1
|
Abstract
BACKGROUND Clonal hematopoiesis of indeterminate potential (CHIP), characterized by the age-related expansion of blood cells carrying preleukemic mutations, is associated with immune aging. This study aimed to investigate the association between CHIP and established autoimmune diseases. METHODS We analyzed baseline data from 456,692 UK Biobank participants with available whole-exome sequences. The primary outcome was 19 autoimmune disorders. Associations among any CHIP (variant allele fraction ≥2%), large CHIP clones (variant allele fraction ≥10%), and gene-specific CHIP subtypes with the incidence of autoimmune diseases were assessed using Cox regression. Mediation analysis was performed to explore the role of inflammation in the link between CHIP and autoimmune diseases. RESULTS We identified 17,433 any CHIP and 11,970 large CHIP at baseline. Participants with any and large CHIP were associated with 44% and 43% higher risk for Crohn's disease, 25% and 33% higher risk for psoriasis, 13% and 14% higher risk for rheumatoid arthritis, and 35% and 55% higher risk for vasculitis, respectively. Participants with CHIP status were associated with increased levels of inflammatory markers, including white blood cell, platelets, neutrophils, and neutrophil-to-lymphocyte ratio, with overall mediation ratios of 16.3% for Crohn's disease, 7.1% for psoriasis, 23.2% for rheumatoid arthritis, and 7.2% for vasculitis. CONCLUSIONS CHIP was associated with an increased risk for incident multiple autoimmune diseases, including Crohn's disease, psoriasis, vasculitis, and rheumatoid arthritis, potentially mediated by elevated inflammatory levels. Future research is needed to clarify the mechanisms underlying these associations and to explore potential interventions to reduce the associated risk.
Collapse
Affiliation(s)
- Hanzhang Wu
- Department of Big Data in Health Science, Zhejiang University School of Public Health and Department of Psychiatry, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang ProvinceHangzhouChina
| | - Jiahe Wei
- Department of Big Data in Health Science, Zhejiang University School of Public Health and Department of Psychiatry, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang ProvinceHangzhouChina
| | - Yuefeng Yu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ningjian Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiao Tan
- Department of Big Data in Health Science, Zhejiang University School of Public Health and Department of Psychiatry, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang ProvinceHangzhouChina
- Department of Medical SciencesUppsala UniversityUppsalaSweden
| |
Collapse
|
2
|
Kronzer VL, Williamson KA, Hanson AC, Sletten JA, Sparks JA, Davis JM, Crowson CS. Quantifying and improving rheumatoid arthritis algorithm performance in biobank settings. Semin Arthritis Rheum 2025; 72:152668. [PMID: 40024070 PMCID: PMC12048211 DOI: 10.1016/j.semarthrit.2025.152668] [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: 08/16/2024] [Accepted: 02/17/2025] [Indexed: 03/04/2025]
Abstract
OBJECTIVE To quantify and improve the performance of standard rheumatoid arthritis (RA) algorithms in a biobank setting. METHODS This retrospective cohort study within the Mayo Clinic (MC) Biobank and MC Tapestry Study identified RA cases by presence of at least two RA codes OR positive anti-cyclic citrullinated peptide antibodies (CCP) plus disease-modifying anti-rheumatic drug (DMARD) prescription as of 7/18/2022. Rheumatology physicians manually verified all RA cases using RA criteria and/or rheumatology physician diagnosis plus DMARD use. All other biobank participants served as non-RA controls. We defined seropositivity as rheumatoid factor and/or anti-CCP positivity. We assessed rules-based and Electronic Medical Records and Genomics (eMERGE) RA algorithms using positive predictive value (PPV). Finally, we developed a novel RA algorithm using a LASSO-based machine learning approach with five-fold cross validation. RESULTS We identified 1,316 confirmed RA cases (968 MC Biobank, 348 Tapestry, 70 % seropositive) and 82,123 non-RA controls (mean age 65, 61 % female). The PPV of 3 RA codes was 43 %, codes plus DMARD was 54 %, and codes plus DMARD plus seropositivity was 85 %. The PPV of eMERGE was 77 %. Available in the MC Biobank, self-reported RA (PPV 10 %) only minimally improved algorithm performance (PPV from 83 % to 85 %), whereas family history of RA (PPV 3 %) worsened performance. At 90 % PPV, the novel RA algorithm incorporating key variables such as anti-CCP and DMARD use increased sensitivity by 4-11 % compared to eMERGE. CONCLUSION Rules-based and eMERGE RA algorithms had worse performance in biobank than administrative settings. Our novel RA algorithm outperformed these standard algorithms.
