1
|
Keener RM, Shi S, Dalapati T, Wang L, Reinoso-Vizcaino NM, Luftig MA, Miller SI, Wilson TJ, Ko DC. Human genetic variation reveals FCRL3 is a lymphocyte receptor for Yersinia pestis. CELL GENOMICS 2025:100917. [PMID: 40494344 DOI: 10.1016/j.xgen.2025.100917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/31/2025] [Accepted: 05/13/2025] [Indexed: 06/18/2025]
Abstract
Yersinia pestis is the bacterium responsible for plague, one of the deadliest diseases in history. To discover human genetic determinants of Y. pestis infection, we utilized nearly 1,000 genetically diverse lymphoblastoid cell lines in a cellular genome-wide association study. A nonsynonymous SNP, rs2282284 (N721S), in Fc receptor-like 3 (FCRL3) was associated with bacterial invasion of host cells (p = 9 × 10-8). Overexpressed FCRL3 facilitated attachment and invasion of Y. pestis and colocalized with Y. pestis at attachment sites. These properties were variably conserved across the FCRL family, revealing an immunoglobulin-like domain and signaling motifs shared by FCRL3 and FCRL5 to be necessary for attachment and invasion. Direct binding to FCRL5 extracellular domain was confirmed, and B cells (the primary cells that express FCRLs) were preferentially invaded by Y. pestis. Thus, Y. pestis hijacks FCRL proteins, possibly taking advantage of an immune receptor to create a lymphocyte niche during infection.
Collapse
Affiliation(s)
- Rachel M Keener
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA; University Program in Genetics and Genomics, Duke University, Durham, NC 27710, USA
| | - Sam Shi
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Trisha Dalapati
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Nicolás M Reinoso-Vizcaino
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Micah A Luftig
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Samuel I Miller
- Departments of Genome Sciences, Medicine, and Microbiology, University of Washington, Seattle, WA 98195, USA
| | - Timothy J Wilson
- Department of Microbiology, Miami University, Oxford, OH 45056, USA
| | - Dennis C Ko
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA; University Program in Genetics and Genomics, Duke University, Durham, NC 27710, USA; Division of Infectious Diseases, Department of Medicine, School of Medicine, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
2
|
Tao Y, Pan Q, Cai T, Lu ZH, Haque M, Dottorini T, Colvin LA, Smith BH, Meng W. A genome-wide association study identifies novel genetic variants associated with neck or shoulder pain in the UK biobank (N = 430,193). Pain Rep 2025; 10:e1267. [PMID: 40291381 PMCID: PMC12026381 DOI: 10.1097/pr9.0000000000001267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/11/2025] [Accepted: 02/09/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction Neck and shoulder pain are prevalent musculoskeletal disorders that significantly affect the quality of life for a substantial portion of the global population. Studies have shown that women are more susceptible than men. Objective This study aims to discover genetic variants associated with neck or shoulder pain through a genome-wide association study (GWAS), using data from 430,193 participants in the UK Biobank. Methods A genome-wide association study was performed adjusting for age, sex, BMI, and 8 population principal components. Significant and independent genetic variants were replicated by FinnGen. Results The primary GWAS revealed 5 significant genetic loci (including 2 novel) associated with neck or shoulder pain, with the most significant single nucleotide polymorphism (SNP) being rs9889282 (P = 2.63 × 10-12) near CA10 on chromosome 17. Two novel significant associations were detected on chromosomes 18 and 14, with the top SNPs being rs4608411 (P = 8.20 × 10-9) near TCF4 and rs370565192 (P = 3.80 × 10-8) in DCAF5, respectively. Our secondary GWAS identified a single novel genetic locus in SLC24A3 among males and 2 genetic loci (including one novel near LINC02770) among females. In the replication stage, the SLC39A8 locus was weakly supported by the FinnGen cohort. The tissue expression analysis revealed a significant association between brain tissues and neck or shoulder pain. Conclusion In summary, this study has identified novel genetic variants for neck or shoulder pain. Sex-stratified GWAS also suggested that sex played a role in the occurrence of the phenotype.
Collapse
Affiliation(s)
- Yiwen Tao
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, China
| | - Qi Pan
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, China
| | - Tengda Cai
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, China
| | - Zen Huat Lu
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Bandar Seri Begawan, Brunei Darussalam
| | - Mainul Haque
- School of Mathematical Sciences, University of Nottingham Ningbo China, Ningbo, China
| | - Tania Dottorini
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
| | - Lesley A. Colvin
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Blair H. Smith
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - Weihua Meng
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, China
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
- Center for Public Health, Faculty of Medicine, Health and Life Sciences, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
3
|
Lu G, Liu H, Wang H, Luo S, Du M, Christiani DC, Wei Q. Genetic variants of FER and SULF1 in the fibroblast-related genes are associated with non-small-cell lung cancer survival. Int J Cancer 2025; 156:2107-2117. [PMID: 39707607 PMCID: PMC11971011 DOI: 10.1002/ijc.35305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/23/2024]
Abstract
Fibroblasts are important components in the tumor microenvironment and can affect tumor progression and metastasis. However, the roles of genetic variants of the fibroblast-related genes (FRGs) in the prognosis of non-small-cell lung cancer (NSCLC) patients have not been reported. Therefore, we investigated the associations between 26,544 single nucleotide polymorphisms (SNPs) in 291 FRGs and survival of NSCLC patients from the Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial. In Cox regression multivariable analysis, we found that 661 SNPs were associated with NSCLC overall survival (OS). Then we validated these SNPs in another independent replication dataset of 984 patients from the Harvard Lung Cancer Susceptibility (HLCS) Study. Finally, we identified two independent SNPs (i.e., FER rs7716388 A>G and SULF1 rs11785839 G>C) that remained significantly associated with NSCLC survival with hazards ratios (HRs) of 0.87 (95% confidence interval [CI] = 0.77-0.98, p = 0.018) and 0.88 (95% CI = 0.79-0.99, p = 0.033), respectively. Combined analysis for these two SNPs showed that the number of protective alleles was associated with better OS and disease-specific survival. Expression quantitative trait loci analysis indicated that the FER rs7716388 G allele was associated with the up-regulation of FER mRNA expression levels in lung tissue. Our results indicated that these two functional SNPs in the FRGs may be prognostic biomarkers for the prognosis of NSCLC patients, and the possible mechanism may be through modulating the expression of their corresponding genes.
Collapse
Affiliation(s)
- Guojun Lu
- Department of Respiratory Medicine, Nanjing Chest Hospital, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA
| | - Huilin Wang
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Respiratory Oncology, Guangxi Cancer Hospital, Guangxi Medical University Cancer Hospital, Nanning, Guangxi 530021, China
| | - Sheng Luo
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Mulong Du
- Departments of Environmental Health and Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115 USA
| | - David C. Christiani
- Departments of Environmental Health and Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115 USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, Durham, NC 27710, USA
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Duke University Medical Center, Durham, Durham, NC 27710, USA
- Duke Global Health Institute, Duke University Medical Center, Durham, Durham, NC 27710, USA
| |
Collapse
|
4
|
Chan SCW, Lei Y, Yap DYH, Lee PPW, Lai WM, Ying SKY, Leung AMH, Mok CC, Lee KL, Lau CS, Yang W, Li PH. Distinct genetic risk loci between biopsy-proven renal and non-renal lupus: a 10-year longitudinal cohort. Rheumatology (Oxford) 2025; 64:4005-4013. [PMID: 39832277 DOI: 10.1093/rheumatology/keaf027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/24/2024] [Accepted: 12/28/2024] [Indexed: 01/22/2025] Open
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is a heterogeneous disease that manifests as different subphenotypes. Distinct subphenotypes, such as lupus nephritis (LN), have been associated with increased genetic risk, but prior studies have been limited by cross-sectional and imprecisely subphenotyped cohorts. This study investigated the genetic basis for LN using a longitudinal cohort of distinctly subphenotyped patients. METHODS SLE patients with biopsy-proven LN or never developed LN (NLN) were recruited from eight tertiary referral centres. All patients had longitudinal clinical data for at least 10 years, or died during the study period. NLN patients had no renal involvement for at least 10 years. Subjects were genotyped and polygenic risk scores (PRS) were calculated using 230 SLE-associated SNPs. Genome-wide association analyses were also conducted for LN vs control, NLN vs control, and LN vs NLN comparisons, along with heterogeneity tests to assess differences in effect size. RESULTS Among 1462 patients, 824 (56%) and 638 (44%) had LN and NLN, respectively. PRS was significantly higher in the LN cohort. Genome-wide significant variants were identified in HLA, TNFAIP3, BLK and STAT4 loci specifically for LN patients, while STAT4 also remained significant for NLN patients. Direct LN vs NLN associations showed no statistically significant variants but heterogeneity tests revealed other genetic loci, including ELF1, OX40, DUSP22 and TPCN2. CONCLUSION Different subphenotypes of SLE are predisposed by distinct genetic risk loci, which can only be identified in clearly subphenotyped cohorts with sufficient longitudinal data. We identified unique genetic risk loci enriched among patients with biopsy-proven LN, and postulate that individual subphenotypes may have varying genetic predisposition.
Collapse
Affiliation(s)
- Shirley C W Chan
- Division of Rheumatology & Clinical Immunology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | - Yao Lei
- Department of Paediatrics & Adolescent Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | - Desmond Y H Yap
- Division of Nephrology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | - Pamela P W Lee
- Department of Paediatrics & Adolescent Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | - Wai Ming Lai
- Department of Paediatrics & Adolescent Medicine, Princess Margaret Hospital, Hong Kong
| | - Shirley K Y Ying
- Department of Medicine & Geriatrics, Princess Margaret Hospital, Hong Kong
| | | | - Chi Chiu Mok
- Department of Medicine, Tuen Mun Hospital, Hong Kong
| | - Ka Lai Lee
- Department of Medicine, Pamela Youde Nethersole Eastern Hospital, Hong Kong
| | - Chak Sing Lau
- Division of Rheumatology & Clinical Immunology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | - Wanling Yang
- Department of Paediatrics & Adolescent Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| | - Philip H Li
- Division of Rheumatology & Clinical Immunology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong
| |
Collapse
|
5
|
Watts LM, Sparkes PC, Dewhurst HF, Guilfoyle SE, Pollard AS, Komla-Ebri D, Butterfield NC, Williams GR, Bassett JHD. The GWAS candidate far upstream element binding protein 3 (FUBP3) is required for normal skeletal growth, and adult bone mass and strength in mice. Bone 2025; 195:117472. [PMID: 40139337 DOI: 10.1016/j.bone.2025.117472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/14/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Bone mineral density (BMD) and height are highly heritable traits for which hundreds of genetic loci have been linked through genome wide association studies (GWAS). FUBP3 is a DNA and RNA binding protein best characterised as a transcriptional regulator of c-Myc, but little is known about its role in vivo. Single nucleotide polymorphisms in FUBP3 at the 9q34.11 locus have been associated with BMD, fracture and height in multiple GWAS, but FUBP3 has no previously established role in the skeleton. We analysed Fubp3-deficient mice to determine the consequence of FUBP3 deficiency in vivo. Mice lacking Fubp3 had reduced survival to adulthood and impaired skeletal growth. Bone mass was decreased, most strikingly in the vertebrae, with altered trabecular micro-architecture. Fubp3 deficient bones were also weak. These data provide the first functional demonstration that Fubp3 is required for normal skeletal growth and development and maintenance of adult bone structure and strength, indicating that FUBP3 contributes to the GWAS association of 9q34.11 with variation in height, BMD and fracture.
Collapse
Affiliation(s)
- Laura M Watts
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Penny C Sparkes
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Hannah F Dewhurst
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Siobhan E Guilfoyle
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Andrea S Pollard
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Davide Komla-Ebri
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Natalie C Butterfield
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Graham R Williams
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| | - J H Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
6
|
Miyamoto-Mikami E, Zempo H, Kumagai H, Hirata K, Takaragawa M, Yoshihara T, Fuku M, Kikuchi N, Kamiya N, Miyamoto N, Fuku N. Genome-Wide Association Study on Muscle Stiffness Identified Novel Locus for Predisposition to Muscle Strain Injury. Med Sci Sports Exerc 2025; 57:1202-1211. [PMID: 39902889 DOI: 10.1249/mss.0000000000003661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
PURPOSE We aimed to screen the entire genome for genetic variants associated with passive muscle stiffness, which has been suggested as a risk factor for muscle strain injury. METHODS This genome-wide association study (GWAS) on passive muscle stiffness included 350 physically active young Japanese individuals. Three hamstring constituents were measured using ultrasound shear wave elastography. Skeletal muscle transcriptomes were compared across the genotypes of GWAS-identified variants in 48 healthy Japanese individuals. Association between GWAS-identified variants and history of muscle strain injury was examined in 1428 Japanese athletes. RESULTS Two loci on chromosome 11 demonstrated a genome-wide significant association with passive muscle stiffness of the biceps femoris long head (rs12807854 T/C: P = 5.19 × 10 -10 , rs78405694 T/C: P = 2.09 × 10 -8 ; linear regression analysis adjusted for sex, age, and stretching exercise habits). Skeletal muscle RNA sequencing revealed significantly elevated expression of extracellular matrix-related genes in muscles carrying stiffness-increasing alleles of these variants. Among athletes, rs12807854 T/C was significantly associated with a history of muscle strain injury ( P = 0.0254; logistic regression analysis adjusted for age, sex, competitive level, and main sport). Carriers of the C allele, associated with increased muscle stiffness, exhibited a heightened risk of muscle strain injury (odds ratio = 1.62; 95% confidence interval = 1.06-2.47 per C allele increase). By contrast, rs78405694 did not show a significant association with muscle strain injury in this population. CONCLUSIONS A novel locus associated with passive muscle stiffness and muscle strain injury was identified. Elucidating the detailed mechanisms linking the identified locus to passive muscle stiffness may lead to the development of new strategies to prevent muscle strain injuries.
Collapse
Affiliation(s)
- Eri Miyamoto-Mikami
- Graduate School of Health and Sports Science, Juntendo University, Chiba, JAPAN
| | | | | | - Kosuke Hirata
- Institute of Health and Sport Sciences, University of Tsukuba, Ibaraki, JAPAN
| | | | - Toshinori Yoshihara
- Graduate School of Health and Sports Science, Juntendo University, Chiba, JAPAN
| | | | - Naoki Kikuchi
- Department of Training Science, Nippon Sport Science University, Tokyo, JAPAN
| | - Nobuhiro Kamiya
- Faculty of Budo and Sport Studies, Tenri University, Nara, JAPAN
| | - Naokazu Miyamoto
- Graduate School of Health and Sports Science, Juntendo University, Chiba, JAPAN
| | - Noriyuki Fuku
- Graduate School of Health and Sports Science, Juntendo University, Chiba, JAPAN
| |
Collapse
|
7
|
Jee YH, Wang Y, Jung KJ, Lee JY, Kimm H, Duan R, Price AL, Martin AR, Kraft P. Genome-wide association studies in a large Korean cohort identify quantitative trait loci for 36 traits and illuminate their genetic architectures. Nat Commun 2025; 16:4935. [PMID: 40436827 PMCID: PMC12120081 DOI: 10.1038/s41467-025-59950-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 05/08/2025] [Indexed: 06/01/2025] Open
Abstract
Genome-wide association studies (GWAS) have predominantly focused on European ancestry populations, limiting biological discoveries across diverse populations. Here we report GWAS findings from 153,950 individuals across 36 quantitative traits in the Korean Cancer Prevention Study-II (KCPS2) Biobank. We discovered 301 previously unreported genetic loci in KCPS2, including an association between thyroid-stimulating hormone and CD36. Meta-analysis with the Korean Genome and Epidemiology Study, Biobank Japan, Taiwan Biobank, and UK Biobank identified 4588 loci that were not significant in any contributing GWAS. We describe differences in genetic architectures across these East Asian and European samples. We also highlight East Asian specific associations, including a known pleiotropic missense variant in ALDH2, which fine-mapping identified as a likely causal variant for multiple traits. Our findings provide insights into the genetic architecture of complex traits in East Asian populations and highlight how broadening the population diversity of GWAS samples can aid discovery.
