDNF), cluster of differentiation 82 (CD82), cadherin 13 (CDH13), cyclin D1 (CCND1), D-Amino acid oxidase activator (DAOA), differentially methylated regions (DMRs), dystrobrevin-binding protein 1 (DTNBP1), enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2), histone 3 lysine 27 (H3K27), histone methyltransferases, homeobox A cluster (HOXA), IFN-γ), isoflavone reductase (IFR), IL-18), IL-6), Jumonji domain-containing protein-3 (JMJD3), lysine acetyltransferase 5 (KAT5), Lysine Demethylase 2B (KDM2B), Neuregulin 1 (NRG1), PHD finger protein 1 (PHF1), polycomb repressive complex 2 (PRC2), RB Binding Protein 4 (RBBP4), RB transcriptional corepressor like 2 (RBL2), regulator of G protein signaling 4 (RGS4), Sirtuin (SIRT5), SLC6A3, SMAD family member 2 (SMAD2), SOX, STAT3, transcription factor AP-2α (FAP2A), transforming growth factor beta (TGF-ß), Vascular endothelial growth factor (VEGF) and zinc finger E-box binding homeobox 1 (ZEB) (36,37,42-44). There was a total of two SNPs each in JARID2 (rs11757092 and rs9383050) and JMJD4 (rs2295994 and rs7419238) and three SNPs in BDNF (rs2242341, rs2353512, and rs10835210; Table V).

Table V

Association of single nucleotide polymorphisms in JARID2 and genes in associated pathways.

Table V

Association of single nucleotide polymorphisms in JARID2 and genes in associated pathways.