Collapse
Affiliation(s)
| | | | - Andrew C Hanson
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA.
| | | | - Jeffrey A Sparks
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, USA.
| | - John M Davis
- Division of Rheumatology, Mayo Clinic, Rochester, Minnesota, USA.
| | - Cynthia S Crowson
- Division of Rheumatology, Mayo Clinic, Rochester, Minnesota, USA; Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA.
| |
Collapse
|
3
|
Bockenhauer D, Konrad M. Phosphate transporters, candidate genes, and the prosecutor's fallacy. Pediatr Nephrol 2025; 40:1825-1829. [PMID: 39841236 DOI: 10.1007/s00467-025-06660-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/23/2025]
Affiliation(s)
- Detlef Bockenhauer
- Department of Paediatric Nephrology, UZ Leuven and Department of Cellular and Molecular Physiology, KUL, Leuven, Belgium.
- Great Ormond Street Hospital for Children and Center for Genetics and Genomics, UCL Department of Renal Medicine, University College London, London, UK.
| | - Martin Konrad
- Department of General Pediatrics, Pediatric Nephrology, University Hospital Münster, Münster, Germany
| |
Collapse
|
4
|
Wang ZY, He XY, Wu BS, Yang L, You J, Liu WS, Feng JF, Cheng W, Yu JT. Whole-exome sequencing identifies 5 novel genes associated with carpal tunnel syndrome. Hum Mol Genet 2025:ddaf076. [PMID: 40382669 DOI: 10.1093/hmg/ddaf076] [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: 01/17/2025] [Revised: 04/07/2025] [Accepted: 05/04/2025] [Indexed: 05/20/2025] Open
Abstract
Carpal tunnel syndrome (CTS), a common peripheral nerve entrapment disorder, has a high estimated heritability index. Although previous genome-wide association studies have assessed common genetic components of CTS, the risk contributed by coding variants is still not well understood. Here, we performed the largest exome-wide analyses using UK Biobank data from 350 770 participants to find coding variants associated with CTS. We then explored the relative contribution of both rare mutations and polygenic risk score (PRS) to CTS risk in survival analyses. Finally, we investigated the functional pathways of the CTS-related coding genes identified above. Aside from conforming 6 known CTS genes, 5 novel genes were identified (SPSB1, SYNC, ITGB5, MUC13 and LOXL4). The associations of most genes we identified with incident CTS were striking in survival analyses. Additionally, we provided evidence that combining rare coding alleles and polygenic risk score can improve the genetic prediction of CTS. Functional enrichment analyses revealed potential roles of the identified coding variants in CTS pathogenesis, where they contributed to extracellular matrix organization. Our results evaluated the contribution to CTS etiology from quantities of coding variants accessible to exome sequencing data.
Collapse
Affiliation(s)
- Zi-Yi Wang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, 12th Wulumuqi Zhong Road, Shanghai 200040, China
| | - Xiao-Yu He
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, 12th Wulumuqi Zhong Road, Shanghai 200040, China
| | - Bang-Sheng Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, 12th Wulumuqi Zhong Road, Shanghai 200040, China
| | - Liu Yang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, 12th Wulumuqi Zhong Road, Shanghai 200040, China
| | - Jia You
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, 220 Handan Rd., Yangpu District, Shanghai 200433, China
| | - Wei-Shi Liu
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, 220 Handan Rd., Yangpu District, Shanghai 200433, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, 220 Handan Rd., Yangpu District, Shanghai 200433, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, 220 Handan Rd., Yangpu District, Shanghai 200433, China
- Department of Computer Science, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, United Kingdom
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, 12th Wulumuqi Zhong Road, Shanghai 200040, China
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, 220 Handan Rd., Yangpu District, Shanghai 200433, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, 220 Handan Rd., Yangpu District, Shanghai 200433, China
- Department of Computer Science, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, United Kingdom
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, 12th Wulumuqi Zhong Road, Shanghai 200040, China
| |
Collapse
|
5
|
Kramarenko DR, Jurgens SJ, Pinto YM, Bezzina CR, Amin AS. Polygenic Risk Scores in Dilated Cardiomyopathy: Towards the Future. Curr Cardiol Rep 2025; 27:87. [PMID: 40369171 PMCID: PMC12078399 DOI: 10.1007/s11886-025-02239-2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/15/2025] [Indexed: 05/16/2025]
Abstract
PURPOSE OF REVIEW Genome-wide association studies (GWASs) have recently shown that common genetic variations significantly affect the risk of developing dilated cardiomyopathy (DCM). This has enabled the development of polygenic scores (PGSs), which aim to aggregate the impact of multiple common genetic variants across the genome to provide an overall genetic risk score for disease manifestation and disease severity. In this review, we discuss the latest findings pertaining to GWASs and PGSs for DCM and various ways in which PGSs could improve the management of patients with DCM or risk of developing DCM. RECENT FINDINGS In 2024 the two largest GWAS meta-analyses for DCM were published. Notably, both studies produced PGSs that were able to discriminate healthy subjects from DCM patients which brings promise for potential clinical application of the scores. Large-scale GWAS have identified common genetic variants associated with DCM, leading to the development of PGS, which show strong associations with disease risk and hold potential for clinical applications. However, before clinical implementation, further research is needed to explore their utility in real-world settings and across diverse populations.