Collapse
Affiliation(s)
- Yon Ho Jee
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Ying Wang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Keum Ji Jung
- Institute for Health Promotion, Department of Epidemiology and Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea.
| | - Ji-Young Lee
- Institute for Health Promotion, Department of Epidemiology and Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| | - Heejin Kimm
- Institute for Health Promotion, Department of Epidemiology and Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| | - Rui Duan
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alkes L Price
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alicia R Martin
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Trans-Divisional Research Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Boston, MD, USA.
| |
Collapse
|
8
|
Wang Y, Zhang Y, Chen H, Zhang XJ, Zhang R, Ng TK, Tham JA, Kam KW, Tam POS, Young AL, Wei Y, Zhang M, Pang CP, Tham CC, Yam JC, Chen LJ. Genome-wide association study identified novel loci and gene-environment interaction for refractive error in children. NPJ Genom Med 2025; 10:44. [PMID: 40410244 PMCID: PMC12102314 DOI: 10.1038/s41525-025-00504-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 05/06/2025] [Indexed: 05/25/2025] Open
Abstract
To identify novel genetic loci for children refractive error, we performed a meta-analysis of two genome-wide association studies (GWASs) of spherical equivalent (SE) in 1,237 children from the population-based Hong Kong Children Eye Study (HKCES) and the Low Concentration Atropine for Myopia Progression (LAMP) study. Replication was conducted in 4,093 Chinese children and 1,814 Chinese adults. Four lead-SNPs (MIR4275 rs292034, TENM3 rs17074027, LOC101928911 rs6925312 and FAM135B rs4609227) showed genome-wide significant association (P ≤ 5.0 × 10-8) with SE. TENM3 had been associated with myopia in adults before, whilst the other three loci, MIR4275, LOC101928911 and FAM135B, were novel. Significant interaction between genetic risk scores (GRS) and near work on SE was also detected (βinteraction = 0.14, Pinteraction = 0.0003). This study identified novel genetic loci for children refractive error and suggested myopia intervention can be individualized based on the genetic risk of children.
Collapse
Affiliation(s)
- Yuyao Wang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yuzhou Zhang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Haoyu Chen
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Guangdong, Shantou, China
| | - Xiu Juan Zhang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Queen Mary Hospital, Pok Fu Lam, Hong Kong, China
| | - Riping Zhang
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Guangdong, Shantou, China
| | - Tsz Kin Ng
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Guangdong, Shantou, China
| | - Jenson A Tham
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ka Wai Kam
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, Hong Kong, China
| | - Pancy O S Tam
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Alvin L Young
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, Hong Kong, China
| | - Yingying Wei
- Department of Statistics, The Chinese University of Hong Kong, Hong Kong, China
| | - Mingzhi Zhang
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Guangdong, Shantou, China
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Guangdong, Shantou, China
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong, China
| | - Clement C Tham
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Eye Hospital, Hong Kong, China
| | - Jason C Yam
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China.
- Joint Shantou International Eye Center of Shantou University and the Chinese University of Hong Kong, Guangdong, Shantou, China.
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong, China.
- Hong Kong Eye Hospital, Hong Kong, China.
| | - Li Jia Chen
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China.
- Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, Hong Kong, China.
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
9
|
Polesskaya O, Boussaty E, Cheng R, Lamonte OA, Zhou TY, Du E, Sanches TM, Nguyen KM, Okamoto M, Palmer AA, Friedman R. Genome-Wide Association Study of Age-Related Hearing Loss in CFW Mice Identifies Multiple Genes and Loci, Including Prkag2. J Assoc Res Otolaryngol 2025:10.1007/s10162-025-00994-1. [PMID: 40399499 DOI: 10.1007/s10162-025-00994-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 04/30/2025] [Indexed: 05/23/2025] Open
Abstract
PURPOSE Age-related hearing loss (ARHL) is one of the most prevalent conditions affecting the elderly. ARHL is influenced by a combination of environmental and genetic factors; the identification of the genes that confer risk will aid in the prevention and treatment of ARHL. The mouse and human inner ears are functionally and genetically homologous. We used Carworth Farms White (CFW) mice to study the genetic basis of ARHL because they are genetically diverse and exhibit variability in the age of onset and severity of ARHL. METHODS Hearing at a range of frequencies was measured using auditory brainstem response (ABR) thresholds in 946 male and female CFW mice at the age of 1, 6, and 10 months. We genotyped the mice using low-coverage (mean coverage 0.27 ×) whole-genome sequencing (lcWGS) followed by imputation using STITCH. To determine the accuracy of the genotypes, we sequenced 8 samples at > 30 × coverage and used those data to estimate the accuracy of lcWGS genotyping, which was > 99.5%. We performed a genome-wide association study (GWAS) for the ABR thresholds for each frequency at each age, and we also performed a GWAS for age at deafness. RESULTS We obtained genotypes at 4.18 million single nucleotide polymorphisms (SNP). The SNP heritability for traits ranged from 0 to 42%. GWAS identified 10 significant associations with ARHL that contained potential candidate genes, including Dnah11, Rapgef5, Cpne4, Prkag2, and Nek11. Genetic ablation of Prkag2 caused ARHL at high frequencies, strongly suggesting that Prkag2 is the causal gene for one of the associations. CONCLUSIONS GWAS for ARHL in CFW outbred mice identified genetic risk factors for ARHL, including Prkag2. Our results will help to define novel therapeutic targets for the treatment and prevention of this common disorder.
Collapse
Affiliation(s)
- Oksana Polesskaya
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Ely Boussaty
- Department of Otolaryngology - Head and Neck Surgery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Riyan Cheng
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Olivia A Lamonte
- Department of Otolaryngology - Head and Neck Surgery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Thomas Y Zhou
- Department of Otolaryngology - Head and Neck Surgery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Eric Du
- Department of Otolaryngology - Head and Neck Surgery, University of California San Diego, La Jolla, CA, 92093, USA
| | | | - Khai-Minh Nguyen
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Mika Okamoto
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Rick Friedman
- Department of Otolaryngology - Head and Neck Surgery, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
10
|
Thakur R, Xu M, Sowards H, Yon J, Jessop L, Myers T, Zhang T, Chari R, Long E, Rehling T, Hennessey R, Funderburk K, Yin J, Machiela MJ, Johnson ME, Wells AD, Chesi A, Grant SFA, Iles MM, Landi MT, Law MH, Melanoma Meta-Analysis Consortium, Choi J, Brown KM. Mapping chromatin interactions at melanoma susceptibility loci uncovers distant cis-regulatory gene targets. Am J Hum Genet 2025:S0002-9297(25)00178-8. [PMID: 40409268 DOI: 10.1016/j.ajhg.2025.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 04/25/2025] [Accepted: 04/28/2025] [Indexed: 05/25/2025] Open
Abstract
Genome-wide association studies (GWASs) of melanoma risk have identified 68 independent signals at 54 loci. For most loci, specific functional variants and their respective target genes remain to be established. Capture-HiC is an assay that links fine-mapped risk variants to candidate target genes by comprehensively mapping chromatin interactions. We performed a melanoma GWAS region-focused capture-HiC assay in human primary melanocytes to identify physical interactions between fine-mapped risk variants and potential causal melanoma-susceptibility genes. Overall, chromatin-interaction data alone nominated potential causal genes for 61 of the 68 melanoma risk signals, identifying many candidates beyond those reported by previous studies. We further integrated these data with epigenomic (chromatin state, accessibility), gene expression (expression quantitative trait locus [eQTL]/transcriptome-wide association study [TWAS]), DNA methylation (methylation QTL [meQTL]/methylome-wide association study [MWAS]), and massively parallel reporter assay (MPRA) data generated from melanoma-relevant cell types to prioritize potentially cis-regulatory variants and their respective candidate gene targets. From the set of fine-mapped variants across these loci, we identified 140 prioritized credible causal variants linked to 195 candidate genes at 42 risk signals. In addition, we developed an integrative scoring system to facilitate candidate gene prioritization, integrating melanocyte and melanoma datasets. Notably, at several GWAS risk signals, we observed long-range chromatin connections (500 kb to >1 Mb) with distant candidate target genes. We validated several such cis-regulatory interactions using CRISPR inhibition, providing evidence for known cancer driver genes MDM4 and CBL, as well as the SRY-box transcription factor SOX4, as likely melanoma risk genes.
Collapse
Affiliation(s)
- Rohit Thakur
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Mai Xu
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Hayley Sowards
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Joshuah Yon
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Lea Jessop
- Laboratory of Genetic Susceptibility, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Timothy Myers
- Laboratory of Genetic Susceptibility, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Tongwu Zhang
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Raj Chari
- Genome Modification Core, Frederick National Lab for Cancer Research, Frederick, MD, USA
| | - Erping Long
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA; Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Thomas Rehling
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Rebecca Hennessey
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Karen Funderburk
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jinhu Yin
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Mitchell J Machiela
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Matthew E Johnson
- Division of Human Genetics, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Andrew D Wells
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alessandra Chesi
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Struan F A Grant
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mark M Iles
- Leeds Institute for Data Analytics, University of Leeds, Leeds, UK; NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Maria Teresa Landi
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Matthew H Law
- Population Health Department, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia; School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | | | - Jiyeon Choi
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Kevin M Brown
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
11
|
Sharma SD, Hum RM, Nair N, Marshall L, Storrie A, Bowes J, MacGregor A, Yates M, Morris AP, Verstappen S, Barton A, van Steenbergen H, Knevel R, van der Helm-van Mil A, Viatte S. Systematic review and independent validation of genetic factors of radiographic outcome in rheumatoid arthritis identifies a genome-wide association with CARD9. Ann Rheum Dis 2025:S0003-4967(25)00897-0. [PMID: 40345877 DOI: 10.1016/j.ard.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/26/2025] [Accepted: 04/05/2025] [Indexed: 05/11/2025]
Abstract
OBJECTIVES This study aimed to investigate non-HLA genetic mechanisms underlying radiographic severity in rheumatoid arthritis (RA). METHODS A systematic review of publications reporting non-HLA genetic associations with radiographic severity in RA across ancestries was undertaken. Experimental validation was performed in the Norfolk Arthritis Register, comprising 1407 patients with available genetic and treatment data followed prospectively for up to 10 years, with 2198 longitudinal radiographs. Genome-wide genotyping was performed with Illumina Human Core Exome Array. Radiographic outcomes (presence of erosions; Larsen score) were modelled longitudinally. Fine mapping and functional annotations to refine associations to potential causative loci were undertaken using FUMA, PolyPhen2, and RegulomeDB. RESULTS The systematic review identified 102 publications reporting 139 independent associations with radiographic outcome. Association with 15 independent polymorphisms were replicated in the Norfolk Arthritis Register data set, implicating adaptive immune processes (Th1, Th2, and Th17 pathways), cytokine regulation, and osteoclast differentiation. Notably, we refined the association of rs59902911 at the CARD9 locus to an intronic polymorphism within an active enhancer (rs78892335), achieving genome-wide significance and with an effect size exceeding the minimal clinically important difference for each copy of the minor allele (4.78 Larsen units/copy; 95% CI, 3.15-6.41; p = 9.01 × 10-9). This polymorphism is associated with the expression of CARD9 in immune cells, including B cells. CONCLUSIONS We provide a comprehensive list of validated genetic associations with RA outcome and demonstrate that non-HLA polymorphisms can associate with radiographic severity independently of disease susceptibility. This highlights the importance of dedicated genetic outcome studies for patient stratification in precision medicine for RA.
Collapse
Affiliation(s)
- Seema Devi Sharma
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Ryan Malcolm Hum
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Nisha Nair
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Lysette Marshall
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom
| | - Alice Storrie
- Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - John Bowes
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Alexander MacGregor
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom; Department of Rheumatology, Norfolk and Norwich University Hospital, United Kingdom
| | - Max Yates
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom; Department of Rheumatology, Norfolk and Norwich University Hospital, United Kingdom
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Suzanne Verstappen
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom; Centre for Epidemiology Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom
| | - Anne Barton
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Hanna van Steenbergen
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rachel Knevel
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Sebastien Viatte
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom; Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
12
|
Hu Y, Liu W, Wu Y, Hu Z, Tao Y, Zhang Q, Chen J, Li M, Hu L, Ding Y. DCC in the cerebral cortex is required for cognitive functions in mouse. Brain Pathol 2025; 35:e13306. [PMID: 39293934 PMCID: PMC11961207 DOI: 10.1111/bpa.13306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 08/26/2024] [Indexed: 09/20/2024] Open
Abstract
Schizophrenia (SZ) is a highly heritable mental disorder, and genome-wide association studies have identified the association between deleted in colorectal cancer (DCC) and SZ. Previous study has shown a lowered expression of DCC in the cerebral cortex of SZ patient. In this study, we identified novel single nucleotide polymorphisms (SNPs) of DCC statistically correlated with SZ. Based on these, we generated DCC conditional knockout (CKO) mice and explored behavioral phenotypes in these mice. We observed that deletion of DCC in cortical layer VI but not layer V led to deficits in fear and spatial memory, as well as defective sensorimotor gating revealed by the prepulse inhibition test (PPI). Critically, the defective sensorimotor gating could be restored by olanzapine, an antipsychotic drug. Furthermore, we found that the levels of p-AKT and p-GSK3α/β were decreased, which was responsible for impaired PPI in the DCC-deficient mice. Finally, the DCC-deficient mice also displayed reduced spine density of pyramidal neurons and disturbed delta-oscillations. Our data, for the first time, identified and explored downstream substrates and signaling pathway of DCC which supports the hypothesis that DCC is a SZ-related risky gene and when defective, may promote SZ-like pathogenesis and behavioral phenotypes in mice.
Collapse
Affiliation(s)
- Yun‐Qing Hu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Wei‐Tang Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain ScienceFudan UniversityShanghaiChina
- Shanghai Institute of Infectious Disease and BiosecurityFudan UniversityShanghaiChina
| | - Yong Wu
- Research Center for Mental Health and Neuroscience, Wuhan Mental Health CenterJianghan UniversityWuhanChina
| | - Zhi‐Bin Hu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Yun‐Chao Tao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain ScienceFudan UniversityShanghaiChina
| | - Qiong Zhang
- Department of Laboratory Animal ScienceFudan UniversityShanghaiChina
| | - Jia‐Yin Chen
- Department of Laboratory Animal ScienceFudan UniversityShanghaiChina
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of ZoologyChinese Academy of SciencesKunmingChina
| | - Ling Hu
- Department of Laboratory Animal ScienceFudan UniversityShanghaiChina
| | - Yu‐Qiang Ding
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain ScienceFudan UniversityShanghaiChina
- Department of Laboratory Animal ScienceFudan UniversityShanghaiChina
| |
Collapse
|
13
|
Bartusel M, Kim SX, Rehimi R, Darnell AM, Nikolić M, Heggemann J, Kolovos P, van Ijcken WFJ, Varineau J, Crispatzu G, Mangold E, Brugmann SA, Vander Heiden MG, Laugsch M, Ludwig KU, Rada-Iglesias A, Calo E. A non-syndromic orofacial cleft risk locus links tRNA splicing defects to neural crest cell pathologies. Am J Hum Genet 2025; 112:1097-1116. [PMID: 40250422 PMCID: PMC12120183 DOI: 10.1016/j.ajhg.2025.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/20/2025] Open
Abstract
Orofacial clefts are the most common form of congenital craniofacial malformation worldwide. The etiology of these birth defects is multifactorial, involving genetic and environmental factors. However, in most cases, the underlying causes remain unexplained, precluding a molecular understanding of disease mechanisms. Here, we integrated genome-wide association data, targeted resequencing of case and control cohorts, tissue- and cell-type-specific epigenomic profiling, and genome architecture analyses to molecularly dissect a genomic locus associated with an increased risk of non-syndromic orofacial cleft. We found that common and rare risk variants associated with orofacial cleft intersect with an enhancer (e2p24.2) that is active in human embryonic craniofacial tissue. We mapped e2p24.2 long-range interactions to a topologically associated domain harboring MYCN, DDX1, and CYRIA. We found that MYCN and DDX1, but not CYRIA, are required during craniofacial development in chicken embryos. We investigated the role of DDX1, a key component of the tRNA splicing complex, in cranial neural crest cells (cNCCs). The loss of DDX1 in cNCCs resulted in the accumulation of unspliced tRNA fragments, depletion of mature intron-containing tRNAs, and ribosome stalling at codons decoded by these tRNAs. This was accompanied by defects in both global protein synthesis and cNCC migration. We further showed that the induction of tRNA fragments is sufficient to disrupt craniofacial development. Together, these results uncovered a molecular mechanism in which impaired tRNA splicing affects cNCCs and craniofacial development and positioned MYCN, DDX1, and tRNA processing defects as risk factors in the pathogenesis of orofacial clefts.