Reference alleleAlternative alleleGeneRSTarget gene
TCAEBP2rs56666201AE Binding Protein 2
AGAEBP2rs4369485 
AGAEBP1rs2595701 
TAAEBP1rs11979310 
GAAEBP1rs41279612 
AGAEBP1rs13928 
AGARID4Brs4659654A-T Rich Interaction Domain
TCARID4Brs12731746 
ACARID4Brs7518225 
TCARID4Brs139681730 
GAARID5Brs201704836 
GAARID2rs2059404 
CGATXN2rs2301622Ataxin 1
CTATXN2rs2301621 
AGATXN2rs2285518 
CTATXN2rs7969300 
AGATXN2rs593226 
TGATXN3rs3092822 
GTATXN3rs7158733 
GAATXN3rs761552 
CTATXN3rs1048755 
TCATXN3rs2268007 
TCATXN3rs8003041 
GCATXN3rs4904834 
GAATXN2Lrs12920514 
AGATXN2Lrs4344749 
TAATXN2Lrs7498491 
CTATXN2Lrs35725751 
CTATXN7rs117130898 
CTATXN7rs3733125 
GAATXN7rs3774729 
GAATXN1rs16885 
GAATXN1rs2072737 
TCATXN1rs2075974 
AGATXN1rs179990 
GAATXN7L1rs940370 
CTATXN3Lrs4830842 
GCBDNF-ASrs2242341Brain-derived neurotrophic factor
TCBDNFrs2353512 
CABDNF-ASrs10835210 
CTBDNFrs13306221 
GACD82rs2303865CD82
AGCD82rs1139971 
CGCD82rs7107335 
AGCDHR4NACadherin-13
TACDHR4rs143469719 
GACDHR4rs76282326 
GAPCDH7rs28481709 
GAPCDH7rs28387015 
GAPCDH7rs73216844 
GAPCDH7rs1047012 
GAPCDH7rs977931 
GCPCDH7rs4580617 
AGPCDH7rs10006845 
TGPCDH7rs16867990 
GAPCDH18rs144391842 
TCPCDH18rs151070417 
AGPCDH18rs10018837 
CGPCDH18rs10006580 
GAPCDH18rs28566714 
CTCDH18rs73760020 
TCCDH18rs12187552 
AGCDH12rs6451992 
AGCDH12rs6451993 
CTCDH10rs62349562 
CTCDH9rs116872912 
CTCDH6rs2287582 
CTCDH6rs2302904 
AGCDH6rs2302903 
TCCDH6rs2229575 
CTCCND1rs3862792Cyclin D1
GACCND1rs9344 
AGCCND1rs3212892 
GADAOArs2391191D-amino acid oxidase activator
TCDMRTC2rs2305809Differentially methylated region MRs
GCDMRT1rs279895 
TCDMRT3rs6477419 
GADMRT2rs202027041 
GCDMRT2rs3824419 
CGDMRTA1rs201444816 
CTDMRTC1Brs201167658 
CTDTNBP1rs4236167Dystrobrevin binding protein 1
GTDTNBP1rs6926401 
TCDTNBP1rs7758659 
AGDBNDD2rs1127497 
AGEZH2rs740949Enhancer of zeste 2 polycomb repressive complex 2
ACEZH2rs41277434 
AGEZH2rs2072407 
CGEZH2rs2302427 
AGEZH2rs10274535 
TCCHD5rs6696489Methylation of histone H3
CACHD5rs59788818 
AGCHD5rs2843493 
AGCHD5rs2273041 
GACHD5rs55930553 
GCCHD5rs2273033 
CTCHD5rs2235790 
TGCHD5rs3765452 
AGCHD5rs2250358 
AGCHD5rs2746066 
TACHD5rs2785582 
ACCHD5NA 
CGCHD5rs17489787 
AGCHD5rs9435102 
TCCHD5rs12565328 
GACHD5rs9434711 
CGCHD5rs12074369 
CTPRMT6rs2232016Histone methyltransferase
TCASH1Lrs4971053 
ACASH1Lrs10908466 
GARBBP5rs11240356 
GARBBP5rs7515178 
GASMYD2rs6540819 
GASMYD2rs1134647 
TGSMYD2rs2291830 
TCSMYD2rs2270704 
CTSMYD2rs1874804 
GASETD2rs2290547 
GASETD2rs4082155 
AGSETD2rs6767907 
CGSETD2rs6442059 
AGNSD2rs489550 
GCHOXA1NAHomeobox A cluster
GAHOXA1rs778380747 
TCHOXA1NA 
TGHOXA1NA 
TCHOXA1NA 
CTHOXA1NA 
CTHOXA1rs10951154 
GAHOXA1rs577426612 
TCHOXA4rs17449108 
ACHOXA4rs6957209 
AGHOXA-AS3rs62454420 
ACHOXA7rs2301720 
CTHOXA7rs2301721 
AGHOXA9NA 
AGIFNLR1rs946671Interferon-γ
TCIFNLR1rs4649195 
TCIFNLR1rs7552000 
TCIFNAR2rs2834158 
AGIFNAR2rs2252639 
CTIFNAR2rs9984273 
AGIFNGR2rs9808753 
CGIFNGR2rs11910627 
TCIFNGR2rs1532 
AGIFNGR1rs1887415 
ACIFNGR1rs11914 
GAIFNGR1rs1327475 
CGIFNGR1rs9376269 
AGIFNGR1rs2234711 
GAIFNB1rs1051922 
AGIFNA21rs2939 
AGIFNA4rs3750479 
TAIFNA4rs3750480 
CTIFNA4rs1062571 
TCIFNA7rs76644201 
GTIFNA10rs56035072 
TCIFNA16rs3919593 
ACIFNA17rs9298814 
AGIFNA17rs7025879 
GTIFNA17rs10117962 
GAIFNA5rs10757212 
GAIFNA6rs2988573 
AGIFNKrs700785 
GAIFRD2rs2229647Isoflavone reductase
AGIFRD2rs2071205 
AGIFRD2rs1076872 
CTIFRD1rs55884191 
TCIFRD1rs34349457 
CTIFRD1rs6968084 
TGIFRD1rs2253962 
GAIFRD1rs2074796 
ACIL18BPrs2298455Interleukin-18
CTIL18R1rs1035130 
AGIL18R1rs4851570 
AGIL18RAPrs1558651 
GAIL18RAPrs6708413 
GAIL6R; SHErs11265621Interleukin-6
AGIL6STrs4865999 
TAJARID2rs11757092Jumonji and AT rich interactive domain 2
AGJARID2rs9383050 
CTJARID2NA 
ACJMJD4rs2295994Jumonji domain-containing protein-3
GAJMJD4rs7419238 
CGJMJD1Crs1935 
GAJMJD1Crs376398448 
TAJMJD1Crs10740107 
GAJMJD1Crs3211105 
ATJMJD1Crs1904294 
GCJMJD1Crs41274074 
ATJMJD1Crs10761725 
TCJMJD1Crs7477425 
GCJMJD1C-AS1rs1061259 
AGJMJD1C-AS1rs10761770 
GCJMJD7rs2241523 
CGJMJD7rs2303516 
GAJMJD7rs890505 
TCJMJD7rs890504 
CGJMJD7rs11547012 
CGJMJD7rs2077543 
ACJMJD7rs1648835 
GCJMJD7rs1648834 
CTJMJD7rs1672466 
GAJMJD7rs1648829 
AGJMJD7rs1197670 
GAJMJD7rs890508 
CTJMJD8rs767035682 
AGJMJD8NA 
AGJMJD8NA 
GAJMJD8rs773965622 
GTKAT5rs2236682Lysine acetyltransferase
AGKAT5rs616250 
TCKAT5rs1151500 
TGKAT5rs487264 
AGKATNAL1rs202087 
TCKATNBL1rs74482733 
AGKAT8rs1549295 
AGKAT8rs1549294 
CTKAT8rs1549293 
AGKATNB1rs2967152 
CGKATNB1rs2967153 
CTKATNB1rs9938236Katanin regulatory subunit B1-like 1
CTKATNB1rs12922275 
GAKATNB1rs76370907 
CTKDM1Ars967605Lysine demethylase 2B
ACKDM1Ars2072945 
TCKDM1Ars2072944 
CAKDM4Ars586339 
GAKDM5Brs4310498 
GCKDM5Brs1141109 
GAKDM5Brs1141108 
CTKDM5Brs3196669 
GAKDM5Brs12028388 
TCKDM2Ars3741189 
AGKDM4Drs76057256 
GAKDM4Drs3740853 
GAKDM4Ers2020210 
AGKDM4Ers10752685 
AGKDM4Ers28412010 
AGKDM5ANA 
TGKDM5ANA 
AGKDM5ANA 
TCKDM5Ars771421847 
GAKDM5Ars2229351 
AGKDM5Ars11062385 
CTKDM5Ars4980885 
ATKDM5Ars7965303 
GAKDM2Brs12427382 
ACKDM2Brs11065575 
CTKDM2Brs1064951 
GCKDM2Brs2288154 
GAKDM2Brs3751131 
GAKDM2Brs10849885 
TGKDM2Brs28461264 
GAKDM8rs877585 
ACKDM8rs908382 
TGKDM8rs11645703 
AGKDM6Brs80152199 
CTKDM6Brs2270516 
TCKDM6Brs2270517 
CAKDM6Brs3744247 
CTKDM6Brs3744248 
CAKDM6Brs3736306 
TCKDM4Brs2240678 
TGKDM4Brs1017820 
CTKDM4Brs148048943 
AGKDM4Brs2620836 
GAKDM4Brs2613786 
AGKDM3Ars2030259 
GAKDM3Ars12714187 
GAKDM3Ars61748134 
TCKDM3Ars4832290 
AGKDM3Ars75816635 
CTKDM3Brs4835678 
TGKDM3Brs12522867 
GAKDM3Brs10073922 
GAKDM3Brs6865472 
AGKDM3Brs2269951 
TCKDM3Brs7726234 
AGKDM3Brs192834842 
CTKDM3Brs17599026 
TCKDM1Brs429158 
GAKDM1Brs214596 
TCKDM1Brs214585 
GAKDM7Ars12703533 
GTKDM7Ars6950119 
TGKDM7Ars1062277 
CGKDM7Ars59225858 
TCKDM4Crs7040131 
GAKDM4Crs10815499 
GAKDM4Crs2296067 
AGKDM4Crs35389625 
GTKDM4Crs818883 
GAKDM4Crs1570512 
CTKDM4Crs3763651 
GCKDM4Crs10758825 
CGKDM4Crs1407863Lysine demethylase 4C
CAKDM6Ars6611055 
CAKDM6Ars2230018 
GAKDM6Ars20539 