Collapse
Affiliation(s)
- Daria R Kramarenko
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Amsterdam, The Netherlands
| | - Sean J Jurgens
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Yigal M Pinto
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Amsterdam, The Netherlands
- Department of Clinical Cardiology, Heart Center, Amsterdam University Medical Centers, University of Amsterdam, Location AMC, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - Connie R Bezzina
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Amsterdam, The Netherlands
| | - Ahmad S Amin
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.
- European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Amsterdam, The Netherlands.
- Department of Clinical Cardiology, Heart Center, Amsterdam University Medical Centers, University of Amsterdam, Location AMC, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands.
| |
Collapse
|
6
|
Jing X, Liu Z, Li W, Ma K, Zhang J, Yan Z, Zhang S, Lin J, Zhao J, Ong KK, Perry JRB, Zhao Y. Protein-truncating variants in UQCRC1 are associated with Parkinson's disease: evidence from half-million people. NPJ Parkinsons Dis 2025; 11:120. [PMID: 40346065 PMCID: PMC12064775 DOI: 10.1038/s41531-025-00987-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/30/2025] [Indexed: 05/11/2025] Open
Abstract
Recent studies have suggested a potential but inconsistent link between UQCRC1 and Parkinson's disease (PD). For the first time, we systematically investigated the association between non-synonymous variants in UQCRC1 and PD risk using data from the UK Biobank with half-million participants, which provide evidence supporting the role of UQCRC1 Protein-truncating variants (PTVs) in PD (P = 1.20 × 10-6, OR = 6.59) and highlight the importance of large-scale population studies in identifying rare genetic risk factors.
Collapse
Affiliation(s)
- Xiaoxi Jing
- Changping Laboratory, Yard-28, Science Park Road, Changping District, Beijing, P. R. China
| | - Zongzhi Liu
- Changping Laboratory, Yard-28, Science Park Road, Changping District, Beijing, P. R. China
| | - Wenwen Li
- Innovation Center for Neurological Disorders and Department of Neurology, National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Kaiyan Ma
- Changping Laboratory, Yard-28, Science Park Road, Changping District, Beijing, P. R. China
| | - Jiaxiang Zhang
- Changping Laboratory, Yard-28, Science Park Road, Changping District, Beijing, P. R. China
| | - Zeqi Yan
- Changping Laboratory, Yard-28, Science Park Road, Changping District, Beijing, P. R. China
| | - Shuo Zhang
- Changping Laboratory, Yard-28, Science Park Road, Changping District, Beijing, P. R. China
| | - Jiecong Lin
- Changping Laboratory, Yard-28, Science Park Road, Changping District, Beijing, P. R. China
| | - Junpeng Zhao
- Changping Laboratory, Yard-28, Science Park Road, Changping District, Beijing, P. R. China
| | - Ken K Ong
- Innovation Center for Neurological Disorders and Department of Neurology, National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - John R B Perry
- MRC Epidemiology Unit and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Metabolic Research Laboratories, MRC Metabolic Diseases Unit and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Yajie Zhao
- Changping Laboratory, Yard-28, Science Park Road, Changping District, Beijing, P. R. China.