Collapse
Affiliation(s)
- Michaela Bartusel
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| | - Skylar X Kim
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rizwan Rehimi
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Institute for Pharmacology and Toxicology (IPT), University Hospital Bonn, Venusberg-Campus 1, Biomedical Center 1 (Building 13), 53127 Bonn, Germany
| | - Alicia M Darnell
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Miloš Nikolić
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Julia Heggemann
- Institute of Human Genetics, University of Bonn, University Hospital Bonn, Medical Faculty, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Petros Kolovos
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | | | - Jade Varineau
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Giuliano Crispatzu
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Elisabeth Mangold
- Institute of Human Genetics, University of Bonn, University Hospital Bonn, Medical Faculty, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Samantha A Brugmann
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MLC 7007, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Matthew G Vander Heiden
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Dana-Farber Cancer Institute, Boston, MA, USA
| | - Magdalena Laugsch
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Kerstin U Ludwig
- Institute of Human Genetics, University of Bonn, University Hospital Bonn, Medical Faculty, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Alvaro Rada-Iglesias
- Institute of Biomedicine and Biotechnology of Cantabria (IBBTEC), CSIC/University of Cantabria, Santander, Spain
| | - Eliezer Calo
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
14
|
Somerville EN, Krohn L, Senkevich K, Yu E, Ahmad J, Asayesh F, Ruskey JA, Spiegelman D, Fahn S, Waters C, Sardi SP, Alcalay RN, Gan-Or Z. Genome-Wide Association Study of Glucocerebrosidase Activity Modifiers. Mol Neurobiol 2025:10.1007/s12035-025-04996-1. [PMID: 40299299 DOI: 10.1007/s12035-025-04996-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 04/23/2025] [Indexed: 04/30/2025]
Abstract
One of the most common genetic risk factors for Parkinson's disease (PD) is variants in GBA1, which encodes the lysosomal enzyme glucocerebrosidase (GCase). GCase deficiency has been associated with an increased PD risk, but not all individuals with low GCase activity are carriers of GBA1 mutations, suggesting other factors may be acting as modifiers. We aimed to discover common variants associated with GCase activity, as well as replicate previously reported associations, by performing a genome-wide association study using two independent cohorts: a Columbia University cohort consisting of 697 PD cases and 347 controls and the Parkinson's Progression Markers Initiative (PPMI) cohort consisting of 357 PD cases and 163 controls. As expected, GBA1 variants have the strongest association with decreased activity, led by N370S (beta = - 4.36, se = 0.32, p = 5.05e - 43). We also identify a novel association in the GAA locus (encoding for acid alpha-glucosidase, beta = - 0.96, se = 0.17, p = 5.23e - 09) that may be the result of an interaction between GCase and acid alpha-glucosidase based on various interaction analyses. Lastly, we show that several PD-risk loci are potentially associated with GCase activity. Further research will be needed to replicate and validate our findings and to uncover the functional connection between acid alpha-glucosidase and GCase.
Collapse
Affiliation(s)
- Emma N Somerville
- The Neuro (Montréal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Lynne Krohn
- The Neuro (Montréal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Konstantin Senkevich
- The Neuro (Montréal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue WestRoom 312, Ludmer PavilionMontréal, QC, H3 A 1 A1, Canada
| | - Eric Yu
- The Neuro (Montréal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Jamil Ahmad
- The Neuro (Montréal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue WestRoom 312, Ludmer PavilionMontréal, QC, H3 A 1 A1, Canada
| | - Farnaz Asayesh
- The Neuro (Montréal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue WestRoom 312, Ludmer PavilionMontréal, QC, H3 A 1 A1, Canada
| | - Jennifer A Ruskey
- The Neuro (Montréal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue WestRoom 312, Ludmer PavilionMontréal, QC, H3 A 1 A1, Canada
| | - Dan Spiegelman
- The Neuro (Montréal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue WestRoom 312, Ludmer PavilionMontréal, QC, H3 A 1 A1, Canada
| | - Stanley Fahn
- Department of Neurology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Cheryl Waters
- Department of Neurology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - S Pablo Sardi
- Rare and Neurological Diseases Therapeutic Area, Sanofi, Cambridge, MA, USA
| | - Roy N Alcalay
- Department of Neurology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
- Taub Institute for Research On Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA
| | - Ziv Gan-Or
- The Neuro (Montréal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada.
- Department of Human Genetics, McGill University, Montréal, QC, Canada.
- Department of Neurology and Neurosurgery, McGill University, 1033 Pine Avenue WestRoom 312, Ludmer PavilionMontréal, QC, H3 A 1 A1, Canada.
| |
Collapse
|
15
|
Huang H, Liu J, Xiao Q, Mao C, She L, Yu L, Yu B, Lei M, Gao Y, He B, Pan P, Li X, Yin J, Liu Z. GWAS study of myelosuppression among NSCLC patients receiving platinum-based combination chemotherapy. Acta Biochim Biophys Sin (Shanghai) 2025. [PMID: 40296719 DOI: 10.3724/abbs.2025013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
Platinum-based chemotherapy remains the mainstay for non-small cell lung cancer (NSCLC), but it frequently causes dose-limiting myelosuppression, with significant individual variability in susceptibility. However, the genetic basis of myelosuppression side effects remains elusive, greatly hindering personalized therapeutic approaches. In this study, we perform a comprehensive genome-wide association analysis on 491 NSCLC patients receiving platinum-based chemotherapy, examining 4,690,998 single-nucleotide polymorphisms (SNPs) to identify relevant genetic variants. LDBlockShow, FUMA, and MAGMA are utilized to explore linkage disequilibrium, expression quantitative trait loci (eQTLs), chromatin interaction, and conduct gene-based and gene set-based analysis of candidate SNPs. The GWAS results reveal that rs6856089 and its linked SNPs are significantly associated with platinum-based chemotherapy-induced myelosuppression. Specifically, patients with the A allele of rs6856089 have a significantly lower risk of myelosuppression (odds ratio (OR) = 0.1300, P = 7.59 × 10 -8). Furthermore, gene-based analysis reveals that EMCN ( P = 2.47 × 10 -5), which encodes endomucin, a marker for hematopoietic stem cells, might mediate myelosuppression. This study provides a scientific basis for the individual differences in platinum-based chemotherapy-induced myelosuppression.
Collapse
Affiliation(s)
- Hanxue Huang
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, China
| | - Junyan Liu
- Departments of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qi Xiao
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, China
| | - Chenxue Mao
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, China
| | - Lei She
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, China
| | - Lulu Yu
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, China
| | - Bing Yu
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, China
| | - Mengrong Lei
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, China
| | - Ying Gao
- Departments of Gerontology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Baimei He
- Departments of Gerontology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Pinhua Pan
- Departments of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xi Li
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, China
| | - Jiye Yin
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, China
| | - Zhaoqian Liu
- Department of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute of Clinical Pharmacology, Engineering Research Center for Applied Technology of Pharmacogenomics of Ministry of Education, Central South University, Changsha 410078, China
| |
Collapse
|
16
|
Sakai T, Furutani M, Nakashima M, Ishibashi N, Maeda J, Oguri N, Miyamoto S, Miyauchi S, Okamura S, Okubo Y, Tokuyama T, Oda N, Mitsumori R, Niida S, Ozaki K, Shigemizu D, Nakano Y. Genome-Wide Association Study of Atrial Fibrillation Recurrence After Radiofrequency Catheter Ablation in a Japanese Population. J Cardiovasc Electrophysiol 2025. [PMID: 40296270 DOI: 10.1111/jce.16674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/14/2025] [Accepted: 03/26/2025] [Indexed: 04/30/2025]
Abstract
INTRODUCTION Catheter ablation of atrial fibrillation (AF) is an established treatment; however, recurrence remains a major issue. METHODS This study included 606 patients with AF who underwent catheter ablation in Hiroshima University Hospital, including 143 and 463 patients with and without AF recurrence within 3 years after ablation, respectively. A logistic regression analysis and genome-wide association study (GWAS) were conducted to identify the clinical and genetic factors, respectively, associated with the AF recurrence risk. RESULTS A logistic regression analysis revealed persistent AF, nonpulmonary vein AF triggers, the N-terminal pro-brain natriuretic peptide level, and the left atrial volume before catheter ablation were significant factors for recurrence (false discovery rate < 0.05). Additionally, six variants (rs2106865, rs12577119, rs12574466, rs4902609, rs8027532, and rs2032303) were associated with the AF recurrence risk, based on the GWAS results' suggestive significance (p < 5 × 10-6). An expression quantitative trait locus analysis revealed a significant association between rs2106865 and ATP-binding cassette subfamily C member 8 (ABCC8) expression in heart tissues (atrial appendage and left ventricle). A linear regression analysis demonstrated a significant association between the ABCC8 variant rs2106865 and left atrium volume before ablation. Finally, the Cox proportional hazard model showed a significant association between the ABCC8 variant and AF recurrence (p = 1.30 × 10-5 by log-rank test; hazard ratio 1.74, 95% confidence interval 1.38-2.20). CONCLUSIONS Our findings, which highlight both the clinical and genetic factors associated with AF recurrence in Japanese patients, may contribute to future efforts to improve treatment strategies for AF.
Collapse
Affiliation(s)
- Takumi Sakai
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Motoki Furutani
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Mika Nakashima
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Naoki Ishibashi
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Junji Maeda
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Naoto Oguri
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Shogo Miyamoto
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Shunsuke Miyauchi
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Sho Okamura
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yousaku Okubo
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Takehito Tokuyama
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Noboru Oda
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Risa Mitsumori
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Shumpei Niida
- Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Kouichi Ozaki
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Daichi Shigemizu
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
- Medical Genome Center, Research Institute, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Yukiko Nakano
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| |
Collapse
|
17
|
Park JH, Park KJ. Genetic variants associated with metabolic dysfunction-associated fatty liver diseases in a Korean population. Eur J Med Res 2025; 30:318. [PMID: 40264238 PMCID: PMC12016408 DOI: 10.1186/s40001-025-02576-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Accepted: 04/09/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Susceptibility to metabolic dysfunction-associated fatty liver diseases (MAFLD) shows a large inter-ethnic variability. Currently, large-scale genome-wide association studies (GWAS) on MAFLD in a Korean population are limited. This study aimed to investigate genes underlying MAFLD in a Korean population. METHODS A total of 13,457 Korean adults (4061 cases and 9396 controls) who underwent abdominal ultrasonography, biochemical testing, and genetic studies at a comprehensive health promotion center from 2019 to 2023 were included. Genome-wide genotyping was conducted using Infinium Asian Screening Array and an iSCAN system (Illumina, San Diego, CA, USA). Gene-based approach was conducted with Multi-Marker Analysis for Genomic Annotation (MAGMA) and Functional Mapping and Annotation (FUMA). Expression quantitative trait loci (eQTLs) mapping was done using GTEx v8 data. RESULTS The 22q13.3, 19p13.11, and 2p23.3 loci were associated with MAFLD after adjusting for age, sex, and body mass index (p < 5 × 10-8). Of these, 154 (89%) variants were identified as eQTLs (FDR < 0.05). Gene-based approach showed that PNPLA3, SAMM50, and PARVB were significantly associated with MAFLD (Bonferroni-corrected p < 2.99 × 10-6), followed by PDLIM4, GCKR, APOB, GPAM, HMGA1, C5orf56, and APOC1. CONCLUSIONS This is the largest-scale GWAS of MAFLD in a Korean adult population. Genotyping PARVB as well as PNPLA3 might help us identify individuals with the highest risk of MAFLD in Korean adults. These findings would contribute to our understanding of genetic pathogenesis of MAFLD in the Korean population.
Collapse
Affiliation(s)
- Jong-Ho Park
- Precision Medicine Center, Seoul National University Bundang Hospital, Seongnam, South Korea
- Department of Genomic Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Kyoung-Jin Park
- Department of Laboratory Medicine & Genetics, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, South Korea.
| |
Collapse
|
18
|
Guo Y, Zheng W, Yue T, Baimakangzhuo, Qi X, Liu K, Li L, He Y, Su B. GCH1 contributes to high-altitude adaptation in Tibetans by regulating blood nitric oxide. J Genet Genomics 2025:S1673-8527(25)00114-6. [PMID: 40254159 DOI: 10.1016/j.jgg.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 04/22/2025]
Abstract
Nitric oxide (NO) is a key vasodilator that regulates vascular pressure and blood flow. Tibetans have developed a "blunted" mechanism for regulating NO levels at high altitude, with GTP cyclohydrolase 1 (GCH1) identified as a key candidate gene. Here, we present comprehensive genetic and functional analyses of GCH1, which exhibits strong Darwinian positive selection in Tibetans. We show that Tibetan-enriched GCH1 variants down-regulate its expression in the blood of Tibetans. Based on this observation, we generate the heterozygous Gch1 knockout (Gch1+/-) mouse model to simulate its downregulation in Tibetans. We find that under prolonged hypoxia, the Gch1+/- mice have relatively higher blood NO and blood oxygen saturation levels compared to the wild-type (WT) controls, providing better oxygen supplies to the cardiovascular and pulmonary systems. Markedly, hypoxia-induced cardiac hypertrophy and pulmonary remodeling are significantly attenuated in the Gch1+/- mice compared with the WT controls, likely due to the adaptive changes in molecular regulations related to metabolism, inflammation, circadian rhythm, extracellular matrix, and oxidative stress. This study sheds light on the role of GCH1 in regulating blood NO, contributing to the physiological adaptation of the cardiovascular and pulmonary systems in Tibetans at high altitude.
Collapse
Affiliation(s)
- Yongbo Guo
- State Key Laboratory of Genetic Resources and Evolution, and Yunnan Key Laboratory of Integrative Anthropology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Wangshan Zheng
- State Key Laboratory of Genetic Resources and Evolution, and Yunnan Key Laboratory of Integrative Anthropology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100101, China; School of Biological and Pharmaceutical Engineering, Lanzhou Jiaotong University, Lanzhou, Gansu 730070, China
| | - Tian Yue
- State Key Laboratory of Genetic Resources and Evolution, and Yunnan Key Laboratory of Integrative Anthropology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Baimakangzhuo
- High Altitude Medical Research Center, School of Medicine, Tibetan University, Lhasa, Xizang 850000, China
| | - Xuebin Qi
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650223, China
| | - Kai Liu
- State Key Laboratory of Genetic Resources and Evolution, and Yunnan Key Laboratory of Integrative Anthropology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Liya Li
- State Key Laboratory of Genetic Resources and Evolution, and Yunnan Key Laboratory of Integrative Anthropology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Yaoxi He
- State Key Laboratory of Genetic Resources and Evolution, and Yunnan Key Laboratory of Integrative Anthropology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.
| | - Bing Su
- State Key Laboratory of Genetic Resources and Evolution, and Yunnan Key Laboratory of Integrative Anthropology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, Yunnan 650223, China.
| |
Collapse
|
19
|
Herzig AF, Rubinacci S, Marenne G, Perdry H, Deleuze JF, Dina C, Barc J, Redon R, Delaneau O, Génin E. SURFBAT: a surrogate family based association test building on large imputation reference panels. G3 (BETHESDA, MD.) 2025; 15:jkae287. [PMID: 39657733 PMCID: PMC12005154 DOI: 10.1093/g3journal/jkae287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/07/2024] [Accepted: 11/29/2024] [Indexed: 12/12/2024]
Abstract
Genotype-phenotype association tests are typically adjusted for population stratification using principal components that are estimated genome-wide. This lacks resolution when analyzing populations with fine structure and/or individuals with fine levels of admixture. This can affect power and precision, and is a particularly relevant consideration when control individuals are recruited using geographic selection criteria. Such is the case in France where we have recently created reference panels of individuals anchored to different geographic regions. To make correct comparisons against case groups, who would likely be gathered from large urban areas, new methods are needed. We present SURFBAT (a surrogate family based association test), which performs an approximation of the transmission-disequilibrium test. Our method hinges on the application of genotype imputation algorithms to match similar haplotypes between the case and control groups. This permits us to approximate local ancestry informed posterior probabilities of un-transmitted parental alleles of each case individual. This is achieved by assuming haplotypes from the imputation panel are well-matched for ancestry with the case individuals. When the first haplotype of an individual from the imputation panel matches that of a case individual, it is assumed that the second haplotype of the same reference individual can be used as a locally ancestry matched control haplotype and to approximately impute un-transmitted parental alleles. SURFBAT provides an association test that is inherently robust to fine-scale population stratification and opens up the possibility of efficiently using large imputation reference panels as control groups for association testing. In contrast to other methods for association testing that incorporate local-ancestry inference, SURFBAT does not require a set of ancestry groups to be defined, nor for local ancestry to be explicitly estimated. We demonstrate the interest of our tool on simulated datasets, as well as on a real-data example for a group of case individuals affected by Brugada syndrome.