TCKDM5CNA 
TCKDM5CNA 
AGKDM5Crs1536247 
CAKDM5Crs1977364 
CGMIR4711NAMicroRNA 4740
GTMIR4711NA 
AGMIR4711rs111566161 
CAMIR4707rs2273626 
GAMIR4706rs2296320 
AGMIR4708rs12881755 
ACMIR4713HGrs4646 
TAMIR4713HGrs28757202 
GAMIR4713HGrs28757201 
ACMIR4713HGrs4324076 
TCMIR4713HGrs3759811 
CAMIR4713HGrs727479 
CTMIR4721rs548287959 
CGMIR4719rs34106659 
AGMIR4719rs58353328 
TCMIR4719rs7500280 
GAMIR4719rs7499278 
GCMIR4752rs4112253 
GTMIR4752rs375002929 
TAMIR4752rs3890103 
CTMIR4752rs7247101 
ACMIR4752rs7246998 
TCMIR4752rs7248086 
TCMIR4752rs7248089 
GCMIR4779rs77373668 
TCMIR4771-1rs866077379 
GAMIR4771-1rs4591335 
GAMIR4771-1rs200567153 
TCMIR4771-1rs4047215 
TGMIR4771-1rs2245238 
TCMIR4771-1rs1522044 
CAMIR4776-1;-2NA 
TGMIR4776-1;-2NA 
ACMIR4799NA 
GAMIR4799NA 
GANRG1rs3924999Neuregulin 1
CTNRG1rs57944175 
CANRG1rs3735776 
GASETDB2-PHF11rs2077848PHD finger protein 1
GTSETDB2-PHF11rs7996852 
GASETDB2-PHF11rs2057413 
AGSETDB2-PHF11rs11619265 
AGPHF11rs2031532 
AGPHF11rs3765526 
CGPHF11rs2274277 
GAPHF11rs1046295 
GAPHF1rs3116713 
GTPHF1rs3106196 
GAPHF10rs12663375 
CTPHF10rs7760142 
AGPHF10rs3807063 
GAPHF14rs218965 
GAPHF14rs2301960 
CAPHF14rs28394821 
TAPHF20L1rs2244885 
CTPHF20L1rs16904746 
CTPHF20L1rs756201 
GAPHF24rs2279790 
GAPHF24rs2279791 
GAPHF2rs10992812 
AGPHF2rs3763605 
TAPHF2rs10761251 
TCPHF2rs10992836 
GAPHF2rs3750359 
AGPHF2NA 
CTPHF2NA 
GAPHF2rs374331864 
ATPHF2NA 
TGPHF2NA 
TCPHF2NA 
GCPHF19rs3753029 
AGPHF19rs1056567 
GCPHF19rs4836833 
TCPHC2rs72658237Polycomb repressive complex 2
CAPHC2rs41265891 
AGPHC2rs6425816 
TCPHC2NA 
GAPHC2NA 
GAPHC2rs34710284 
TCRBBP4rs2762904RB binding protein 4
ACRBBP4rs359955 
AGRBBP4rs359956 
ACRBBP4rs607202 
TCRBBP4rs1320511 
CTRBBP4rs55707864 
TCRBBP6rs7195386 
TCRBBP8rs2336916 
TCRBBP8rs78439784 
AGRBBP9rs3748450 
AGRBBP9rs2424217 
AGRBBP9rs2247746 
ACRBBP8NLrs2236200 
ACRBBP7rs5924532 
CTRBBP7rs67984110 
CTREPS2rs73189116 
TAREPS2rs1946395 
AGRBL2rs555878756RB transcriptional corepressor like 2
GARGS4rs10917671Regulator of G protein signaling 4
TCRGSL1rs266508 
AGRGSL1rs7535533 
CARGSL1rs266534 
GCRGSL1rs6657620 
TCRGS16rs1144566 
TCRGS16rs509476 
TCRGS7rs2275742 
TARGS7rs4659599 
GCRGS6rs2238186 
CARGS6rs10131300 
TGRGS6rs2239277 
CGRGS6rs551152070 
ACRGS6rs4617784 
AGRGS6rs4346151 
AGRGS6NA 
GARGS6NA 
AGRGS6rs769852120 
AGRGS6NA 
ATRGS6rs11628539 
TCRGS11rs57268939 
CTRGS11rs74003728 
CGRGS11rs117215141 
AGRGS9rs2585858 
AGRGS9rs9896245 
CTRGS9rs12452285 
AGRGS9rs2292592 
CGRGS9BPrs3826926 
AGRGS12rs2236052 
CTRGS12rs10006362 
CTRGS12rs118013842 
GCRGS12rs3213507 
GARGS12rs374843920 
AGRGS12rs2269497 
AGRGS7BPrs889248 
TARGS7BPrs10939996 
TARGS17rs685449 
AGRGS22rs6468700 
CGRGS22rs3101322 
CTRGS22rs922207 
GARGS22rs2980542 
AGRGS22rs1471293 
TCRGS22rs2453626 
TCRGS22rs1460933 
CTRGS3rs10981790 
ATRGS3NA 
GCRGS3rs12350531 
AGRGS3rs12341266 
CGRGS3rs10817493 
CTSIRT3rs11246020Sirtuin 5
CASIRT3rs11555236 
TGSIRT3rs12365010 
CTSIRT3rs34700713 
TGSIRT3rs9795476 
AGSIRT7rs1879568 
AGSIRT7rs1879569 
AGSIRT6rs350845 
TGSIRT6rs7246235 
ACSIRT6rs350843 
TGSIRT6rs7260071 
CTSIRT6rs352493 
GCSIRT2rs45496398 
GASIRT2rs11879010 
GASIRT2rs11879029 
CTSIRT2rs11667030 
AGSIRT2rs11083483 
TCSIRT2rs10410544 
GASLC66A1rs58790041Solute carrier family 6
TCSLC6A17rs7527375 
GASLC6A17rs12737742 
ATSLC6A17rs1007692 
CTSLC6A17rs12033312 
CTSLC6A5rs76857783 
GASLC6A5rs1443547 
CTSLC6A5rs7109418 
CTSLC6A5rs2241941 
TCSLC6A5rs1443548 
CGSLC6A5rs1443549 
CGSLC6A5rs72927519 
TCSLC6A5rs2278649 
GASLC6A5rs1443551 
AGSLC6A5rs7925597 
ACSLC6A5rs7925624 
GASLC6A5rs11827415 
CTSLC6A5rs4923548 
GASLC6A5rs2276433 
AGSLC6A12rs646569 
TCSLC6A12rs216248 
AGSLC6A12rs216250 
CTSLC6A13rs2289954 
CTSLC6A13rs74057642 
CGSLC6A10Prs395061 
CTSLC6A10Prs3878528 
TCSLC6A10Prs432506 
GASLC6A10Prs865876957 
AGSLC6A10Prs796806794 
GTSLC6A10Prs796465823 
CTSLC6A10Prs542763347 
GASLC6A10Prs62046808 
CTSLC6A10Prs28510469 
AGSLC6A2rs5564 
TGSLC6A2rs28613651 
TCSLC6A2rs2279805 
GASLC6A2rs5569 
GASLC6A2rs998424 
CTSLC6A2rs2242447 
GTSLC6A16rs2278405 
GASLC6A16rs536601412 
CTSLC66A3rs2271622 
TCSLC66A3rs6432182 
TCSLC6A11rs2304725 
TCSLC6A11rs2272395 
CTSLC6A11rs2272399 
CTSLC6A20rs2251347 
AGSLC6A20rs758386 
CTSLC12A7rs6554618 
TASLC12A7rs116648138 
AGSLC6A19NA 
GASLC6A19rs7732589 
AGSLC6A19rs6554663 
AGSLC6A19rs4975629 
TCSLC6A19rs12513763 
GASLC6A19NA 
ACSLC6A19NA 
GASLC6A18rs7704058 
TASLC6A18rs7728667 
CTSLC6A18rs7705355 
TCSLC6A18rs7728814 
AGSLC6A18rs7724858 
GCSLC6A18rs4975623 
CASLC6A3rs40358 
GTSLC6A3rs460000 
CTSLC6A8rs376038235 
CASMAD9rs3748305SMAD family member 2
GTSMAD9rs9576126 
TCSMAD9rs182137303 
CASMAD3rs1866319 
AGSMAD3rs1065080 
GASMAD3rs758586312 
CTSMAD5-AS1rs3764942 
TGSMAD5-AS1rs3764941 
GTSMAD5-AS1rs548522421 
TCSMAD5-AS1rs3764939 
AGSMAD5rs4585442 
AGQSOX1rs2298206Sry-related HMG box
CTQSOX1rs7521513 
CTQSOX1rs1050154 
GTQSOX1rs67343209 
TCQSOX1rs7544147 
TCSOX8rs237674 
CTSOX8rs11542179 
ACSOX9rs1042667 
CTSOX3rs396467 
CTSOX13rs2250538 
AGSOX13rs7551756 
AGSOX6rs4617548 
TCSOX6rs7926424 
GASOX6rs12277740 
TCSOX5rs4636755 
CTSOX5rs7485662 
TCSOX5rs7980561 
CGSOX21rs1060474 
TGSOX21rs2253604 
CGSOX14rs1385306 
GASOX30rs35793864 
GTSOX30rs12188040 
CTSOX30rs777105078 
TCQSOX2rs9696116 
TCQSOX2rs10858247 
CTQSOX2rs12684650 
CAQSOX2NA 
TCQSOX2rs12380852 
TGQSOX2NA 
CTQSOX2rs79849109 
TCQSOX2rs567434269 
GAQSOX2rs62579871 
CTSYNCrs360042Signal transducer and activator of transcription 3
GATFAP2Ars9367875Transcription factor AP-2 alpha
AGTFAP2Ars9370818 
CTTFAP2Ars11968445 
AGTFAP2Ars1925775 
GATFAP2Ars9477310 
CTTGFBR3rs2493202Transforming growth factor beta
AGTGFBR3rs284878 
CTTGFBR3rs1805112 
GATGFBR3rs11165441 
GATGFBR3rs1805110 
CTTGFBR3rs1805109 
CTTGFB3rs3917201 
AGTGFB1rs1800469 
CTTGFBR1rs11568753 
TCTGFBR1NA 
AGTGFBR1rs7041311 
GATGFBR1rs334354 
CTVEGFCrs7664413Vascular endothelial growth factor A
CAVEGFArs2146323 
GAVEGFArs3025009 
TCVEGFArs3025010 
CTYARSrs144866833 
GAYARSrs4951787 
AGZEB1rs189276857Zinc finger E-box binding homeobox 1
GAZEB1rs220060 