| |
Collapse
|
7
|
Tan Y, Zhu X, Huang Y, Zhao C, Cheng X, Li J, Zhang G, Ma T, Yang S, Bai Y. Clonal Hematopoiesis of Indeterminate Potential Is Associated With Incident Abdominal Aortic Aneurysm. Arterioscler Thromb Vasc Biol 2025. [PMID: 40336477 DOI: 10.1161/atvbaha.124.322630] [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: 02/14/2025] [Accepted: 04/22/2025] [Indexed: 05/09/2025]
Abstract
BACKGROUND Clonal hematopoiesis of indeterminate potential (CHIP) is an emerging risk factor for cardiovascular diseases. Genetic IL (interleukin)-6 signaling deficiency reduced cardiovascular disease risk in CHIP carriers. However, the association between CHIP and incident abdominal aortic aneurysm (AAA) and whether IL-6 signaling inhibition attenuates AAA risk among individuals with CHIP remained unclear. METHODS Participants without prevalent AAA from the UK Biobank were included. The associations of any CHIP (variant allele fraction, ≥2%), large CHIP (variant allele fraction, ≥10%), and gene-specific CHIP subtypes with incident AAA were investigated. The protection role of IL6R p.Asp358Ala, a genetic proxy for IL-6 deficiency, was tested after stratification by CHIP status. Furthermore, the interaction and joint effects of CHIP and genetic susceptibility on AAA risk were tested. RESULTS This study included 425 211 participants. Any CHIP and large CHIP was identified in 13 768 (3.2%) and 8576 (2.0%) participants, respectively. CHIP was associated with an increased risk of incident AAA (hazard ratio [HR], 1.21 [95% CI, 1.01-1.44]; P=0.034), with large CHIP clones exhibiting greater effect size (HR, 1.35 [95% CI, 1.10-1.66]; P=0.0045). Driver gene-specific analyses revealed that ASXL1-mediated CHIP exerted the strongest effect size on AAA risk (HR, 2.10 [95% CI, 1.54-2.88]; P<0.001). The presence of 2 IL6R p.Asp358Ala alleles attenuated the risk of AAA in large CHIP carriers (HR, 0.48 [95% CI, 0.23-0.99]; P=0.046). In the joint analysis, participants with CHIP and high genetic risk had a higher risk of developing AAA than those without CHIP and with low genetic risk (HR, 2.15 [95% CI, 1.63-2.85]; P<0.001). CONCLUSIONS CHIP is associated with an increased risk of AAA. Genetic IL-6 signaling deficiency attenuates the risk of AAA in large CHIP carriers. CHIP may serve as an attractive target for the prevention and treatment of AAA.
Collapse
Affiliation(s)
- Yu Tan
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
| | - Xuanmeng Zhu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
| | - Yuanfeng Huang
- Department of Geriatrics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China. (Y.H., J.L.)
- Bioinformatics Center, Xiangya Hospital and Furong Laboratory, Central South University, Changsha, Hunan, China. (Y.H., J.L.)
| | - Chenxuan Zhao
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
| | - Xunjie Cheng
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
| | - Jinchen Li
- Department of Geriatrics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China. (Y.H., J.L.)
- Bioinformatics Center, Xiangya Hospital and Furong Laboratory, Central South University, Changsha, Hunan, China. (Y.H., J.L.)
| | - Guogang Zhang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
| | - Tianqi Ma
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
| | - Shujun Yang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China. (Y.T., X.Z., C.Z., X.C., G.Z., T.M., S.Y.)
| | - Yongping Bai
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China. (Y.B.)
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China. (Y.B.)
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, China. (Y.B.)
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan, China. (Y.B.)