Collapse
Affiliation(s)
- Anthony F Herzig
- Inserm, Université de Bretagne-Occidentale, EFS, UMR 1078, GGB, Brest F-29200, France
| | - Simone Rubinacci
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki 00290, Finland
| | - Gaëlle Marenne
- Inserm, Université de Bretagne-Occidentale, EFS, UMR 1078, GGB, Brest F-29200, France
| | - Hervé Perdry
- CESP Inserm U1018, Université Paris-Saclay, Villejuif F-94807, France
| | - Jean-François Deleuze
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), Evry F-91000, France
- CEPH, Fondation Jean Dausset, Paris F-75010, France
| | - Christian Dina
- Nantes Université, CNRS, INSERM UMR 1087, L’Institut du Thorax, Nantes F-44000, France
| | - Julien Barc
- Nantes Université, CNRS, INSERM UMR 1087, L’Institut du Thorax, Nantes F-44000, France
| | - Richard Redon
- Nantes Université, CNRS, INSERM UMR 1087, L’Institut du Thorax, Nantes F-44000, France
| | | | - Emmanuelle Génin
- Inserm, Université de Bretagne-Occidentale, EFS, UMR 1078, GGB, Brest F-29200, France
- CHU Brest, Brest F-29200, France
| |
Collapse
|
20
|
Hayat M, Chen WC, Babb de Villiers C, Hyuck Lee S, Curtis C, Newton R, Waterboer T, Sitas F, Bradshaw D, Muchengeti M, Singh E, Lewis CM, Ramsay M, Mathew CG, Brandenburg JT. Genome-wide association study identifies common variants associated with breast cancer in South African Black women. Nat Commun 2025; 16:3542. [PMID: 40229280 PMCID: PMC11997036 DOI: 10.1038/s41467-025-58789-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 04/01/2025] [Indexed: 04/16/2025] Open
Abstract
Genome-wide association studies (GWAS) have characterized the contribution of common variants to breast cancer (BC) risk in populations of European ancestry, however GWAS have not been reported in resident African populations. This GWAS included 2485 resident African BC cases and 1101 population matched controls. Two risk loci were identified, located between UNC13C and RAB27A on chromosome 15 (rs7181788, p = 1.01 × 10-08) and in USP22 on chromosome 17 (rs899342, p = 4.62 × 10-08). Several genome-wide significant signals were also detected in hormone receptor subtype analysis. The novel loci did not replicate in BC GWAS data from populations of West Africa ancestry suggesting genetic heterogeneity in different African populations, but further validation of these findings is needed. A European ancestry derived polygenic risk model for BC explained only 0.79% of variance in our data. Larger studies in pan-African populations are needed to further define the genetic contribution to BC risk.
Collapse
Affiliation(s)
- Mahtaab Hayat
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
- School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, South Africa.
| | - Wenlong C Chen
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- National Cancer Registry, National Health Laboratory Service, Johannesburg, South Africa
- Strengthening Oncology Services Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Chantal Babb de Villiers
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sang Hyuck Lee
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- National Institute for Health and Care Research Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London, UK
| | - Charles Curtis
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- National Institute for Health and Care Research Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London, UK
| | - Rob Newton
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- University of York, University of York, York, UK
| | - Tim Waterboer
- Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Freddy Sitas
- Burden of Disease Research Unit, South African Medical Research Council, Cape Town, South Africa
- UNSW International Centre for Future Health Systems, Sydney, NSW, Australia
- School of Population Health, University of New South Wales, Sydney, NSW, Australia
| | - Debbie Bradshaw
- Burden of Disease Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Mazvita Muchengeti
- National Cancer Registry, National Health Laboratory Service, Johannesburg, South Africa
- South African DSI-NRF Centre of Excellence in Epidemiological Modelling and Analysis (SACEMA), Stellenbosch University, Stellenbosch, South Africa
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Elvira Singh
- National Cancer Registry, National Health Laboratory Service, Johannesburg, South Africa
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Cathryn M Lewis
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Michele Ramsay
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Christopher G Mathew
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Jean-Tristan Brandenburg
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
- Strengthening Oncology Services Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
21
|
Fusco D, Marinelli C, André M, Troiani L, Noè M, Pizzagalli F, Marnetto D, Provero P. Exploring the molecular basis of the genetic correlation between body mass index and brain morphological traits. PLoS Genet 2025; 21:e1011658. [PMID: 40209151 PMCID: PMC12048161 DOI: 10.1371/journal.pgen.1011658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 05/02/2025] [Accepted: 03/17/2025] [Indexed: 04/12/2025] Open
Abstract
Several studies have demonstrated significant phenotypic and genetic correlations between body mass index (BMI) and brain morphological traits derived from structural magnetic resonance imaging (sMRI). We use the sMRI, BMI, and genetic data collected by the UK Biobank to systematically compute the genetic correlations between area, volume, and thickness measurements of hundreds of brain structures on one hand, and BMI on the other. In agreement with previous literature, we find many such measurements to have negative genetic correlation with BMI. We then dissect the molecular mechanisms underlying such correlations using brain eQTL data and summary-based Mendelian randomization, thus producing an atlas of genes whose genetically regulated expression in brain tissues is pleiotropic with brain morphology and BMI. Fine-mapping followed by colocalization analysis allows, in several cases, the identification of credible sets of variants likely to be causal for both the macroscopic phenotypes and for gene expression. In particular, epigenetic fine mapping identifies variant rs7187776 in the 5' UTR of the TUFM gene as likely to be causal of increased BMI and decreased caudate volume, possibly through the creation, by the alternate allele, of an ETS binding site leading to increased chromatin accessibility, specifically in microglial cells.
Collapse
Affiliation(s)
- Daniela Fusco
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | - Camilla Marinelli
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | - Mathilde André
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | - Lucia Troiani
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | - Martina Noè
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | - Fabrizio Pizzagalli
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | - Davide Marnetto
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | - Paolo Provero
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
- Center for Omics Sciences, IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
22
|
Foco L, De Bortoli M, Fabiola Del Greco M, Frommelt LS, Volani C, Riekschnitz DA, Motta BM, Fuchsberger C, Delerue T, Völker U, Huan T, Gögele M, Winkelmann J, Dörr M, Levy D, Waldenberger M, Teumer A, Pramstaller PP, Rossini A, Pattaro C. Genomic and molecular evidence that the lncRNA DSP-AS1 modulates Desmoplakin expression. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.29.25324867. [PMID: 40236443 PMCID: PMC11998815 DOI: 10.1101/2025.03.29.25324867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Cardiac desmosomes are specialized cell junctions responsible for cardiomyocytes mechanical coupling. Mutation in desmosomal genes cause autosomal dominant and recessive familial arrhythmogenic cardiomyopathy. Motivated by evidence that Mendelian diseases share genetic architecture with common complex traits, we assessed whether common variants in any desmosomal gene were associated with cardiac conduction traits in the general population. We analysed data of N=4342 Cooperative Health Research in South Tyrol (CHRIS) study participants. We tested associations between genotype imputed variants covering the five desmosomal genes DSP, JUP, PKP2, DSG2, and DSC2, and P-wave, PR, QRS, and QT electrocardiographic intervals, using linear mixed models. Functional annotation and interrogation of publicly available genome-wide association study resources implicated potential connection with antisense lncRNAs, DNA methylation sites, and complex traits. Causality was tested via two-sample Mendelian randomization (MR) analysis and validated with functional in vitro follow-up in human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs). DSP variant rs2744389 was associated with QRS (P=3.5×10-6), with replication in the Microisolates in South Tyrol (MICROS) study (n=636; P=0.010). Observing that rs2744389 was associated with DSP-AS1 antisense lncRNA but not with DSP expression in multiple GTEx-v8 tissues, we conducted two-sample Mendelian randomization analyses that identified causal effects of DSP-AS1 on DSP expression (P=6.33×10-5; colocalization posterior probability=0.91) and QRS (P=0.015). In hiPSC-CMs, DSP-AS1 expression downregulation through a specific GapmerR matching sequence led to significant DSP upregulation at both mRNA and protein levels. The evidence that DSP-AS1 has a regulatory role on DSP opens the venue for further investigations on DSP-AS1's therapeutic potential for conditions caused by reduced desmoplakin production.
Collapse
Affiliation(s)
- Luisa Foco
- Institute for Biomedicine, Eurac Research, Bolzano, Italy
| | | | | | - Laura S. Frommelt
- Institute for Biomedicine, Eurac Research, Bolzano, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cardiovascular Biology Laboratory, Trieste, Italy
| | - Chiara Volani
- Institute for Biomedicine, Eurac Research, Bolzano, Italy
- Università degli Studi di Milano, The Cell Physiology MiLab, Department of Biosciences, Milano, Italy
| | | | | | | | - Thomas Delerue
- Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Munich, 85764 Neuherberg, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Germany
| | - Tianxiao Huan
- Framingham Heart Study, Framingham, MA, United States of America
- The Population Studies Branch, National Heart, Lung, and Blood Institute of the National Institutes of Health, Bethesda, MD, United States of America
| | - Martin Gögele
- Institute for Biomedicine, Eurac Research, Bolzano, Italy
| | - Juliane Winkelmann
- Institute of Neurogenomics, Helmholtz Munich, Helmholtz Munich, 85764 Neuherberg, Germany
| | - Marcus Dörr
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Germany
| | - Daniel Levy
- Framingham Heart Study, Framingham, MA, United States of America
- The Population Studies Branch, National Heart, Lung, and Blood Institute of the National Institutes of Health, Bethesda, MD, United States of America
| | - Melanie Waldenberger
- Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Munich, 85764 Neuherberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 81377 Munich, Germany
| | - Alexander Teumer
- Department SHIP/Clinical-Epidemiological Research, Institute for Community Medicine, University Medicine Greifswald, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Germany
| | | | | | | |
Collapse
|
23
|
Khan AH, Bagley JR, LaPierre N, Gonzalez-Figueroa C, Spencer TC, Choudhury M, Xiao X, Eskin E, Jentsch JD, Smith DJ. Differing genetics of saline and cocaine self-administration in the hybrid mouse diversity panel. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.04.626933. [PMID: 39713377 PMCID: PMC11661131 DOI: 10.1101/2024.12.04.626933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
To identify genes that regulate the response to the potentially addictive drug cocaine, we performed a control experiment using genome-wide association studies (GWASs) and RNA-Seq of a panel of inbred and recombinant inbred mice undergoing intravenous self-administration of saline. A linear mixed model increased statistical power for analysis of the longitudinal behavioral data, which was acquired over 10 days. A total of 145 loci were identified for saline compared to 17 for the corresponding cocaine GWAS. Only one locus overlapped. Transcriptome-wide association studies (TWASs) using RNA-Seq data from the nucleus accumbens and medial frontal cortex identified 5031434O11Rik and Zfp60 as significant for saline self-administration. Two other genes, Myh4 and Npc1, were nominated based on proximity to loci for multiple endpoints or a cis locus regulating expression. All four genes have previously been implicated in locomotor activity, despite the absence of a strong relationship between saline taking and distance traveled in the open field. Our results indicate a distinct genetic basis for saline and cocaine self-administration, and suggest some common genes for saline self-administration and locomotor activity.
Collapse
Affiliation(s)
- Arshad H Khan
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095
- Current address: Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048
| | - Jared R Bagley
- Department of Psychology, Binghamton University, Binghamton, NY 13902
- Current address: Department of Pharmaceutical Sciences, Binghamton University, Binghamton, NY 13902
| | - Nathan LaPierre
- Department of Computer Science, UCLA, Los Angeles, CA 90095
- Current address: Department of Human Genetics, University of Chicago, Chicago, IL 60637
| | | | - Tadeo C Spencer
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095
| | - Mudra Choudhury
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095
- Current address: Sanford Burnham Prebys, La Jolla, CA 92037
| | - Xinshu Xiao
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA 90095
| | - Eleazar Eskin
- Department of Computational Medicine, UCLA, Los Angeles, CA 90095
| | - James D Jentsch
- Department of Psychology, Binghamton University, Binghamton, NY 13902
| | - Desmond J Smith
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095
- Department of Molecular and Medical Pharmacology David Geffen School of Medicine, UCLA Box 951735, 23-151A CHS Los Angeles, CA 90095-1735
| |
Collapse
|
24
|
Wu B, Woo JS, Hasiakos S, Pan C, Cokus S, Benincá C, Stiles L, Sun Z, Pellegrini M, Shirihai OS, Lusis AJ, Srikanth S, Gwack Y. Mitochondrial reactive oxygen species regulate acetyl-CoA flux between cytokine production and fatty acid synthesis in effector T cells. Cell Rep 2025; 44:115430. [PMID: 40088449 PMCID: PMC12007815 DOI: 10.1016/j.celrep.2025.115430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 01/13/2025] [Accepted: 02/21/2025] [Indexed: 03/17/2025] Open
Abstract
Genetic and environmental factors shape an individual's susceptibility to autoimmunity. To identify genetic variations regulating effector T cell functions, we used a forward genetics screen of inbred mouse strains and uncovered genomic loci linked to cytokine expression. Among the candidate genes, we characterized a mitochondrial inner membrane protein, TMEM11, as an important determinant of Th1 responses. Loss of TMEM11 selectively impairs Th1 cell functions, reducing autoimmune symptoms in mice. Mechanistically, Tmem11-/- Th1 cells exhibit altered cristae architecture, impaired respiration, and increased mitochondrial reactive oxygen species (mtROS) production. Elevated mtROS hindered histone acetylation while promoting neutral lipid accumulation. Further experiments using genetic, biochemical, and pharmacological tools revealed that mtROS regulate acetyl-CoA flux between histone acetylation and fatty acid synthesis. Our findings highlight the role of mitochondrial cristae integrity in directing metabolic pathways that influence chromatin modifications and lipid biosynthesis in Th1 cells, providing new insights into immune cell metabolism.
Collapse
Affiliation(s)
- Beibei Wu
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jin Seok Woo
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Spyridon Hasiakos
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Division of Oral Biology and Medicine, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Calvin Pan
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shawn Cokus
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Cristiane Benincá
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Linsey Stiles
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zuoming Sun
- Department of Immunology & Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Orian S Shirihai
- Department of Medicine, Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Metabolism Theme, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA; Molecular Cellular Integrative Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Aldon J Lusis
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sonal Srikanth
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
25
|
Baranova A, Liu D, Chandhoke V, Cao H, Zhang F. Unraveling the genetic links between depression and type 2 diabetes. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111258. [PMID: 39837361 DOI: 10.1016/j.pnpbp.2025.111258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/06/2025] [Accepted: 01/13/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND Type 2 diabetes (T2D) is a chronic metabolic disorder that has high comorbidity with mental disorders. The genetic relationships between T2D and depression are far from being well understood. METHODS We performed genetic correlation, polygenic overlap, Mendelian randomization (MR) analyses, cross-trait meta-analysis, and Bayesian colocalization analysis to assess genetic relationships between T2D and depression, in the forms of major depressive disorder (MDD) and depressed affect (DAF). Then, the summary data-based MR (SMR) analysis was performed to prioritize genes contributing to MDD and to T2D from functional perspective. MDD-driven signaling pathways were constructed to understand the influence of MDD on T2D at the molecular level. RESULTS T2D has positive genetic correlations both with MDD (rg = 0.14) and with DAF (rg = 0.19). The polygenic overlap analysis showed that about 60 % of causal variants for T2D are shared with MDD and DAF. The MR analysis indicated that genetic liabilities to both MDD (OR: 1.24, 95 % CI: 1.11-1.38) and DAF (OR: 1.48, 95 % CI: 1.23-1.78) are associated with an increased risk for T2D, while genetic liability to T2D is not associated with the risk for MDD (OR: 1.00, 95 % CI: 0.99-1.01) or DAF (OR: 1.01, 95 % CI: 1.00-1.02). The cross-trait meta-analysis identified 271 genomic loci, of which 29 were novel. Genetic predisposition to MDD and T2D shares six overlapping loci, involving some well-characterized genes, such as TCF4 and NEGR1. Colocalization analysis revealed three shared chromosome regions between MDD and T2D, which covers mediator genes including SCYL1, DENND1A, and MAD1L1. Molecular pathway analysis suggests mechanisms that promote the development of T2D through inflammatory pathways overactive in patients with MDD. The SMR analysis and the meta-analysis highlighted seven genes with functional implications for both MDD and T2D, including TNKS2, CCDC92, FADS1, ERI1, THUMPD3, NUCKS1, and PM20D1. CONCLUSIONS Our study points out that depression, in the forms of MDD and DAF, may increase the risk of T2D. Analysis of underlying genetic variation and the molecular pathways, connecting depression and T2D, indicate that the pathophysiological foundations of these two conditions have a notable overlap.