[i] RS, Reference single nucleotide polymorphism; NA, not applicable.

Discussion

The present study aimed to identify genetic factors associated with MDD. SLC6A3, BDNF and JARID2 were chosen because of their potential involvement in MDD pathways and previous research findings (28,30,31,34,36,38). To the best of our knowledge, the present study is the first to report on JARID2 and BDNF variations between patients with MDD and healthy individuals in Thailand. The present findings revealed an association between the SNP rs40184 in SLC6A3 and MDD, aligning with the results of other studies (34,45). This SNP alters SLC6A3 expression, which is associated with dopamine levels in the brain and depression severity (46,47). Previous studies have reported associations between dopamine transporter (DAT) levels and various psychiatric disorders including MDD and bipolar disorder (33,45,48). However, further investigation is necessary to elucidate these relationships. To comprehend the role of SLC6A3 (which encodes DAT) in psychiatric conditions, it must be explored within the broader context of phenotypical and allelic heterogeneity, as well as gene-environment interactions. Various SNPs in BDNF have been associated with MDD and suicide, including rs12273363, rs7124442, rs10767664, rs962369, rs908867 and especially rs6265 (Val66Met) (49-54). The rs6265 SNP decreases the activity-dependent release of BDNF (55). Furthermore, a previous study indicated an association between low BDNF levels and MDD pathogenesis (30).

To date, 500,000-1,000,000 SNPs have been identified in individuals with MDD compared with healthy controls. However, GWASs have not definitively demonstrated the influence of these SNPs on underlying mechanisms of mental disorders. These SNPs typically exhibit minimal phenotypic effects, with ORs ranging from 1.0 to 1.2 (9,56-59). Despite the subtle effects of individual SNPs, twin studies have revealed a significant genetic component in MDD (60,61). For example, one study found 37% heritability for recurrent unipolar MDD, with only minor environmental risk factors (62). More broadly, various studies have estimated the heritability of MDD to be between 30 and 50% (21,24). SNPs and other factors appear to be weakly associated with MDD. However, genetic variation varies between population, genetic background, and sequencing technology. The genetic mechanism for heritability remains unclear. Viruses or other factors may contribute to MDD pathophysiology (8,10,59). HHV-6 is often transmitted from mother to child and within families and can persist for life, this genetic polymorphism in HHV-6 may impact the heritability of MDD (21).

The present study identified a novel association between HHV-6 status and an SNP in JARID2. To the best of our knowledge, the present study is the first to explore the relationship between JARID2 and HHV-6, including both infection status and viral load, in the context of MDD. Variant A allele of SNP rs9383046 in JARID2 was more prevalent among individuals positive for HHV-6 infection and was positively associated with viral load. Previous research has linked pathogenic polymorphisms in JARID2 to a neurodevelopmental syndrome marked by developmental delays, cognitive impairment, hypotonia, autism and behavioral abnormalities (39,63). Additionally, rs326221458 SNP has been shown to affect JARID2 expression, which influences aggressive behavior in weaned pigs (64).

A total of six main hypotheses have been proposed to explain MDD pathogenesis: Hypothalamic/pituitary/adrenal axis dysfunction, monoamine imbalance, inflammation, genetic and epigenetic anomalies, structural and functional brain remodeling and social psychological factors. However, no single hypothesis can fully explain the pathology of MDD (8,10). The present study did not identify a direct association between MDD and JARID2. However, JARID2 may be directly associated with HHV-6 infection and indirectly with neurodevelopmental disorders, potentially increasing the risk of developing MDD. JARID2 polymorphisms may increase HHV-6 viral load, which is associated with inflammation and may contribute to MDD development (22,40). JARID2 serves as a transcriptional repressor protein and regulates histone methyltransferase complexes (65). Viral infections alter the expression of lysine demethylase 2B (KDM2B/NDY1), enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) and JARID2. This alteration actively degrades growth factor-independent 1 transcriptional repressor (GFI1), thereby promoting activation of the major immediate early promoter (MIEP) of human cytomegalovirus (HCMV). The combination of KDM2B/NDY1, EZH2 and JARID2 promotes histone H3 K27 trimethylation, which is associated with transcriptional suppression. EZH2, responsible for histone H3 K27 trimethylation, interacts with JARID2, a non-canonical jumonji domain protein that modulates EZH2 methyltransferase activity. HCMV infects cells by targeting GFI1 through various mechanisms. GFI1 mRNA and protein levels are rapidly downregulated upon viral exposure, likely via degradation (18,66-68). HHV-6 is similarly activated, with GFI1 rapidly degrading after HHV-6 infection, activating the MIEP.