| |
Collapse
|
8
|
Dutta D, Chatterjee N. Expanding scope of genetic studies in the era of biobanks. Hum Mol Genet 2025:ddaf054. [PMID: 40312842 DOI: 10.1093/hmg/ddaf054] [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: 01/13/2025] [Revised: 03/25/2025] [Accepted: 04/08/2025] [Indexed: 05/03/2025] Open
Abstract
Biobanks have become pivotal in genetic research, particularly through genome-wide association studies (GWAS), driving transformative insights into the genetic basis of complex diseases and traits through the integration of genetic data with phenotypic, environmental, family history, and behavioral information. This review explores the distinct design and utility of different biobanks, highlighting their unique contributions to genetic research. We further discuss the utility and methodological advances in combining data from disease-specific study or consortia with that of biobanks, especially focusing on summary statistics based meta-analysis. Subsequently we review the spectrum of additional advantages offered by biobanks in genetic studies in representing population differences, calibration of polygenic scores, assessment of pleiotropy and improving post-GWAS in silico analyses. Advances in sequencing technologies, particularly whole-exome and whole-genome sequencing, have further enabled the discovery of rare variants at biobank scale. Among recent developments, the integration of large-scale multi-omics data especially proteomics and metabolomics, within biobanks provides deeper insights into disease mechanisms and regulatory pathways. Despite challenges like ascertainment strategies and phenotypic misclassification, biobanks continue to evolve, driving methodological innovation and enabling precision medicine. We highlight the contributions of biobanks to genetic research, their growing integration with multi-omics, and finally discuss their future potential for advancing healthcare and therapeutic development.
Collapse
Affiliation(s)
- Diptavo Dutta
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, Rockville, MD, 20879, United States
| | - Nilanjan Chatterjee
- Department of Biostatistics, Johns Hopkins University, 615 N Wolfe Street, Baltimore, MD, 21205, United States
- Department of Oncology, Johns Hopkins University, 615 N Wolfe Street, Baltimore, MD, 21205, United States
| |
Collapse
|
9
|
Holleman AM, Deaton AM, Hoffing RA, Krohn L, LoGerfo P, Nioi P, Plekan ME, Akle Serrano S, Ticau S, Walshe TE, Borodovsky A, Ward LD. Rare predicted loss-of-function and damaging missense variants in CFHR5 associate with protection from age-related macular degeneration. Am J Hum Genet 2025; 112:1062-1080. [PMID: 40250423 DOI: 10.1016/j.ajhg.2025.03.016] [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: 11/09/2024] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 04/20/2025] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of blindness among older adults worldwide, but treatment options are limited. Genetics studies have implicated the CFH locus, containing CFH and five CFHR genes, CFHR1-5, in AMD. While CFH has been robustly linked with AMD risk, potential additional roles for the CFHR genes remain unclear, obscured by strong linkage disequilibrium across the locus. Investigating rare coding variants can help to identify causal genes in such regions. We used whole-exome sequencing data from 406,952 UK Biobank participants to examine AMD associations with genes at the CFH locus. For each gene, we used burden testing to examine associations of rare (minor-allele frequency [MAF] < 1%) predicted loss-of-function (pLoF) and predicted damaging missense variants with AMD. We considered "broadly defined AMD" (ICD-10 35.3; ncases = 10,700) and "strictly defined AMD" (dry or wet AMD; ncases = 346). Adjusting for CFH-region variants known to independently associate with AMD, we find that CFHR5 rare variant burden significantly associates with a decreased risk of broadly defined AMD (odds ratio [OR] = 0.75, p = 7 × 10-4), with this association primarily driven by pLoF variants. Furthermore, the association of CFHR5 rare variants with AMD protection is estimated to be stronger for individuals with the CFH rs1061170 AMD risk allele (p.Tyr402His [p.Y402H]; interaction p = 0.04). Corresponding analyses of strict AMD were underpowered. However, we observe that thinning of the photoreceptor layer outer segment strongly predicts strict AMD and find that CFHR5 rare variant burden is significantly associated with increased thickness of this retinal layer (+0.34 SD, p = 4 × 10-4, n = 45,365). These findings suggest CFHR5 inhibition as a potential therapeutic approach for AMD.