Collapse
Affiliation(s)
- Ancha Baranova
- School of Systems Biology, George Mason University, Fairfax 22030, USA; Research Centre for Medical Genetics, Moscow 115478, Russia
| | - Dongming Liu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China; Department of Neurosurgery, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Vikas Chandhoke
- School of Systems Biology, George Mason University, Fairfax 22030, USA
| | - Hongbao Cao
- School of Systems Biology, George Mason University, Fairfax 22030, USA
| | - Fuquan Zhang
- Institute of Neuropsychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China; Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
26
|
Yu A, Yoon D, An HB, Choi U, Heo HR, Chae DH, Park HS, Park JH, Han MH, Heo J, Cho K, Park KS, Yoo HM, Kim HS, Kang KS, Oh MK, Lim HJ, Yu KR. The regulatory role of ACP5 in the diesel exhaust particle-induced AHR inflammatory signaling pathway in a human bronchial epithelial cell line. Sci Rep 2025; 15:8826. [PMID: 40087344 PMCID: PMC11909186 DOI: 10.1038/s41598-024-84280-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 12/23/2024] [Indexed: 03/17/2025] Open
Abstract
Exposure to diesel exhaust particles (DEPs), which are major constituents of urban air pollution, is associated with adverse health outcomes. Previous studies have shown that DEPs enhance the expression of pro-inflammatory cytokines and immune responses. However, few studies have focused on genomic variants that regulate DEP-induced signaling. Here, we identify a frequently found genomic variant, ACP5, in allergic diseases, and establish an ACP5 knock-out (KO) human bronchial epithelial cell line (BEAS-2B) using CRISPR/Cas9 editing to mimic the ACP5 mutation. DEP-induced apoptosis and intracellular reactive oxygen species (ROS) were significantly increased in the ACP5 KO cells compared with controls, suggesting that ACP5 KO cells were at increased risk from DEP exposure. A gene expression profile revealed an activated aryl hydrocarbon receptor (AHR)-CYP1A1 axis followed by upregulated pro-inflammatory signaling. Treatment of a DEP-exposed ACP5 KO BEAS-2B conditioned medium (CM) supernatant induced an inflammatory response and tissue damage in mice, and AHR inhibition effectively prevented inflammation-induced damage, suggesting that AHR-CYP1A1-inflammatory signaling is a prominent mechanism responsible for detrimental effects. Collectively, our findings reveal a novel link between ACP5 KO and the AHR-CYP1A1 inflammatory signaling pathway in DEP-exposed cells, and identify the AHR-CYP1A1 axis as a potential therapeutic target in individuals suffering from DEP-induced toxicity, particularly those with ACP5 mutations.
Collapse
Affiliation(s)
- Aaron Yu
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea
| | - Dankyu Yoon
- Division of Cardiovascular Disease Research, Department of Chronic Disease Convergence, Korea National Institute of Health, Cheongju, 28159, Korea
| | - Hye Bin An
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea
| | - Uimook Choi
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, North Bethesda, USA
| | - Hye-Ryeon Heo
- Division of Cardiovascular Disease Research, Department of Chronic Disease Convergence, Korea National Institute of Health, Cheongju, 28159, Korea
| | - Dong-Hoon Chae
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea
| | - Hyun Sung Park
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea
| | - Jae Han Park
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea
| | - Myoung Hee Han
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea
| | - Jiyoung Heo
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea
| | - Keonwoo Cho
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea
| | - Ki-Sun Park
- KM Science Research Division, Korea Institute of Oriental Medicine, Daejeon, 34054, Korea
| | - Hee Min Yoo
- Biometrology Group, Korea Research Institute of Standards and Science (KRISS), Daejeon, 34113, Korea
- Department of Bio-Analytical Science, University of Science and Technology (UST), Daejeon, 34113, Korea
| | - Hyung-Sik Kim
- Department of Life Science in Dentistry, School of Dentistry, Pusan National University, Yangsan, 50612, Korea
- Dental and Life Science Institute, Pusan National University, Yangsan, 50612, Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center and Research, Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Korea
| | - Mi-Kyung Oh
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea
| | - Hyun Joung Lim
- Division of Cardiovascular Disease Research, Department of Chronic Disease Convergence, Korea National Institute of Health, Cheongju, 28159, Korea.
| | - Kyung-Rok Yu
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea.
| |
Collapse
|
27
|
Yang G, González P, Moneró M, Carrasquillo K, Renta JY, Hernandez-Suarez DF, Botton MR, Melin K, Scott SA, Ruaño G, Roche-Lima A, Alarcon C, Ritchie MD, Perera MA, Duconge J. Discovery of ancestry-specific variants associated with clopidogrel response among Caribbean Hispanics. NPJ Genom Med 2025; 10:20. [PMID: 40055373 PMCID: PMC11889249 DOI: 10.1038/s41525-025-00479-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
High on-treatment platelet reactivity (HTPR) with clopidogrel predicts ischemic events in adults with coronary artery disease, and while HTPR varies by ethnicity, no genome-wide association study (GWAS) of clopidogrel response has been conducted in Caribbean Hispanics. This study aimed to identify genetic predictors of HTPR in a cohort of 511 Puerto Rican cardiovascular patients treated with clopidogrel, stratified by P2Y12 reaction units (PRU) into responders and non-responders (HTPR). Local ancestry inference (LAI) and traditional GWAS identified variants in the CYP2C19 region associated with HTPR, primarily in individuals with European ancestry. Three variants (OSBPL10 rs1376606, DERL3 rs5030613, RGS6 rs9323567) showed suggestive significance, and a variant in UNC5C was linked to increased HTPR risk. These findings highlight the unique genetic landscape of Caribbean Hispanics and challenge the significance of CYP2C19*2 in predicting clopidogrel response in patients with high non-European ancestry. Further studies are needed to replicate these results in other diverse cohorts.
Collapse
Affiliation(s)
- Guang Yang
- Department of Pharmacology, Center for Pharmacogenomics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Pablo González
- Department of Pharmacology, School of Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA
| | - Mariangeli Moneró
- Department of Pharmacology, School of Medicine, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA
| | - Kelvin Carrasquillo
- Research Centers in Minority Institutions (RCMI) Program, Center for Collaborative Research in Health Disparities (CCRHD), Academic Affairs Deanship, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA
| | - Jessicca Y Renta
- Research Centers in Minority Institutions (RCMI) Program, Center for Collaborative Research in Health Disparities (CCRHD), Academic Affairs Deanship, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA
| | - Dagmar F Hernandez-Suarez
- Department of Translational Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Mariana R Botton
- Transplant Immunology and Personalized Medicine Unit, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Kyle Melin
- Department of Pharmacy Practice, School of Pharmacy, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA
| | - Stuart A Scott
- Department of Pathology, Stanford University, Palo Alto, CA, USA
| | | | - Abiel Roche-Lima
- Research Centers in Minority Institutions (RCMI) Program, Center for Collaborative Research in Health Disparities (CCRHD), Academic Affairs Deanship, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA
| | - Cristina Alarcon
- Department of Pharmacology, Center for Pharmacogenomics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Marylyn D Ritchie
- Department of Genetics and Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Minoli A Perera
- Department of Pharmacology, Center for Pharmacogenomics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jorge Duconge
- Research Centers in Minority Institutions (RCMI) Program, Center for Collaborative Research in Health Disparities (CCRHD), Academic Affairs Deanship, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Puerto Rico-Medical Sciences Campus, San Juan, PR, USA.
| |
Collapse
|
28
|
Jiang Y, Zhang H. Empowering genome-wide association studies via a visualizable test based on the regional association score. Proc Natl Acad Sci U S A 2025; 122:e2419721122. [PMID: 39999171 PMCID: PMC11892588 DOI: 10.1073/pnas.2419721122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
The genome-wide association studies identified genes associated with many diseases, but the identification and verification of disease variants are still challenging due to small effects and large number of individual variants. In this paper, we propose a powerful method that first quantifies the strength of regional associations at each single nucleotide polymorphism and converts these measures into time series data before using a change point detection algorithm to identify significant regions. In our extensive simulation study, the proposed method consistently demonstrates greater power than existing alternatives, achieving a relative increase of over 20% in challenging scenarios where true causal variants are sparse and multiple association regions exist at the same time, while maintaining a lower false positive rate.
Collapse
Affiliation(s)
- Yiran Jiang
- Department of Biostatistics, Yale University, New Haven, CT06511
| | - Heping Zhang
- Department of Biostatistics, Yale University, New Haven, CT06511
| |
Collapse
|
29
|
Ray NR, Bradley J, Yilmaz E, Kizil C, Kurup JT, Martin ER, Klein HU, Kunkle BW, Bennett DA, De Jager PL, Alzheimer’s Disease Genetics Consortium, Beecham GW, Cruchaga C, Reitz C. Local genetic covariance analysis with lipid traits identifies novel loci for early-onset Alzheimer's Disease. PLoS Genet 2025; 21:e1011631. [PMID: 40096060 PMCID: PMC11984970 DOI: 10.1371/journal.pgen.1011631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 04/10/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025] Open
Abstract
The genetic component of early-onset Alzheimer disease (EOAD), accounting for ~10% of all Alzheimer's disease (AD) cases, is largely unexplained. Recent studies suggest that EOAD may be enriched for variants acting in the lipid pathway. The current study examines the shared genetic heritability between EOAD and the lipid pathway using genome-wide multi-trait genetic covariance analyses. Summary statistics were obtained from the GWAS meta-analyses of EOAD by the Alzheimer's Disease Genetics Consortium (n=19,668) and five blood lipid traits by the Global Lipids Genetics Consortium (n=1,320,016). The significant results were compared between the EOAD and lipids GWAS and genetic covariance analyses were performed via SUPERGNOVA. Genes in linkage disequilibrium (LD) with top EOAD hits in identified regions of covariance with lipid traits were scored and ranked for causality by combining evidence from gene-based analysis, AD-risk scores incorporating transcriptomic and proteomic evidence, eQTL data, eQTL colocalization analyses, DNA methylation data, and single-cell RNA sequencing analyses. Direct comparison of GWAS results showed 5 loci overlapping between EOAD and at least one lipid trait harboring APOE, TREM2, MS4A4E, LILRA5, and LRRC25. Local genetic covariance analyses identified 3 regions of covariance between EOAD and at least one lipid trait. Gene prioritization nominated 3 likely causative genes at these loci: ANKDD1B, CUZD1, and MS4A64.The current study identified genetic covariance between EOAD and lipids, providing further evidence of shared genetic architecture and mechanistic pathways between the two traits.
Collapse
Affiliation(s)
- Nicholas R. Ray
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Joseph Bradley
- NeuroGenomics and Informatics Center, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Elanur Yilmaz
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Caghan Kizil
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Jiji T. Kurup
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Eden R. Martin
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Hans-Ulrich Klein
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | - Brian W. Kunkle
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - David A. Bennett
- RUSH Alzheimer’s Disease Center, RUSH University, Chicago, Illinois, United States of America
| | - Philip L. De Jager
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| | | | - Gary W. Beecham
- The John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Carlos Cruchaga
- NeuroGenomics and Informatics Center, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Christiane Reitz
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
| |
Collapse
|
30
|
Wonkam A, Esoh K, Levine RM, Ngo Bitoungui VJ, Mnika K, Nimmagadda N, Dempsey EAD, Nkya S, Sangeda RZ, Nembaware V, Morrice J, Osman F, Beer MA, Makani J, Mulder N, Lettre G, Steinberg MH, Latanich R, Casella JF, Drehmer D, Arking DE, Chimusa ER, Yen JS, Newby GA, Antonarakis SE. FLT1 and other candidate fetal haemoglobin modifying loci in sickle cell disease in African ancestries. Nat Commun 2025; 16:2092. [PMID: 40025045 PMCID: PMC11873275 DOI: 10.1038/s41467-025-57413-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/20/2025] [Indexed: 03/04/2025] Open
Abstract
Known fetal haemoglobin (HbF)-modulating loci explain 10-24% variation of HbF level in Africans with Sickle Cell Disease (SCD), compared to 50% among Europeans. Here, we report fourteen candidate loci from a genome-wide association study (GWAS) of HbF level in patients with SCD from Cameroon, Tanzania, and the United States of America. We present results of cell-based experiments for FLT1 candidate, demonstrating expression in early haematopoiesis and a possible involvement in hypoxia associated HbF induction. Our study employed genotyping arrays that capture a broad range of African and non-African genetic variation and replicated known loci (BCL11A and HBS1L-MYB). We estimated the heritability of HbF level in SCD at 94%, higher than estimated in unselected Europeans, and suggesting a robust capture of HbF-associated loci by these arrays. Our approach, which involved genotype imputation against six reference haplotype panels and association analysis with each of the panels, proved superior over selecting a best-performing panel, evidenced by a substantial proportion of panel-specific (up to 18%) and a low proportion of shared (28%) imputed variants across the panels.
Collapse
Affiliation(s)
- Ambroise Wonkam
- McKusick-Nathans Institute and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| | - Kevin Esoh
- McKusick-Nathans Institute and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Rachel M Levine
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Khuthala Mnika
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Nikitha Nimmagadda
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Erin A D Dempsey
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Siana Nkya
- Department of Biochemistry and Molecular Biology, Muhimbili University of Health and Allied Sciences, Dar Es Salaam, Tanzania
| | - Raphael Z Sangeda
- Department of Pharmaceutical Microbiology, Muhimbili University of Health and Allied Sciences, Dar Es Salaam, Tanzania
| | - Victoria Nembaware
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Jack Morrice
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Fujr Osman
- McKusick-Nathans Institute and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael A Beer
- McKusick-Nathans Institute and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Julie Makani
- Sickle Cell Programme, Department of Haematology and Blood Transfusion, Muhimbili University of Health & Allied Sciences (MUHAS), Dar Es Salaam, Tanzania
- SickleInAfrica Clinical Coordinating Center, Muhimbili University of Health & Allied Sciences (MUHAS), Dar Es Salaam, Tanzania
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Nicola Mulder
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, CIDRI-Africa Wellcome Trust Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Guillaume Lettre
- Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
| | - Martin H Steinberg
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Rachel Latanich
- McKusick-Nathans Institute and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - James F Casella
- Department of Pediatrics, Division of Hematology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daiana Drehmer
- Armstrong Oxygen Biology Research Center, Institute for Cell Engineering, and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dan E Arking
- McKusick-Nathans Institute and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Emile R Chimusa
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle, Tyne and Wear, UK
| | - Jonathan S Yen
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Gregory A Newby
- McKusick-Nathans Institute and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Stylianos E Antonarakis
- Department of Genetic Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
31
|
Kim J, Lee HJ, Park HJ, Lee JH, Kim WJ. Genome-Wide Association Study Identifying a Novel Gene Related to a History of Febrile Convulsions in Patients With Focal Epilepsy. J Clin Neurol 2025; 21:123-130. [PMID: 40065453 PMCID: PMC11896740 DOI: 10.3988/jcn.2024.0296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/07/2024] [Accepted: 12/11/2024] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND AND PURPOSE The risk factors for developing epilepsy following febrile convulsion (FC) have been studied extensively, but the underlying genetic components remain largely unexplored. Our objective here was to identify the risk loci related to FC through a genome-wide association study of Korean epilepsy patients. METHODS We examined associations between a history of FC and single-nucleotide polymorphisms (SNPs) in data obtained from 125 patients with focal epilepsy: 28 with an FC history and 97 without an FC history. RESULTS Among 288,394 SNPs, 5 candidate SNPs showed p<1×10⁻⁴. Regional association plots of these SNPs identified a novel locus adjacent to PROX1 that is implicated in hippocampal neurogenesis and epileptogenesis. The allele frequencies of the SNPs upstream of PROX1 including two candidate SNPs (rs1159179 and rs7554295 on chromosome 1) differed significantly between the groups with and without an FC history. In contrast, the allele frequencies of the SNPs inside PROX1 showed no differences, indicating dysregulated expression of PROX1 rather than a functional alteration in the PROX1 protein. CONCLUSIONS This novel discovery of SNPs upstream of PROX1 suggests that the dysregulated expression of PROX1 contributes to the development of focal epilepsy following FC. We propose that these SNPs are potential genetic markers for focal epilepsy following FC, and that PROX1 represents a potential therapeutic target of antiseizure medications.