Future research should further examine the roles of KDM2B/NDY1, EZH2, JARID2 and JMJD3 using the SNPs in JMJD1, JMJD4, JMJD7, JMJD8, and JARID2 identified in the present study (18,66-68). Moreover, a common feature of HHV is the association of latent genomes with polycomb group proteins, specifically PRC2.2, which includes JARID2 and is associated with H3K27me3 (43,69).

Several mechanisms have been proposed for MDD onset, including monoamine theory, neuroendocrine mechanisms, neurotrophic hypothesis and neuroinflammation (8,10). JARID2 may influence HHV-6 and tumor necrosis factor-alpha (TNF-α)-induced inflammation through various indirect pathways. Neuroinflammation and cytokines are connected to monoamine deficiency, suppressed neurogenesis, and dysregulation of the hypothalamic/pituitary/adrenal axis. Chronic stress and inflammation can cause peripheral inflammatory markers to cross the blood-brain barrier into the central nervous system, activating microglia (70-73). TNF-α promoter SNP (G>A) and elevated HHV-6 viral load, including factors like JARID2, lead to TNF-α overexpression and subsequent inflammation (39). Therefore, the current data suggest that HHV-6 may enhance viral load by promoting neuroinflammation through JARID2.

The present study had limitations. The primary limitation was small sample size. Another was the difference in mean ages between the MDD group and the healthy control group, which may cause bias in the results. The present study investigated SLC6A3 as a biomarker for MDD diagnosis, as well as JARID2 and HHV-6 as potential therapeutic targets. The role of JARID2 in MDD should be assessed by investigating its expression patterns.

In conclusion, the present study demonstrated significant association between SNPs in BDNF (rs6265) and SLC6A3 (rs28363170) and MDD. A higher HHV-6 viral load was found in participants carrying variant A allele of SNP rs9383046 (G>A) in JARID2. Whole-exome sequencing identified SNPs in JARID2 in patients with MDD, indicating interactions between HHV-6 infection, SNPs and MDD development.

Acknowledgements

The authors would like to thank the Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand, for use of the qPCR machine.

Funding

Funding: The present study was supported by Faculty of Pharmaceutical Sciences, Ubon Ratchathani University (grant no. Phar.UBU.0604.11-4/2567), The University of Phayao (grant no. FF64-RIB005) and Department of Medical Technology, Faculty of Allied Health Sciences, Burapha University (grant no. AHS04/2567).

Availability of data and materials

The data generated in the present study may be found in the Sequence Read Archive database under accession number PRJNA1146872 or at the following URL: ncbi.nlm.nih.gov/sra/?term=PRJNA1146872.

Authors' contributions

SBum, SBud and NP conceived the study and designed and performed experiments. SS performed experiments. SBum and NP wrote and reviewed the manuscript. SD and SP conceived the study and designed experiments. SBum, SBud and NP confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Committee on Human Research Ethics in Health Sciences and Science and Technology, University of Phayao, Thailand (UP-HEC 1.3/013/65). The participants provided written informed consent before the start of the study. All protocols involving human subjects adhered to the Declaration of Helsinki, Belmont Report, the International Conference on Harmonization in Good Clinical Practice and the Council for International Organizations of Medical Sciences.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Shadrina M, Bondarenko EA and Slominsky PA: Genetics factors in major depression disease. Front Psychiatry. 9(334)2018.PubMed/NCBI View Article : Google Scholar

2 

Wangtongkum S, Sucharitakul P, Wongjaroen S and Maneechompoo S: Prevalence of depression among a population aged over 45 years in Chiang Mai, Thailand. J Med Assoc Thai. 91:1812–1816. 2008.PubMed/NCBI

3 

Singkhorn O, Apidechkul T, Pitchalard K, Moonpanane K, Hamtanon P, Sunsern R, Leaungsomnapa Y and Thepsaw J: Prevalence of and factors associated with depression in the hill tribe population aged 40 years and older in northern Thailand. Int J Ment Health Syst. 15(62)2021.PubMed/NCBI View Article : Google Scholar

4 

Pengpid S, Peltzer K and Anantanasuwong D: Prevalence and determinants of incident and persistent depressive symptoms among middle-aged and older adults in Thailand: prospective cohort study. BJPsych Open. 9(e99)2023.PubMed/NCBI View Article : Google Scholar

5 

Musumari PM, Tangmunkongvorakul A, Srithanaviboonchai K, Techasrivichien T, Suguimoto SP, Ono-Kihara M and Kihara M: Grit is associated with lower level of depression and anxiety among university students in Chiang Mai, Thailand: A cross-sectional study. PLoS One. 13(e0209121)2018.PubMed/NCBI View Article : Google Scholar

6 

Charoensakulchai S, Usawachoke S, Kongbangpor W, Thanavirun P, Mitsiriswat A, Pinijnai O, Kaensingh S, Chaiyakham N, Chamnanmont C, Ninnakala N, et al: Prevalence and associated factors influencing depression in older adults living in rural Thailand: A cross-sectional study. Geriatr Gerontol Int. 19:1248–1253. 2019.PubMed/NCBI View Article : Google Scholar

7 

Aung TNN, Moolphate S, Koyanagi Y, Angkurawaranon C, Supakankunti S, Yuasa M and Aung MN: Depression and associated factors among community-dwelling Thai older adults in Northern Thailand: The relationship between history of fall and geriatric depression. Int J Environ Res Public Health. 19(10574)2022.PubMed/NCBI View Article : Google Scholar

8 

Cui L, Li S, Wang S, Wu X, Liu Y, Yu W, Wang Y, Tang Y, Xia M and Li B: Major depressive disorder: Hypothesis, mechanism, prevention and treatment. Signal Transduct Target Ther. 9(30)2024.PubMed/NCBI View Article : Google Scholar

9 

Marx W, Penninx BWJH, Solmi M, Furukawa TA, Firth J, Carvalho AF and Berk M: Major depressive disorder. Nat Rev Dis Primers. 9(44)2023.PubMed/NCBI View Article : Google Scholar

10 

Belmaker RH and Agam G: Major depressive disorder. N Engl J Med. 358:55–68. 2008.PubMed/NCBI View Article : Google Scholar

11 

Wang X, Zhang L, Lei Y, Liu X, Zhou X, Liu Y, Wang M, Yang L, Zhang L, Fan S and Xie P: Meta-analysis of infectious agents and depression. Sci Rep. 4(4530)2014.PubMed/NCBI View Article : Google Scholar

12 

Yu X, Wang S, Wu W, Chang H, Shan P, Yang L, Zhang W and Wang X: Exploring new mechanism of depression from the effects of virus on nerve cells. Cells. 12(1767)2023.PubMed/NCBI View Article : Google Scholar