Collapse
Affiliation(s)
- Joonho Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Jeong Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
- Department of Neurology, Chung-Ang University Gwangmyeong Hospital, Gwangmyeong, Korea
| | - Hyung Jun Park
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Ji Hyun Lee
- Department of Clinical Pharmacology and Therapeutics, Kyung Hee University College of Medicine, Seoul, Korea
| | - Won-Joo Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
32
|
Park K, Shin CM, Kim N, Won S, Song CH, Ohn JH, Lee S, Park JH, Yie GE, Kang SJ, Kim JS, Lee DH. rs762855 single nucleotide polymorphism modulates the risk for diffuse-type gastric cancer in females: a genome-wide association study in the Korean population. Gastric Cancer 2025; 28:145-159. [PMID: 39862296 PMCID: PMC11842433 DOI: 10.1007/s10120-024-01575-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 12/08/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Intestinal-type gastric cancer (IGC) and diffuse-type gastric cancer (DGC) exhibit different prevalence rates between sexes. While environmental factors like Helicobacter pylori infection and alcohol consumption contribute to these differences, they do not fully account for them, suggesting a role for host genetic factors. METHODS We conducted a meta-analysis to explore associations between single nucleotide polymorphisms (SNPs) and the risk of IGC or DGC. The analysis included the SNUBH cohort (998 participants: 159 DGCs, 303 IGCs, 4,962,361 variants) and the GC_HC cohort (6,233 participants: 389 DGCs, 405 IGCs, 4,541,617 variants). Significant variants were validated in the SNUBH2_AA cohort (5,511 participants: 40 DGCs, 49 IGCs, 3,668,632 variants). RESULTS The meta-analysis identified that rs762855 (chr4:3,074,795; hg19) is significantly associated with DGC risk in females (OR [95% CI]: 1.758 [1.438-2.150], P = 3.91 × 10-8), a finding replicated in the SNUBH2_AA datasets (OR [95% CI]: 3.356 [1.031-10.92], P = 4.43 × 10-2). Gene-set and transcriptomic analyses revealed that the Myb/SANT DNA Binding Domain Containing 1 (MSANTD1) gene is significantly linked to DGC susceptibility in females. In addition, Mendelian randomization analyses suggested that increased serum total protein and non-albumin protein (NAP) levels elevate DGC risk in females (P < 0.05), but not in males. CONCLUSION The rs762855 SNP, MSANTD1, and serum NAP levels are associated with DGC risk in Korean females.
Collapse
Affiliation(s)
- Kyungtaek Park
- Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
| | - Cheol Min Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-Do, South Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-Do, South Korea.
- Research Center for Sex- and Gender-Specific Medicine, Seoul National University Bundang Hospital, 173-82, Gumi-ro, Bundang-gu, Seongnam, Gyeonggi-Do, South Korea.
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea.
| | - Sungho Won
- Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
- Department of Public Health Sciences, Seoul National University, Seoul, South Korea
- Interdisciplinary Program of Bioinformatics, Seoul National University, Seoul, Korea
| | - Chin-Hee Song
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-Do, South Korea
| | - Jung Hun Ohn
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-Do, South Korea
| | - Sejoon Lee
- Precision Medicine Center, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Ji Hyun Park
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Ga-Eun Yie
- Interdisciplinary Program of Bioinformatics, Seoul National University, Seoul, Korea
| | - Seung Joo Kang
- Department of Internal Medicine and Healthcare Research Institute, Healthcare System Gangnam Center, Seoul National University Hospital, Seoul, Korea
| | - Joo Sung Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
- Department of Internal Medicine and Healthcare Research Institute, Healthcare System Gangnam Center, Seoul National University Hospital, Seoul, Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-Do, South Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
33
|
Ismail Umlai UK, Toor SM, Al-Sarraj YA, Mohammed S, Al Hail MSH, Ullah E, Kunji K, El-Menyar A, Gomaa M, Jayyousi A, Saad M, Qureshi N, Al Suwaidi JM, Albagha OME. A multi-ancestry genome-wide association study and evaluation of polygenic scores of LDL-C levels. J Lipid Res 2025; 66:100752. [PMID: 39909172 PMCID: PMC11927683 DOI: 10.1016/j.jlr.2025.100752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/12/2024] [Accepted: 02/02/2025] [Indexed: 02/07/2025] Open
Abstract
The genetic determinants of low-density lipoprotein cholesterol (LDL-C) levels in blood have been predominantly explored in European populations and remain poorly understood in Middle Eastern populations. We investigated the genetic architecture of LDL-C variation in Qatar by conducting a genome-wide association study (GWAS) on serum LDL-C levels using whole genome sequencing data of 13,701 individuals (discovery; n = 5,939, replication; n = 7,762) from the population-based Qatar Biobank (QBB) cohort. We replicated 168 previously reported loci from the largest LDL-C GWAS by the Global Lipids Genetics Consortium (GLGC), with high correlation in allele frequencies (R2 = 0.77) and moderate correlation in effect sizes (Beta; R2 = 0.53). We also performed a multi-ancestry meta-analysis with the GLGC study using MR-MEGA (Meta-Regression of Multi-Ethnic Genetic Association) and identified one novel LDL-C-associated locus; rs10939663 (SLC2A9; genomic control-corrected P = 1.25 × 10-8). Lastly, we developed Qatari-specific polygenic score (PGS) panels and tested their performance against PGS derived from other ancestries. The multi-ancestry-derived PGS (PGS000888) performed best at predicting LDL-C levels, whilst the Qatari-derived PGS showed comparable performance. Overall, we report a novel gene associated with LDL-C levels, which may be explored further to decipher its potential role in the etiopathogenesis of cardiovascular diseases. Our findings also highlight the importance of population-based genetics in developing PGS for clinical utilization.
Collapse
Affiliation(s)
- Umm-Kulthum Ismail Umlai
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Salman M Toor
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Yasser A Al-Sarraj
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar; Qatar Genome Program (QGP), Qatar Foundation Research, Development and Innovation, Qatar Foundation, Doha, Qatar
| | - Shaban Mohammed
- Department of Pharmacy, Hamad Medical Corporation, Doha, Qatar
| | | | - Ehsan Ullah
- Qatar Computing Research Institute (QCRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Khalid Kunji
- Qatar Computing Research Institute (QCRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Ayman El-Menyar
- Trauma and Vascular Surgery, Hamad Medical Corporation (HMC), Doha, Qatar
| | - Mohammed Gomaa
- Adult Cardiology, Heart Hospital, Hamad Medical Corporation (HMC), Doha, Qatar
| | - Amin Jayyousi
- Department of Diabetes, Hamad Medical Corporation (HMC), Doha, Qatar
| | - Mohamad Saad
- Qatar Computing Research Institute (QCRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Nadeem Qureshi
- Primary Care Stratified Medicine Research Group, Centre for Academic Primary Care, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Jassim M Al Suwaidi
- Adult Cardiology, Heart Hospital, Hamad Medical Corporation (HMC), Doha, Qatar
| | - Omar M E Albagha
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar; Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
34
|
Park J, Peña-Tauber A, Talozzi L, Greicius MD, Le Guen Y. Rare genetic associations with human lifespan in UK Biobank are enriched for oncogenic genes. Nat Commun 2025; 16:2064. [PMID: 40021682 PMCID: PMC11871019 DOI: 10.1038/s41467-025-57315-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 02/18/2025] [Indexed: 03/03/2025] Open
Abstract
Human lifespan is shaped by genetic and environmental factors. To enable precision health, understanding how genetic variants influence mortality is essential. We conducted a survival analysis in European ancestry participants of the UK Biobank, using age-at-death (N=35,551) and last-known-age (N=358,282). The associations identified were predominantly driven by cancer. We found lifespan-associated loci (APOE, ZSCAN23) for common variants and six genes where burden of loss-of-function variants were linked to reduced lifespan (TET2, ATM, BRCA2, CKMT1B, BRCA1, ASXL1). Additionally, eight genes with pathogenic missense variants were associated with reduced lifespan (DNMT3A, SF3B1, TET2, PTEN, SOX21, TP53, SRSF2, RLIM). Many of these genes are involved in oncogenic pathways and clonal hematopoiesis. Our findings highlight the importance of understanding genetic factors driving the most prevalent causes of mortality at a population level, highlighting the potential of early genetic testing to identify germline and somatic variants increasing one's susceptibility to cancer and/or early death.
Collapse
Affiliation(s)
- Junyoung Park
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA.
| | - Andrés Peña-Tauber
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Lia Talozzi
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Yann Le Guen
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA, 94304, USA
| |
Collapse
|
35
|
Fricke DR, Cvejkus RK, Barinas-Mitchell E, Feitosa MF, Murabito JM, Acharya S, Brent MR, Daw EW, Minster RL, Zmuda JM, Kuipers AL. Epidemiology and genetic determination of measures of peripheral vascular health in the Long Life Family Study. Aging (Albany NY) 2025; 17:464-481. [PMID: 40013929 PMCID: PMC11892930 DOI: 10.18632/aging.206204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 02/03/2025] [Indexed: 02/28/2025]
Abstract
Peripheral artery disease (PAD) is a major contributor to morbidity in older adults. We aimed to determine genetic and non-genetic determinants of PAD and ankle-brachial index (ABI) in the Long Life Family Study (LLFS). 3006 individuals had ABI assessment, including 1090 probands (mean age 89), 1554 offspring (mean age 60) and 362 spousal controls (mean age 61). Outcomes include minimum of right and left ABIs and PAD (ABI <0.9). Stepwise regression determined independent significant non-genetic correlates of ABI and PAD. Genomewide association and linkage analyses were adjusted for age, sex, study center, significant principal components, and independent predictors. All analyses accounted for familial relatedness. Median ABI was 1.16 and 7.4% had PAD (18.2% probands, 1.0% offspring, 1.9% controls). Correlates of PAD and lower ABI included age, SBP, and creatinine (ABI only); BMI (ABI only), HDL (ABI only) and DBP (PAD only); and antihypertensive use, current smoking, female sex (ABI only), and high school noncompletion (ABI only). Genomewide linkage identified 1 region (15q12-q13) and association identified 3 single nucleotide polymorphisms (rs780213, rs12512857, rs79644420) of interest. In these families, PAD prevalence was low compared to other studies of older adults. We identified four genomic sites that may harbor variants associated with protection from PAD.
Collapse
Affiliation(s)
- Deidra R. Fricke
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Ryan K. Cvejkus
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | | | - Mary F. Feitosa
- Department of Genetics, Division of Statistical Genomics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joanne M. Murabito
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
- Section of General Internal Medicine, Boston Medical Center, Boston, MA 02119, USA
| | - Sandeep Acharya
- Division of Computational and Data Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael R. Brent
- Department of Computer Science and Engineering, Washington University, St. Louis, MO 63130, USA
| | - E. Warwick Daw
- Department of Genetics, Division of Statistical Genomics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ryan L. Minster
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Joseph M. Zmuda
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Allison L. Kuipers
- Department of Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| |
Collapse
|
36
|
Kumari P, Friedman RZ, Pi L, Curtis SW, Paraiso K, Visel A, Rhea L, Dunnwald M, Patni AP, Mar D, Bomsztyk K, Mathieu J, Ruohola-Baker H, Leslie EJ, White MA, Cohen BA, Cornell RA. Identification of functional non-coding variants associated with orofacial cleft. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.01.596914. [PMID: 40027800 PMCID: PMC11870446 DOI: 10.1101/2024.06.01.596914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Oral facial cleft (OFC) is a multifactorial disorder that can present as a cleft lip with or without cleft palate (CL/P) or a cleft palate only. Genome wide association studies (GWAS) of isolated OFC have identified common single nucleotide polymorphisms (SNPs) at the 1q32/ IRF6 locus and many other loci where, like IRF6 , the presumed OFC-relevant gene is expressed in embryonic oral epithelium. To identify the functional subset of SNPs at eight such loci we conducted a massively parallel reporter assay in a cell line derived from fetal oral epithelium, revealing SNPs with allele-specific effects on enhancer activity. We filtered these against chromatin-mark evidence of enhancers in relevant cell types or tissues, and then tested a subset in traditional reporter assays, yielding six candidates for functional SNPs in five loci (1q32/ IRF6 , 3q28/ TP63 , 6p24.3/ TFAP2A , 20q12/ MAFB , and 9q22.33/ FOXE1 ). We further tested two SNPs near IRF6 and one near FOXE1 by engineering the genome of induced pluripotent stem cells, differentiating the cells into embryonic oral epithelium, and measuring expression of IRF6 or FOXE1 and binding of transcription factors; the results strongly supported their candidacy. Conditional analyses of a meta-analysis of GWAS suggest that the two functional SNPs near IRF6 account for the majority of risk for CL/P associated with variation at this locus. This study connects genetic variation associated with orofacial cleft to mechanisms of pathogenesis.
Collapse
|
37
|
Galan C, Lu G, Gill R, Li D, Liu Y, Huh JR, Hang S. RTF1 mediates epigenetic control of Th17 cell differentiation via H2B monoubiquitination. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:vkae043. [PMID: 40073106 PMCID: PMC11952878 DOI: 10.1093/jimmun/vkae043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/02/2024] [Indexed: 03/14/2025]
Abstract
A gene encoding the transcription factor RTF1 has been associated with an increased risk of ulcerative colitis (UC). In this study, we investigated its function in modulating T cells expressing interleukin-17A (Th17 cells), a cardinal cell type promoting intestinal inflammation. Our results indicate that Rtf1 deficiency disrupts the differentiation of Th17 cells, while leaving regulatory T cells (Treg) unaffected. Mechanistically, RTF1 facilitates histone H2B monoubiquitination (H2Bub1), which requires its histone modification domain (HMD), for supporting Th17 cell function. Impaired Th17 differentiation was also observed in cells lacking the H2Bub1 E3 ligase subunit RNF40, an enzyme known to physically interact with RTF1. Thus, our study underscores the essential role of RTF1 in H2Bub1-mediated epigenetic regulation of Th17 cell differentiation. Understanding this process will likely provide valuable insights into addressing Th17-associated inflammatory disorders. (Images were created with BioRender).
Collapse
Affiliation(s)
| | - Guangqing Lu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, United States
| | - Richard Gill
- Genentech, Inc, South San Francisco, CA, United States
| | - Dun Li
- Genentech, Inc, South San Francisco, CA, United States
| | - Yifang Liu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, United States
| | - Jun R Huh
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, United States
- Bio2Q, Keio University, Tokyo, Japan
| | - Saiyu Hang
- Genentech, Inc, South San Francisco, CA, United States
| |
Collapse
|
38
|
Ohyama N, Matsunami M, Imamura M, Yoshida A, Javed A, Liu X, Kimura R, Matsuda K, Terao C, Maeda S. A variant in HMMR/HMMR-AS1 is associated with serum alanine aminotransferase levels in the Ryukyu population. Sci Rep 2025; 15:6494. [PMID: 39987337 PMCID: PMC11846991 DOI: 10.1038/s41598-025-90195-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/11/2025] [Indexed: 02/24/2025] Open
Abstract
The Ryukyu archipelago is located southwest of the Japanese islands, and people originally from this region, the Ryukyu population, have a unique genetic background distinct from that of other populations, including people from mainland Japan. However, few genetic studies have focused on the Ryukyu population. In this study, we performed genome-wide association studies (GWAS) on the serum levels of alanine aminotransferase (ALT, n = 15,224), aspartate aminotransferase (AST, n = 15,203), and gamma-glutamyl transferase (GGT, n = 14,496) in the Ryukyu population. We found 13 loci with a genome-wide significant association (P < 5 × 10-8), three for ALT, four for AST, and six for GGT, including one novel locus associated with ALT: rs117595134-A in HMMR/HMMR-AS1, ß = - 0.131, standard error = 0.024, P = 4.90 × 10-8. Rs117595134-A is common in the Japanese population but is not observed in other ethnic populations in the 1000 genomes database. Additionally, 77 of 80 loci derived from Korean GWAS and 541 of 716 loci from European GWAS showed the same directions of effect (P = 1.41 × 10-19, P = 2.50 × 10-44, binomial test), indicating that most of susceptibility loci are shared between the Ryukyu population and other ethnic populations.
Collapse
Affiliation(s)
- Noriko Ohyama
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
- Department of Cardiovascular Surgery, Okinawa Prefectural Nanbu Medical Center and Children's Medical Center, Haebaru, Japan
| | - Masatoshi Matsunami
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
| | - Minako Imamura
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan.