13 

Zheng H, Webster MJ, Weickert CS, Beasley CL, Paulus MP, Yolken RH and Savitz J: Cytomegalovirus antibodies are associated with mood disorders, suicide, markers of neuroinflammation, and microglia activation in postmortem brain samples. Mol Psychiatry. 28:5282–5292. 2023.PubMed/NCBI View Article : Google Scholar

14 

Simon MS, Ioannou M, Arteaga-Henríquez G, Wijkhuijs A, Berghmans R, Musil R, Müller N and Drexhage HA: Premature T cell aging in major depression: A double hit by the state of disease and cytomegalovirus infection. Brain Behav Immun Health. 29(100608)2023.PubMed/NCBI View Article : Google Scholar

15 

Jones-Brando L, Dickerson F, Ford G, Stallings C, Origoni A, Katsafanas E, Sweeney K, Squire A, Khushalani S and Yolken R: Atypical immune response to Epstein-Barr virus in major depressive disorder. J Affect Disord. 264:221–226. 2020.PubMed/NCBI View Article : Google Scholar

16 

Bayturan S, Sapmaz ŞY, Uzun AD, Kandemir H and Ecemiş T: Relationship of herpesvirus (HSV1, EBV, CMV, HHV6) seropositivity with depressive disorder and its clinical aspects: The first study in children. J Med Virol. 94:5484–5491. 2022.PubMed/NCBI View Article : Google Scholar

17 

Sourvinos G, Morou A, Sanidas I, Codruta I, Ezell SA, Doxaki C, Kampranis SC, Kottakis F and Tsichlis PN: The Downregulation of GFI1 by the EZH2-NDY1/KDM2B-JARID2 Axis and by Human Cytomegalovirus (HCMV) Associated Factors Allows the Activation of the HCMV Major IE promoter and the transition to productive infection. PLoS Pathog. 10(e1004136)2014.PubMed/NCBI View Article : Google Scholar

18 

Kobayashi N, Oka N, Takahashi M, Shimada K, Ishii A, Tatebayashi Y, Shigeta M, Yanagisawa H and Kondo K: Human Herpesvirus 6B greatly increases risk of depression by activating hypothalamic-pituitary-adrenal axis during latent phase of infection. iScience. 23(101187)2020.PubMed/NCBI View Article : Google Scholar

19 

Pantry SN and Medveczky PG: Latency, integration, and reactivation of human herpesvirus-6. Viruses. 9(194)2017.PubMed/NCBI View Article : Google Scholar

20 

Strenger V, Aberle SW, Nacheva EP and Urban C: Chromosomal integration of the HHV-6 genome in a patient with nodular sclerosis Hodgkin lymphoma. Br J Haematol. 161:594–595. 2013.PubMed/NCBI View Article : Google Scholar

21 

Kobayashi N, Shimada K, Ishii A, Osaka R, Nishiyama T, Shigeta M, Yanagisawa H, Oka N and Kondo K: Identification of a strong genetic risk factor for major depressive disorder in the human virome. iScience. 27(109203)2024.PubMed/NCBI View Article : Google Scholar

22 

Levey DF, Stein MB, Wendt FR, Pathak GA, Zhou H, Aslan M, Quaden R, Harrington KM, Nuñez YZ, Overstreet C, et al: Bi-ancestral depression GWAS in the Million Veteran Program and meta-analysis in >1.2 million individuals highlight new therapeutic directions. Nat Neurosci. 24:954–963. 2021.PubMed/NCBI View Article : Google Scholar

23 

Howard DM, Adams MJ, Clarke TK, Hafferty JD, Gibson J, Shirali M, Coleman JRI, Hagenaars SP, Ward J, Wigmore EM, et al: Genome-wide meta-analysis of depression identifies 102 independent variants and highlights the importance of the prefrontal brain regions. Nat Neurosci. 22:343–352. 2019.PubMed/NCBI View Article : Google Scholar

24 

Flint J: The genetic basis of major depressive disorder. Mol Psychiatry. 28:2254–2265. 2023.PubMed/NCBI View Article : Google Scholar

25 

Alshaya DS: Genetic and epigenetic factors associated with depression: An updated overview. Saudi J Biol Sci. 29(103311)2022.PubMed/NCBI View Article : Google Scholar

26 

Fries GR, Saldana VA, Finnstein J and Rein T: Molecular pathways of major depressive disorder converge on the synapse. Mol Psychiatry. 28:284–297. 2023.PubMed/NCBI View Article : Google Scholar

27 

Yuan M, Yang B, Rothschild G, Mann JJ, Sanford LD, Tang X, Huang C, Wang C and Zhang W: Epigenetic regulation in major depression and other stress-related disorders: molecular mechanisms, clinical relevance and therapeutic potential. Signal Transduct Target Ther. 8(309)2023.PubMed/NCBI View Article : Google Scholar

28 

Ribeiro L, Busnello JV, Cantor RM, Whelan F, Whittaker P, Deloukas P, Wong ML and Licinio J: The brain-derived neurotrophic factor rs6265 (Val66Met) polymorphism and depression in Mexican-Americans. Neuroreport. 18:1291–1293. 2007.PubMed/NCBI View Article : Google Scholar

29 

Nguyen VT, Hill B, Sims N, Heck A, Negron M, Lusk C and Galindo CL: Brain-derived neurotrophic factor rs6265 (Val66Met) single nucleotide polymorphism as a master modifier of human pathophysiology. Neural Regen Res. 18:102–106. 2023.PubMed/NCBI View Article : Google Scholar

30 

Szarowicz CA, Steece-Collier K and Caulfield ME: New frontiers in neurodegeneration and regeneration associated with brain-derived neurotrophic factor and the rs6265 single nucleotide polymorphism. Int J Mol Sci. 23(8011)2022.PubMed/NCBI View Article : Google Scholar

31 

Bieliński M, Jaracz M, Lesiewska N, Tomaszewska M, Sikora M, Junik R, Kamińska A, Tretyn A and Borkowska A: Association between COMT Val158Met and DAT1 polymorphisms and depressive symptoms in the obese population. Neuropsychiatr Dis Treat. 13:2221–2229. 2017.PubMed/NCBI View Article : Google Scholar

32 

Tonelli E, Pascale E, Troianiello M, D'Addario C and Adriani W: DAT1 gene methylation as an epigenetic biomarker in attention deficit hyperactivity disorder: A commentary. Front Genet. 11(444)2020.PubMed/NCBI View Article : Google Scholar

33 

Pinsonneault JK, Han DD, Burdick KE, Kataki M, Bertolino A, Malhotra AK, Gu HH and Sadee W: Dopamine transporter gene variant affecting expression in human brain is associated with bipolar disorder. Neuropsychopharmacology. 36:1644–1655. 2011.PubMed/NCBI View Article : Google Scholar

34 

Gontijo BR, Possatti I, Fratelli CF, Pereira ASR, Bonasser LSS, de Souza Silva CM and Rodrigues da Silva IC: The 3'UTR VNTR SLC6A3 genetic variant and major depressive disorder: A systematic review. Biomedicines. 11(2270)2023.PubMed/NCBI View Article : Google Scholar

35 

Pattarachotanant N, Sritharathikhun T, Suttirat S and Tencomnao T: Association of C/T polymorphism in intron 14 of the dopamine transporter gene (rs40184) with major depression in a northeastern Thai population. Genet Mol Res. 9:565–572. 2010.PubMed/NCBI View Article : Google Scholar

36 

Chen G, Liu Y, Norton N, Liu W, Zhu H, Zhou P, Luan M, Yang S, Chen X, Carroll L, et al: Whole genome association study in a homogenous population in Shandong peninsula of China reveals JARID2 as a susceptibility gene for schizophrenia. J Biomed Biotechnol. 2009(536918)2009.PubMed/NCBI View Article : Google Scholar

37 

Pedrosa E, Ye K, Nolan KA, Morrell L, Okun JM, Persky AD, Saito T and Lachman HM: Positive association of schizophrenia to JARID2 gene. Am J Med Genet B Neuropsychiatr Genet. 144B:45–51. 2007.PubMed/NCBI View Article : Google Scholar

38 

Verberne EA, van der Laan L, Haghshenas S, Rooney K, Levy MA, Alders M, Maas SM, Jansen S, Lieden A, Anderlid BM, et al: DNA Methylation Signature for JARID2-Neurodevelopmental Syndrome. Int J Mol Sci. 23(8001)2022.PubMed/NCBI View Article : Google Scholar

39 

Sumala S, Ekalaksananan T, Pientong C, Buddhisa S, Passorn S, Duangjit S, Janyakhantikul S, Suktus A and Bumrungthai S: The Association of HHV-6 and the TNF-α (-308G/A) promotor with major depressive disorder patients and healthy controls in Thailand. Viruses. 15(1898)2023.PubMed/NCBI View Article : Google Scholar

40 

American Psychiatric Association: DSM-5 Task Force: Diagnostic and Statistical Manual of Mental Disorders: DSM-5™. 5th edition. American Psychiatric Publishing, Inc., Washington DC, 2013.

41 

Faris A, Hadi BH and Abidin ZS: Development and validation of high resolution melting assays for high-throughput screening of BDNF rs6265 and DAT1 rs40184. Malays J Med Res. 14:2636–9346. 2018.

42 

Chen X, Long F, Cai B, Chen X and Chen G: A novel relationship for schizophrenia, bipolar and major depressive disorder Part 5: A hint from chromosome 5 high density association screen. Am J Transl Res. 9:2473–2491. 2017.PubMed/NCBI

43 

Dochnal SA, Francois AK and Cliffe AR: De Novo polycomb recruitment: Lessons from latent herpesviruses. Viruses. 13(1470)2021.PubMed/NCBI View Article : Google Scholar

44 

Bergé-Lefranc JL, Jay P, Massacrier A, Cau P, Mattei MG, Bauer S, Marsollier C, Berta P and Fontes M: Characterization of the human jumonji gene. Hum Mol Genet. 5:1637–1641. 1996.PubMed/NCBI View Article : Google Scholar

45 

Haeffel GJ, Getchell M, Koposov RA, Yrigollen CM, Deyoung CG, Klinteberg BA, Oreland L, Ruchkin VV and Grigorenko EL: Association between polymorphisms in the dopamine transporter gene and depression evidence for a gene-environment interaction in a sample of juvenile detainees. Psychol Sci. 19:62–69. 2008.PubMed/NCBI View Article : Google Scholar

46 

Rafikova EI, Shibalev DV, Shadrina MI, Slominsky PA, Guekht AB, Ryskov AP and Vasilyev VA: Influence of polymorphic gene variants of the dopaminergic system on the risk of disorders with depressive symptoms. Russ J Genet. 57:942–948. 2021.

47 

Pizzagalli DA, Berretta S, Wooten D, Goer F, Pilobello KT, Kumar P, Murray L, Beltzer M, Boyer-Boiteau A, Alpert N, et al: Assessment of striatal dopamine transporter binding in individuals with major depressive disorder: In Vivo positron emission tomography and postmortem evidence. JAMA Psychiatry. 76:854–861. 2019.PubMed/NCBI View Article : Google Scholar

48 

Reith MEA, Kortagere S, Wiers CE, Sun H, Kurian MA, Galli A, Volkow ND and Lin Z: The dopamine transporter gene SLC6A3: Multidisease risks. Mol Psychiatry. 27:1031–1046. 2022.PubMed/NCBI View Article : Google Scholar

49 

Lin E, Hong CJ, Hwang JP, Liou YJ, Yang CH, Cheng D and Tsai SJ: Gene-gene interactions of the brain-derived neurotrophic-factor and neurotrophic tyrosine kinase receptor 2 genes in geriatric depression. Rejuvenation Res. 12:387–393. 2009.PubMed/NCBI View Article : Google Scholar

50 

Zhao M, Chen L, Yang J, Han D, Fang D, Qiu X, Yang X, Qiao Z, Ma J, Wang L, et al: BDNF Val66Met polymorphism, life stress and depression: A meta-analysis of gene-environment interaction. J Affect Disord. 227:226–235. 2018.PubMed/NCBI View Article : Google Scholar

51 

Hwang JP, Tsai SJ, Hong CJ, Yang CH, Lirng JF and Yang YM: The Val66Met polymorphism of the brain-derived neurotrophic-factor gene is associated with geriatric depression. Neurobiol Aging. 27:1834–1837. 2006.PubMed/NCBI View Article : Google Scholar

52 

Porter GA and O'Connor JC: Brain-derived neurotrophic factor and inflammation in depression: Pathogenic partners in crime? World J Psychiatry. 12:77–97. 2022.PubMed/NCBI View Article : Google Scholar

53 

Hing B, Davidson S, Lear M, Breen G, Quinn J, McGuffin P and MacKenzie A: A polymorphism associated with depressive disorders differentially regulates brain derived neurotrophic factor promoter IV activity. Biol Psychiatry. 71:618–626. 2012.PubMed/NCBI View Article : Google Scholar

54 

Ropret S, Zupanc T, Komel R and Videtič Paska A: Single nucleotide polymorphisms in the BDNF gene and suicide in the Slovenian sample. Neurosci Lett. 602:12–16. 2015.PubMed/NCBI View Article : Google Scholar

55 

Egan MF, Kojima M, Callicott JH, Goldberg TE, Kolachana BS, Bertolino A, Zaitsev E, Gold B, Goldman D, Dean M, et al: The BDNF val66met polymorphism affects activity-dependent secretion of BDNF and human memory and hippocampal function. Cell. 112:257–269. 2003.PubMed/NCBI View Article : Google Scholar

56 

Bosker FJ, Hartman CA, Nolte IM, Prins BP, Terpstra P, Posthuma D, van Veen T, Willemsen G, DeRijk RH, de Geus EJ, et al: Poor replication of candidate genes for major depressive disorder using genome-wide association data. Mol Psychiatry. 16:516–532. 2011.PubMed/NCBI View Article : Google Scholar