- Division of Clinical Laboratory and Blood Transfusion, University of the Ryukyus Hospital, Nishihara, Japan.
| | - Akihiro Yoshida
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
- Department of Obstetrics and Gynecology, Okinawa Hokubu Hospital, Nago, Japan
| | - Azeem Javed
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
| | - Xiaoxi Liu
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan
| | - Ryosuke Kimura
- Department of Human Biology and Anatomy, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
| | - Koichi Matsuda
- Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan
- Department of Applied Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Shiro Maeda
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
- Division of Clinical Laboratory and Blood Transfusion, University of the Ryukyus Hospital, Nishihara, Japan
| |
Collapse
|
39
|
Liu J. PSMB4: a potential biomarker and therapeutic target for depression, perspective from integration analysis of depression GWAS data and human plasma proteome. Transl Psychiatry 2025; 15:62. [PMID: 39979251 PMCID: PMC11842700 DOI: 10.1038/s41398-025-03279-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 01/19/2025] [Accepted: 02/10/2025] [Indexed: 02/22/2025] Open
Abstract
Depression is a common and severe mental disorder that affects more than 300 million people worldwide. While it is known to have a moderate genetic component, identifying specific genes that contribute to the disorder has been challenging. Previous Genome-wide association studies (GWASs) have identified over 100 genomic loci that are significantly associated with depression. But finding useful therapeutic targets and diagnostic biomarkers from this information has proven difficult. To address this challenge, I conducted a plasma protein proteome-wide association study (PWAS) for depression, using human plasma protein QTL (pQTL) and depression GWAS data. I identified four proteins that were significantly associated with depression: BTN3A3 (P value = 6.41 × 10-06), PSMB4 (P value = 1.42 × 10-05), TIMP4 (P value = 3.77 × 10-05), and ITIH1 (P value = 7.86 × 10-05). Specifically, I found that BTN3A3 and PSMB4 play a causal role in depression, as confirmed by colocalization and Mendelian Randomization (MR) analysis. Interestingly, I also discovered that PSMB4 was significantly associated with depression in both the brain proteome studies and the plasma PWAS results, which suggests that it may be a particularly promising candidate for further study. Overall, this work has identified 4 new risk proteins for depression and highlights the potential of plasma proteome data for uncovering novel therapeutic targets and diagnostic biomarkers.
Collapse
Affiliation(s)
- Jiewei Liu
- Department of Psychiatry, Wuhan Mental Health Center, Wuhan, 430012, Hubei Province, China.
- Department of Psychiatry, Wuhan Hospital for Psychotherapy, Wuhan, 430012, Hubei Province, China.
| |
Collapse
|
40
|
Zheng R, Li Z, Fang W, Lou H, Liu F, Sun Q, Shi X, Liu H, Chen Q, Shen X, Yao L, Guan L, Chen J, Xie Y, Yang Y, Yang H, Wang L, Qin L, Huang X, Wang J, Liu Z, Liu H, Ge B, Xu J, Sha W. A genome-wide association study identified PRKCB as a causal gene and therapeutic target for Mycobacterium avium complex disease. Cell Rep Med 2025; 6:101923. [PMID: 39848245 PMCID: PMC11866518 DOI: 10.1016/j.xcrm.2024.101923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/01/2024] [Accepted: 12/26/2024] [Indexed: 01/25/2025]
Abstract
Non-tuberculous mycobacterial pulmonary disease (NTM-PD) is a chronic progressive lung disease that is increasing in incidence. Host genetic factors are associated with NTM-PD susceptibility. However, the heritability of NTM-PD is not well understood. Here, we perform a two-stage genome-wide association study (GWAS) and discover a susceptibility locus at 16p21 associated with NTM-PD, especially with pulmonary Mycobacterium avium complex (MAC) disease. As the lead variant, rs194800 C allele augments protein kinase C beta (PRKCB) gene expression and associates with severer NTM-PD. The functional studies show that PRKCB exacerbates M. avium infection and promotes intracellular survival of M. avium in macrophages by inhibiting phagosomal acidification. Mechanistically, PRKCB interacts with subunit G of the vacuolar-H+-ATPase (V-ATPase) and vacuolar protein sorting-associated protein 16 homolog (VPS16), blocking the fusion between lysosomes and mycobacterial phagosomes. PRKCB inhibitor has therapeutic potential against M. avium infection. These findings provide insights into the genetic etiology of NTM-PD and highlight PRKCB as an attractive target for host-directed therapy of MAC disease.
Collapse
Affiliation(s)
- Ruijuan Zheng
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, P.R. China; Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200049, China.
| | - Zhiqiang Li
- The Affiliated Hospital of Qingdao University &The Biomedical Sciences Institute of Qingdao University (Qingdao Branch of SJTU Bio-X Institutes), Qingdao University, Qingdao 266003, P.R. China; Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education) and the Collaborative Innovation Center for Brain Science, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Weijun Fang
- Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200092, P.R. China
| | - Hai Lou
- Department of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Feng Liu
- Department of Otolaryngology Head and Neck Surgery and Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Road 600, Shanghai 200233, P.R. China
| | - Qin Sun
- Department of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Xiang Shi
- Department of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Hua Liu
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, P.R. China
| | - Qing Chen
- School of Public Health, the Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, P.R. China
| | - Xiaona Shen
- Department of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Lan Yao
- Department of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Liru Guan
- Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200092, P.R. China
| | - Jianxia Chen
- Clinical Translation Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, P.R. China
| | - Yingzhou Xie
- Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai, China
| | - Yifan Yang
- Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200092, P.R. China
| | - Hua Yang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, P.R. China; Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200049, China
| | - Ling Wang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, P.R. China; Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200049, China
| | - Lianhua Qin
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, P.R. China
| | - Xiaochen Huang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, P.R. China
| | - Jie Wang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, P.R. China
| | - Zhonghua Liu
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, P.R. China
| | - Haipeng Liu
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, P.R. China; Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Baoxue Ge
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, P.R. China; Key Laboratory of Pathogen-Host Interaction, Ministry of Education, Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200049, China; Department of Microbiology and Immunology, Tongji University School of Medicine, Shanghai 200092, P.R. China; Clinical Translation Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, P.R. China.
| | - Jinfu Xu
- Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai, China; Department of Respiratory and Critical Care Medicine, Huadong Hospital, Fudan University, Shanghai, China.
| | - Wei Sha
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, P.R. China; Department of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China.
| |
Collapse
|
41
|
Zhang Y, Zhang CY, Yuan J, Jiang H, Sun P, Hui L, Xu L, Yu L, Guo Z, Wang L, Yang Y, Li M, Li SW, Yang J, Li W, Teng Z, Xiao X. Human mood disorder risk gene Synaptotagmin-14 contributes to mania-like behaviors in mice. Mol Psychiatry 2025:10.1038/s41380-025-02933-1. [PMID: 39966626 DOI: 10.1038/s41380-025-02933-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/30/2025] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
Bipolar disorder (BD) and major depressive disorder (MDD) are the most prevalent mood disorders and cause considerable burden worldwide. Compelling evidence suggests a pronounced overlap between these two disorders in clinical symptoms, treatment strategies, and genetic etiology. Here we leverage a BD GWAS (1822 cases and 4650 controls) and a MDD GWAS (5303 cases and 5337 controls), followed by independent replications, to investigate their shared genetic basis among Han Chinese. We have herein identified a lead SNP rs126277 at the 1q32.2 locus, which also exhibited nominal associations with mood disorders and several relevant sub-clinical phenotypes (e.g., mania) in European populations. Bulk tissue and single-cell eQTL analyses suggest that the risk G-allele of rs126277 predicted lower SYT14 mRNA expression in human brains. We generated mice lacking Syt14 (Syt14-/-) and mice with insufficient expression of Syt14 in the hippocampus (Syt14-KD), and found that depletion of Syt14 resulted in mania-like behaviors including hyperactivity and anti-depressive behaviors, resembling aspects of mood disorders. We also confirmed that deficiency of this gene in the hippocampus was sufficient to induce hyperactivity in mice. RNA-sequencing analyses of the hippocampus of Syt14-/- mice revealed significant upregulation of Per1 as well as downregulation of Slc7a11 and Ptprb. Ultrastructural analyses showed significant alteration of the number of vesicles within 50 nm to the active zone and the width of synaptic cleft in the ventral hippocampus of Syt14-/- mice compared with the control mice. Overall, we have identified a novel mood disorder risk gene SYT14, and confirmed its impact on mania-like behaviors. While the current study identifies an essential mood disorder risk gene, further investigations elucidating the detailed mechanisms by which SYT14 contributes to the pathogenesis of the illnesses are needed.
Collapse
Affiliation(s)
- Yue Zhang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Chu-Yi Zhang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jing Yuan
- Department of Psychiatry, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Hongyan Jiang
- Department of Psychiatry, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Ping Sun
- Qingdao Mental Health Center, Qingdao, Shandong, China
| | - Li Hui
- Suzhou Guangji Hospital, The Affiliated Guangji Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Li Xu
- Department of Psychiatry, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Ling Yu
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zeyi Guo
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Lu Wang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yi Yang
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Ming Li
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Shi-Wu Li
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jianzhong Yang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou, Zhejiang, China
| | - Wei Li
- Department of Blood Transfusion, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zhaowei Teng
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan Province, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Xiao Xiao
- State Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
- Yunnan Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China.
| |
Collapse
|
42
|
Meng Z, Zhang C, Liu S, Li W, Wang Y, Zhang Q, Peng B, Ye W, Jiang Y, Song Y, Guo M, Chang X, Shao L. Exploring genetic loci linked to COVID-19 severity and immune response through multi-trait GWAS analyses. Front Genet 2025; 16:1502839. [PMID: 40034745 PMCID: PMC11873281 DOI: 10.3389/fgene.2025.1502839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction COVID-19 severity has been linked to immune factors, with excessive immune responses like cytokine storms contributing to mortality. However, the genetic basis of these immune responses is not well understood. This study aimed to explore the genetic connection between COVID-19 severity and blood cell traits, given their close relationship with immunity. Materials and methods GWAS summary statistics for COVID-19 and blood cell counts were analyzed using Linkage Disequilibrium Score Regression (LDSC) to estimate genetic correlations and heritabilities. For traits with significant correlations, a Multi-Trait GWAS Analysis (MTAG) was performed to identify pleiotropic loci shared between COVID-19 and blood cell counts. Results Our MTAG analysis identified four pleiotropic loci associated with COVID-19 severity, five loci linked to hospitalized cases, and one locus related to general patients. Among these, two novel loci were identified in the high-risk population, with rs55779981 located near RAVER1 and rs73009538 near CARM1. In hospitalized patients, two previously unrecognized loci were detected, namely, rs115545251 near GFI1 and rs3181049 near RAVER1, while in general patients, rs11065822 near CUX2 emerged as a newly identified locus. We also identified potential target genes, including those involved in inflammation signaling (CARM1), endothelial dysfunction (INTS12), and antiviral immune response (RAVER1), which may require further investigation. Conclusion Our study offers insights into the genetic overlap between COVID-19 and immune factors, suggesting potential directions for future research and clinical exploration.
Collapse
Affiliation(s)
- Ziang Meng
- Department of Infectious Disease, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Chumeng Zhang
- The Second School of Clinical Medicine of Shandong First Medical University, Tai’an, Shandong, China
| | - Shuai Liu
- Agricultural Products Quality and Safety Center of Jinan, Jinan, Shandong, China
| | - Wen Li
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Yue Wang
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Qingyi Zhang
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Bichen Peng
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Weiyi Ye
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Yue Jiang
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Yingchao Song
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Miao Guo
- School of Life Sciences, Shandong First Medical University, Shandong, China
| | - Xiao Chang
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Lei Shao
- Department of Infectious Disease, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
43
|
Shen Y, Dong X, Li X, Shi Z, Shao T, Jiang J, Song J. WNT inhibitor SP5-mediated SERPING1 suppresses lung adenocarcinoma progression via TSC2/mTOR pathway. Cell Death Dis 2025; 16:103. [PMID: 39962118 PMCID: PMC11832940 DOI: 10.1038/s41419-025-07440-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/25/2025] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
The long-term outlook for patients grappling with lung cancer (LC) remains bleak, with lung adenocarcinoma (LUAD) emerging as the most predominant histological subtype. Our Mendelian randomization (MR) investigation spotlighted that heightened levels of the circulating protein serpin peptidase inhibitor family G1 (SERPING1) substantially mitigated LC risk. The fusion of multi-omics strategies unveiled that SERPING1 exhibited diminished expression in LUAD patients compared to healthy individuals both in tissues and serum, with LUAD individuals showcasing elevated SERPING1 expression demonstrating improved prognoses. Furthermore, SERPING1 expression exhibited a robust correlation with the efficacy of immunotherapy. Through meticulous in vivo and in vitro analyses, we unraveled that SERPING1 impeded the proliferation, migration, invasion and wound healing of LUAD cells via the tuberous sclerosis 2 (TSC2)/mammalian target of rapamycin (mTOR) pathway. Mechanistically, WNT inhibitor- Specificity Protein (SP5) was delineated as facilitator of SERPING1 transcription by binding to the SERPING1 gene promoter. Intriguingly, aside from the association between SERPING1 and systolic blood pressure, glycosylated hemoglobin (HbA1c), type I diabetes, no discernible link between SERPING1 overexpression and heightened risks of other cardiometabolic conditions and diseases was evident. In summary, SERPING1 emerges as a novel tumor suppressor gene and SP5/SERPING1/TSC2 is a promising therapeutic target in the context of LUAD.
Collapse
Affiliation(s)
- Yefeng Shen
- Institute of Cardiovascular Sciences, Guangxi Academy of Medical Sciences, Nanning, China
- Department of Thoracic Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaofeng Dong
- Department of Hepatobiliary, Pancreas and Spleen Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region (Guangxi Academy of Medical Sciences), Nanning, China
| | - Xujia Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhiyuan Shi
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin, China
| | - Tingting Shao
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Junlan Jiang
- Department of Pathology, the First Affiliated Hospital, Anhui Medical University, Hefei, China
- Pathology Center, Anhui Medical University, Hefei, China
| | - Jian Song
- Institute of Cardiovascular Sciences, Guangxi Academy of Medical Sciences, Nanning, China.
- Department of Radiation Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
44
|
Wang M, Daghlas I, Zhang Z, Gill D, Liu D. MTHFR Polymorphisms, Homocysteine Elevation, and Ischemic Stroke Susceptibility in East Asian and European Populations. Neurology 2025; 104:e210245. [PMID: 39787475 DOI: 10.1212/wnl.0000000000210245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/31/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Methylenetetrahydrofolate reductase (MTHFR) is a key enzyme that regulates folate and homocysteine metabolism. Genetic variation in MTHFR has been implicated in cerebrovascular disease risk, although research in diverse populations is lacking. We thus aimed to investigate the effect of genetically predicted MTHFR activity on risk of ischemic stroke (IS) and its main subtypes using a multiancestry Mendelian randomization (MR) approach. METHODS We proxied reduced MTHFR function using the C677T missense variant that impairs MTHFR function and consequently increases levels of total plasma homocysteine (tHcy) in both East Asian and European populations. Summary data for IS and its subtypes (small vessel stroke [SVS], large artery stroke [LAS], and cardioembolic stroke [CES]) were obtained from the largest available genome-wide association studies. MR estimates were calculated using the Wald ratio and random-effects inverse-variance-weighted methods. We performed sensitivity analyses to evaluate for confounding due to linkage disequilibrium. RESULTS Genetically downregulated MTHFR activity, associated with a consequent SD increase in tHcy levels, was associated with an increased risk of SVS in both East Asian (odds ratio [OR] 1.20, 95% CI 1.08-1.34, p = 8.58 × 10-4) and European populations (OR 1.62, 95% CI 1.24-2.12; p = 3.73 × 10-4). There was no evidence that genetically perturbed MTHFR activity influenced risk of CES or LAS. These findings were consistent in sensitivity analyses. DISCUSSION Our findings provide genetic evidence that reduced MTHFR activity was selectively associated with an increased risk of SVS in both East Asian and European populations. These findings warrant further investigation of genotype-guided nutritional supplementation for the prevention of SVS.
Collapse
Affiliation(s)
- Mengmeng Wang
- Department of Neurology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Iyas Daghlas
- Department of Neurology, University of California, San Francisco
| | - Zhizhong Zhang
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, China
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, United Kingdom; and
| | - Dandan Liu
- Department of Integrated Traditional Chinese and Western Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
45
|
Yi D, Byun MS, Park J, Kim J, Jung G, Ahn H, Lee J, Lee Y, Kim YK, Kang KM, Sohn C, Liu S, Huang Y, Saykin AJ, Lee DY, Nho K, for the KBASE research group. Tau pathway-based gene analysis on PET identifies CLU and FYN in a Korean cohort. Alzheimers Dement 2025; 21:e14416. [PMID: 39625110 PMCID: PMC11848168 DOI: 10.1002/alz.14416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 12/13/2024]
Abstract
INTRODUCTION The influence of genetic variation on tau protein aggregation, a key factor in Alzheimer's disease (AD), remains not fully understood. We aimed to identify novel genes associated with brain tau deposition using pathway-based candidate gene association analysis in a Korean cohort. METHODS We analyzed data for 146 older adults from the well-established Korean AD continuum cohort (Korean Brain Aging Study for the Early Diagnosis and Prediction of Alzheimer's Disease; KBASE). Fifteen candidate genes related to both tau pathways and AD were selected. Association analyses were performed using PLINK: A tool set for whole-genome association and population-based linkage analyses (PLINK) on tau deposition measured by 18F-AV-1451 positron emission tomography (PET) scans, with additional voxel-wise analysis conducted using Statistical Parametric Mapping 12 (SPM12). RESULTS CLU and FYN were significantly associated with tau deposition, with the most significant single-nucleotide polymorphisms (SNPs) being rs149413552 and rs57650567, respectively. These SNPs were linked to increased tau across key brain regions and showed additive effects with apolipoprotein E (APOE) ε4. DISCUSSION CLU and FYN may play specific roles in tau pathophysiology, offering potential targets for biomarkers and therapies. HIGHLIGHTS Gene-based analysis identified CLU and FYN as associated with tau deposition on positron emission tomography (PET). CLU rs149413552 and FYN rs57650567 were associated with brain tau deposition. rs149413552 and rs57650567 were associated with structural brain atrophy. CLU rs149413552 was associated with immediate verbal memory. CLU and FYN may play specific roles in tau pathophysiology.
Collapse
Affiliation(s)
- Dahyun Yi
- Institute of Human Behavioral MedicineMedical Research CenterSeoul National UniversitySeoulSouth Korea
| | - Min Soo Byun
- Department of NeuropsychiatrySeoul National University HospitalSeoulSouth Korea
- Department of PsychiatrySeoul National University College of MedicineSeoulSouth Korea
| | - Jong‐Ho Park
- Precision Medicine CenterSeoul National University Bundang HospitalSeongnam‐siGyeonggi‐doSouth Korea
| | - Jong‐Won Kim
- Department of Laboratory Medicine and GeneticsSamsung Medical CenterSungkyunkwan University School of MedicineGangnam‐guSeoulSouth Korea
| | - Gijung Jung
- Institute of Human Behavioral MedicineMedical Research CenterSeoul National UniversitySeoulSouth Korea
| | - Hyejin Ahn
- Interdisciplinary Program of Cognitive ScienceSeoul National University College of HumanitiesGwanak‐guSeoulSouth Korea
| | - Jun‐Young Lee
- Department of PsychiatrySeoul National University Boramae Medical Center, Dongjak‐guSeoulSouth Korea
| | - Yun‐Sang Lee
- Department of Nuclear MedicineSeoul National University College of MedicineJongro‐guSeoulSouth Korea
| | - Yu Kyeong Kim
- Department of Nuclear MedicineSeoul National University Boramae Medical Center, Dongjak‐guSeoulSouth Korea
| | - Koung Mi Kang
- Department of RadiologySeoul National University Hospital, Jongro‐guSeoulSouth Korea
- Department of RadiologySeoul National University College of Medicine, Jongro‐guSeoulSouth Korea
| | - Chul‐Ho Sohn
- Department of RadiologySeoul National University Hospital, Jongro‐guSeoulSouth Korea
| | - Shiwei Liu
- Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Center for NeuroimagingDepartment of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Yen‐Ning Huang
- Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Center for NeuroimagingDepartment of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Andrew J. Saykin
- Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Center for NeuroimagingDepartment of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Dong Young Lee
- Institute of Human Behavioral MedicineMedical Research CenterSeoul National UniversitySeoulSouth Korea
- Department of NeuropsychiatrySeoul National University HospitalSeoulSouth Korea
- Department of PsychiatrySeoul National University College of MedicineSeoulSouth Korea
| | - Kwangsik Nho
- Indiana Alzheimer's Disease Research CenterIndiana University School of MedicineIndianapolisIndianaUSA
- Center for NeuroimagingDepartment of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | | |
Collapse
|
46
|
Liu J. Proteome-wide association studies have predicted that the protein abundance of LSM6, GMPPB, ICA1L, and CISD2 is associated with attention-deficit/hyperactivity disorder. Eur Child Adolesc Psychiatry 2025; 34:721-728. [PMID: 38954053 DOI: 10.1007/s00787-024-02517-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024]
Abstract
Identification of changes in protein abundance for attention-deficit/hyperactivity disorder (ADHD) is important for potential disease mechanisms and therapeutic study for ADHD. In order to identify candidate proteins that confer risk for ADHD, a proteome-wide association study (PWAS) for ADHD was conducted by integrating two human brain proteome datasets and the ADHD genome-wide association study (GWAS) summary statistics released by the Psychiatric Genomics Consortium (PGC). A total of 11 risk proteins were identified as significant candidates that passed the bonferroni corrected proteome-wide significant (PWS) level. The predicted protein abundance level of LSM6, GMPPB, ICA1L and CISD2 are shown significantly associated with ADHD in both proteome datasets, highlighting their potential role in ADHD pathogenesis. A transcriptome-wide association study (TWAS) of ADHD was also conducted, and 13 genes with predicted expression changes related to ADHD were identified. GMPPB, ICA1L and NAT6 were supported by both TWAS and PWASs analysis. This study uncovers the predicted protein abundance changes that confer risk for ADHD and pinpoints a number of high-confidence protein candidates (e.g. LSM6, GMPPB, ICA1L, CISD2) for further functional exploration studies and drug development targeting these proteins.
Collapse
Affiliation(s)
- Jiewei Liu
- Department of Psychiatry, Wuhan Mental Health Center, Wuhan, 430012, Hubei, China.
- Department of Psychiatry, Wuhan Hospital for Psychotherapy, Wuhan, 430012, Hubei, China.
| |
Collapse
|
47
|
Greenwood E, Cao M, Lee CM, Liu A, Moyo B, Bao G, Gibson G. Haplotype rather than single causal variants effects contribute to regulatory gene expression associations in human myeloid cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635675. [PMID: 39975189 PMCID: PMC11838257 DOI: 10.1101/2025.01.30.635675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Genome-wide association studies typically identify hundreds to thousands of loci, many of which harbor multiple independent peaks, each parsimoniously assumed to be due to the activity of a single causal variant. Fine-mapping of such variants has become a priority and since most associations are located within regulatory regions, it is also assumed that they colocalize with regulatory variants that influence the expression of nearby genes. Here we examine these assumptions by using a moderate throughput expression CROPseq protocol in which Cas9 nuclease is used to induce small insertions and deletions across the credible set of SNPs that may account for expression quantitative trait loci (eQTL) for genes associated with inflammatory bowel disease (IBD). Of the 4,384 SNPs targeted in 88 loci (an average of 50 per locus), 439 were significant and further examined for validation. From these, 98 significantly altered target gene expression in HL-60 myeloid cell line, 74 in induced macrophages from these HL-60 cells, and 78 in induced neutrophils for a total of 201 validated effects (46%), 43 of which were observed in at least two of the cell types. Considering the observed sensitivity and specificity of the controls, we estimate that there are at least 150 true positives per cell type, an average of almost 2.4 for each of the 64 eQTL for which putative causal variants have been fine-mapped. This implies that haplotype effects are likely to explain many of the associations. We also demonstrate that the same approach can be used to investigate the activity of very rare variants in regulatory regions for 89 genes, providing a rapid strategy for establishing clinical relevance of non-coding mutations.
Collapse
Affiliation(s)
- Emily Greenwood
- School of Biological Sciences, Georgia Institute of Technology, Atlanta GA 30332, USA
| | - Mingming Cao
- Department of Bioengineering, Rice University, Houston TX 77005, USA
| | - Ciaran M. Lee
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Aidi Liu
- Department of Bioengineering, Rice University, Houston TX 77005, USA
| | - Buhle Moyo
- Department of Bioengineering, Rice University, Houston TX 77005, USA
| | - Gang Bao
- Department of Bioengineering, Rice University, Houston TX 77005, USA
| | - Greg Gibson
- School of Biological Sciences, Georgia Institute of Technology, Atlanta GA 30332, USA
| |
Collapse
|
48
|
Nguyen H, Lin C, Bell K, Huang A, Hannum M, Ramirez V, Christensen C, Rawson NE, Colquitt L, Domanico P, Sasimovich I, Herriman R, Joseph P, Braimah O, Reed DR. Worldwide study of the taste of bitter medicines and their modifiers. Chem Senses 2025; 50:bjaf003. [PMID: 39902731 PMCID: PMC12010088 DOI: 10.1093/chemse/bjaf003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Indexed: 02/06/2025] Open
Abstract
The bitter taste of medicines hinders patient compliance, but not everyone experiences these difficulties because people worldwide differ in their bitterness perception. To better understand how people from diverse ancestries perceive medicines and taste modifiers, 338 adults, European and recent US and Canadian immigrants from Asia, South Asia, and Africa, rated the bitterness intensity of taste solutions on a 100-point generalized visual analog scale and provided a saliva sample for genotyping. The taste solutions were 5 medicines, tenofovir alafenamide (TAF), moxifloxacin, praziquantel, amodiaquine, and propylthiouracil (PROP), and 4 other solutions, TAF mixed with sucralose (sweet, reduces bitterness) or 6-methylflavone (tasteless, reduces bitterness), sucralose alone, and sodium chloride alone. Bitterness ratings differed by ancestry for 2 of the 5 drugs (amodiaquine and PROP) and for TAF mixed with sucralose. Genetic analysis showed that people with variants in 1 bitter receptor variant gene (TAS2R38) reported PROP was more bitter than did those with a different variant (P = 7.6e-19) and that people with either an RIMS2 or a THSD4 genotype found sucralose more bitter than did others (P = 2.6e-8, P = 7.9e-11, respectively). Our findings may help guide the formulation of bad-tasting medicines to meet the needs of those most sensitive to them.
Collapse
Affiliation(s)
- Ha Nguyen
- Monell Chemical Senses Center, Philadelphia, PA, United States
| | - Cailu Lin
- Monell Chemical Senses Center, Philadelphia, PA, United States
| | - Katherine Bell
- Monell Chemical Senses Center, Philadelphia, PA, United States
| | - Amy Huang
- Monell Chemical Senses Center, Philadelphia, PA, United States
| | | | - Vicente Ramirez
- Monell Chemical Senses Center, Philadelphia, PA, United States
| | | | - Nancy E Rawson
- Monell Chemical Senses Center, Philadelphia, PA, United States
| | - Lauren Colquitt
- Monell Chemical Senses Center, Philadelphia, PA, United States
| | - Paul Domanico
- Clinton Health Access Initiative, Boston, MA, United States
| | | | - Riley Herriman
- Monell Chemical Senses Center, Philadelphia, PA, United States
| | - Paule Joseph
- National Institute of Alcohol Abuse and Alcoholism and National Institute of Nursing Research, Bethesda, MD, United States
| | | | - Danielle R Reed
- Monell Chemical Senses Center, Philadelphia, PA, United States
| |
Collapse
|
49
|
Dou L, Xu Z, Xu J, Zang C, Su C, Pieper AA, Leverenz JB, Wang F, Zhu X, Cummings J, Cheng F. A network-based systems genetics framework identifies pathobiology and drug repurposing in Parkinson's disease. NPJ Parkinsons Dis 2025; 11:22. [PMID: 39837893 PMCID: PMC11751448 DOI: 10.1038/s41531-025-00870-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 01/06/2025] [Indexed: 01/23/2025] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disorder. However, current treatments only manage symptoms and lack the ability to slow or prevent disease progression. We utilized a systems genetics approach to identify potential risk genes and repurposable drugs for PD. First, we leveraged non-coding genome-wide association studies (GWAS) loci effects on five types of brain-specific quantitative trait loci (xQTLs, including expression, protein, splicing, methylation and histone acetylation) under the protein-protein interactome (PPI) network. We then prioritized 175 PD likely risk genes (pdRGs), such as SNCA, CTSB, LRRK2, DGKQ, and CD44, which are enriched in druggable targets and differentially expressed genes across multiple human brain-specific cell types. Integrating network proximity-based drug repurposing and patient electronic health record (EHR) data observations, we identified Simvastatin as being significantly associated with reduced incidence of PD (hazard ratio (HR) = 0.91 for fall outcome, 95% confidence interval (CI): 0.87-0.94; HR = 0.88 for dementia outcome, 95% CI: 0.86-0.89) after adjusting for 267 covariates. In summary, our network-based systems genetics framework identifies potential risk genes and repurposable drugs for PD and other neurodegenerative diseases if broadly applied.
Collapse
Affiliation(s)
- Lijun Dou
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Zhenxing Xu
- Department of Population Health Sciences, Weill Cornell Medical College, Cornell University, New York, NY, 10065, USA
| | - Jielin Xu
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Chengxi Zang
- Department of Population Health Sciences, Weill Cornell Medical College, Cornell University, New York, NY, 10065, USA
| | - Chang Su
- Department of Population Health Sciences, Weill Cornell Medical College, Cornell University, New York, NY, 10065, USA
| | - Andrew A Pieper
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA
- Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
- Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, 44106, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
- Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA
| | - James B Leverenz
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, 44195, USA
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Fei Wang
- Department of Population Health Sciences, Weill Cornell Medical College, Cornell University, New York, NY, 10065, USA
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, Kirk Kerkorian School of Medicine, UNLV, Las Vegas, NV, 89154, USA
| | - Feixiong Cheng
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA.
| |
Collapse
|
50
|
Pan Q, Tao Y, Cai T, Veluchamy A, Hebert HL, Zhu P, Haque M, Dottorini T, Colvin LA, Smith BH, Meng W. A genome-wide association study identifies genetic variants associated with hip pain in the UK Biobank cohort (N = 221,127). Sci Rep 2025; 15:2812. [PMID: 39843573 PMCID: PMC11754597 DOI: 10.1038/s41598-025-85871-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025] Open
Abstract
Hip pain is a common musculoskeletal complaint that leads many people to seek medical attention. We conducted a primary genome-wide association study (GWAS) on the hip pain phenotype within the UK Biobank cohort. Sex-stratified GWAS analysis approach was also performed to explore sex specific variants associated with hip pain. We found seven different loci associated with hip pain at GWAS significance level, with the most significant single nucleotide polymorphism (SNP) being rs77641763 within the EXD3 (p value = 2.20 × 10-13). We utilized summary statistics from the FinnGen cohort and a previous GWAS meta-analysis on hip osteoarthritis as replication cohorts. Four loci (rs509345, rs73581564, rs9597759, rs2018384) were replicated with a p value less than 0.05. Sex-stratified GWAS analyses revealed a unique locus within the CUL1 gene (rs4726995, p = 2.56 × 10-9) in males, and three unique loci in females: rs1651359966 on chromosome 7 (p = 1.15 × 10-8), rs552965738 on chromosome 9 (p = 2.72 × 10-8), and rs1978969 on chromosome 13 (p = 2.87 × 10-9). This study has identified seven genetic loci associated with hip pain. Sex-stratified analysis also revealed sex specific variants associated with hip pain in males and females. This study has provided a foundation for advancing research of hip pain and hip osteoarthritis.
Collapse
Affiliation(s)
- Qi Pan
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, 315100, China
| | - Yiwen Tao
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, 315100, China
| | - Tengda Cai
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, 315100, China
| | - Abi Veluchamy
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD2 4BF, UK
| | - Harry L Hebert
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD2 4BF, UK
| | - Peixi Zhu
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang, China
| | - Mainul Haque
- School of Mathematical Sciences, University of Nottingham Ningbo China, Ningbo, 315100, China
| | - Tania Dottorini
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, LE12 5RD, UK
| | - Lesley A Colvin
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD2 4BF, UK
| | - Blair H Smith
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD2 4BF, UK
| | - Weihua Meng
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, 315100, China.
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD2 4BF, UK.
- Center for Public Health, Faculty of Medicine, Health and Life Sciences, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, BT12 6BA, UK.
| |
Collapse
|