57 

Levinson DF, Mostafavi S, Milaneschi Y, Rivera M, Ripke S, Wray NR and Sullivan PF: Genetic studies of major depressive disorder: Why are there no genome-wide association study. findings, and what can we do about it? Biol Psychiatry. 76:510–512. 2014.PubMed/NCBI View Article : Google Scholar

58 

Lohoff FW: Overview of the genetics of major depressive disorder. Curr Psychiatry Rep. 12:539–546. 2010.PubMed/NCBI View Article : Google Scholar

59 

Li Z, Ruan M, Chen J and Fang Y: Major Depressive Disorder: Advances in neuroscience research and translational applications. Neurosci Bull. 37:863–880. 2021.PubMed/NCBI View Article : Google Scholar

60 

Roberson-Nay R, Lapato DM, Wolen AR, Lancaster EE, Webb BT, Verhulst B, Hettema JM and York TP: An epigenome-wide association study of early-onset major depression in monozygotic twins. Transl Psychiatry. 10(301)2020.PubMed/NCBI View Article : Google Scholar

61 

Huider F, Milaneschi Y, van der Zee MD, de Geus EJC, Helmer Q, Penninx BWJH and Boomsma DI: major depressive disorder and lifestyle: Correlated genetic effects in extended twin pedigrees. Genes (Basel). 12(1509)2021.PubMed/NCBI View Article : Google Scholar

62 

Sullivan PF, Neale MC and Kendler KS: Genetic epidemiology of major depression: Review and meta-analysis. Am J Psychiatry. 157:1552–1562. 2000.PubMed/NCBI View Article : Google Scholar

63 

Aref-Eshghi E, Kerkhof J and Pedro VP: Groupe DI France. Barat-Houari M, Ruiz-Pallares N, Andrau JC, Lacombe D, Van-Gils J, Fergelot P, et al: Evaluation of DNA Methylation Episignatures for Diagnosis and Phenotype Correlations in 42 Mendelian Neurodevelopmental Disorders. Am J Hum Genet. 106:356–370. 2020.PubMed/NCBI View Article : Google Scholar

64 

Yang H, Zhang C, Chao X, Zhao J, Liu M, Chen J, Liu S, Wang T, Muhammad A, Schinckel AP and Zhou B: A functional single nucleotide polymorphism in the 3' untranslated region of the porcine JARID2 gene is associated with aggressive behavior of weaned pigs after mixing. Int J Mol Sci. 25(27)2023.PubMed/NCBI View Article : Google Scholar

65 

van der Laan L, Rooney K, Haghshenas S, Silva A, McConkey H, Relator R, Levy MA, Valenzuela I, Trujillano L, Lasa-Aranzasti A, et al: Functional insight into and refinement of the genomic boundaries of the JARID2-Neurodevelopmental disorder episignature. Int J Mol Sci. 24(14240)2023.PubMed/NCBI View Article : Google Scholar

66 

Cao R, Wang H, He J, Erdjument-Bromage H, Tempst P and Zhang Y: Role of hPHF1 in H3K27 methylation and Hox gene silencing. Mol Cell Biol. 28:1862–1872. 2008.PubMed/NCBI View Article : Google Scholar

67 

Nekrasov M, Klymenko T, Fraterman S, Papp B, Oktaba K, Köcher T, Cohen A, Stunnenberg HG, Wilm M and Müller J: Pcl-PRC2 is needed to generate high levels of H3-K27 trimethylation at Polycomb target genes. EMBO J. 26:4078–4088. 2007.PubMed/NCBI View Article : Google Scholar

68 

Herz HM and Shilatifard A: The JARID2-PRC2 duality. Genes Dev. 24:857–861. 2010.PubMed/NCBI View Article : Google Scholar

69 

Francois AK, Rohani A, Loftus M, Dochnal S, Hrit J, McFarlane S, Whitford A, Lewis A, Krakowiak P, Boutell C, et al: Single-genome analysis reveals a heterogeneous association of the herpes simplex virus genome with H3K27me2 and the reader PHF20L1 following infection of human fibroblasts. mBio. 15(e0327823)2024.PubMed/NCBI View Article : Google Scholar

70 

Bakunina N, Pariante CM and Zunszain PA: Immune mechanisms linked to depression via oxidative stress and neuroprogression. Immunology. 144:365–373. 2015.PubMed/NCBI View Article : Google Scholar

71 

Troubat R, Barone P, Leman S, Desmidt T, Cressant A, Atanasova B, Brizard B, El Hage W, Surget A, Belzung C and Camus V: Neuroinflammation and depression: A review. Eur J Neurosci. 53:151–171. 2021.PubMed/NCBI View Article : Google Scholar

72 

Brites D and Fernandes A: Neuroinflammation and Depression: Microglia activation, extracellular microvesicles and microRNA dysregulation. Front Cell Neurosci. 9(476)2015.PubMed/NCBI View Article : Google Scholar

73 

Peirce JM and Alviña K: The role of inflammation and the gut microbiome in depression and anxiety. J Neurosci Res. 97:1223–1241. 2019.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

December-2024
Volume 21 Issue 6

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Bumrungthai S, Buddhisa S, Duangjit S, Passorn S, Sumala S and Prakobkaew N: Association of HHV‑6 reactivation and SLC6A3 (C&gt;T, rs40184), BDNF (C&gt;T, rs6265), and JARID2 (G&gt;A, rs9383046) single nucleotide polymorphisms in depression. Biomed Rep 21: 181, 2024.
APA
Bumrungthai, S., Buddhisa, S., Duangjit, S., Passorn, S., Sumala, S., & Prakobkaew, N. (2024). Association of HHV‑6 reactivation and SLC6A3 (C&gt;T, rs40184), BDNF (C&gt;T, rs6265), and JARID2 (G&gt;A, rs9383046) single nucleotide polymorphisms in depression. Biomedical Reports, 21, 181. https://doi.org/10.3892/br.2024.1869
MLA
Bumrungthai, S., Buddhisa, S., Duangjit, S., Passorn, S., Sumala, S., Prakobkaew, N."Association of HHV‑6 reactivation and SLC6A3 (C&gt;T, rs40184), BDNF (C&gt;T, rs6265), and JARID2 (G&gt;A, rs9383046) single nucleotide polymorphisms in depression". Biomedical Reports 21.6 (2024): 181.
Chicago
Bumrungthai, S., Buddhisa, S., Duangjit, S., Passorn, S., Sumala, S., Prakobkaew, N."Association of HHV‑6 reactivation and SLC6A3 (C&gt;T, rs40184), BDNF (C&gt;T, rs6265), and JARID2 (G&gt;A, rs9383046) single nucleotide polymorphisms in depression". Biomedical Reports 21, no. 6 (2024): 181. https://doi.org/10.3892/br.2024.1869