Collapse
Affiliation(s)
| | | | | | - Lynne Krohn
- Alnylam Pharmaceuticals, Cambridge, MA 02142, USA
| | | | - Paul Nioi
- Alnylam Pharmaceuticals, Cambridge, MA 02142, USA
| | | | | | - Simina Ticau
- Alnylam Pharmaceuticals, Cambridge, MA 02142, USA
| | | | | | - Lucas D Ward
- Alnylam Pharmaceuticals, Cambridge, MA 02142, USA
| |
Collapse
|
10
|
Li ZY, Fei CJ, Yin RY, Kang JJ, Ma Q, He XY, Wu XR, Zhao YJ, Zhang W, Liu WS, Wu BS, Yang L, Zhu Y, Feng JF, Yu JT, Cheng W. Whole exome sequencing identified six novel genes for depressive symptoms. Mol Psychiatry 2025; 30:1925-1936. [PMID: 39472661 DOI: 10.1038/s41380-024-02804-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 04/24/2025]
Abstract
Previous genome-wide association studies of depression have primarily focused on common variants, limiting our comprehensive understanding of the genetic architecture. In contrast, whole-exome sequencing can capture rare coding variants, helping to explore the phenotypic consequences of altering protein-coding genes. Here, we conducted a large-scale exome-wide association study on 296,199 participants from the UK Biobank, assessing their depressive symptom scores through the Patient Health Questionnaire-4. We identified 22 genes associated with depressive symptoms, including 6 newly discovered genes (TRIM27, UBD, SVOP, ADGRB2, IRF2BPL, and ANKRD12). Both ontology enrichment analysis and plasma proteomics association analysis consistently revealed that the identified genes were associated with immune responses. Furthermore, we identified associations between these genes and brain regions related to depression, such as anterior cingulate cortex and orbitofrontal cortex. Additionally, phenome-wide association analysis demonstrated that TRIM27 and UBD were associated with neuropsychiatric, cognitive, biochemistry, and inflammatory traits. Our findings offer new insights into the potential mechanisms and genetic architecture of depressive symptoms.
Collapse
Affiliation(s)
- Ze-Yu Li
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Chen-Jie Fei
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Rui-Ying Yin
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Ju-Jiao Kang
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Qing Ma
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Xiao-Yu He
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Xin-Rui Wu
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yu-Jie Zhao
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Wei Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Wei-Shi Liu
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Bang-Sheng Wu
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Liu Yang
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Ying Zhu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
- Department of Computer Science, University of Warwick, Coventry, CV4 7AL, UK
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Jinhua, China
| | - Jin-Tai Yu
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| | - Wei Cheng
- Institute of Science and Technology for Brain-Inspired Intelligence, Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China.
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-inspired Intelligence, Zhejiang Normal University, Jinhua, China.
| |
Collapse
|
11
|
Ruth KS. Genetic insights into infertility from large-scale analyses. Nat Genet 2025; 57:1062-1063. [PMID: 40307585 DOI: 10.1038/s41588-025-02186-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Affiliation(s)
- Katherine S Ruth
- Faculty of Health and Life Sciences, University of Exeter, Exeter, UK.
| |
Collapse
|
12
|
Wijdeveld LFJM, Ajufo E, Challa SP, Rämö JT, Wang X, Kany S, Halford JL, Weng LC, Choi SH, Aragam KG, van Tintelen JP, Brundel BJJM, Jurgens SJ, Ellinor PT. Cardiomyopathy-Associated Gene Variants in Atrial Fibrillation. JAMA Cardiol 2025:2832996. [PMID: 40305039 PMCID: PMC12044542 DOI: 10.1001/jamacardio.2025.0460] [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: 10/29/2024] [Accepted: 02/11/2025] [Indexed: 05/02/2025]
Abstract
Importance Patients with atrial fibrillation (AF), a common morbid arrhythmia, are more likely to carry rare genetic variants associated with inherited cardiomyopathies. Prior studies on rare pathogenic variants in AF relied on small, hospital referral populations, and knowledge on clinical outcomes remains limited. Objective To evaluate the prevalence and prognostic implications of cardiomyopathy-associated pathogenic or likely pathogenic (CMP-PLP) genetic variants in patients with AF. Design, Setting, and Participants In 2 prospective cohort studies, the prevalence of CMP-PLP variants was assessed in the population of patients with AF and early-onset AF. The association between carrying a CMP-PLP variant and the risk of incident cardiomyopathy or heart failure (CMP/HF) after AF diagnosis was evaluated. Finally, the joint contributions of CMP-PLP variants, clinical risk, and polygenic risk were assessed. Included in this study were 2 large longitudinal cohort studies, the UK Biobank (UKB) (data 2006-2023) and the All of Us Research Program (AllofUs) (2018-2022). The UKB and AllofUs cohorts, respectively, contained 393 768 and 193 232 unrelated genotyped participants. Exposures CMP-PLP variants. Main Outcomes and Measures Prevalence of CMP-PLP variants and risk of incident CMP/HF after AF diagnosis. Results In the UKB cohort, 32 281 participants (8%) had AF (mean [SD] age, 62 [6] years; 20 459 male [63.4%]). In the AllofUs cohort, 11 901 participants (6%) had AF (mean [SD] age, 67 [12] years; 6576 male [55.3%]). Compared with the biobank populations, CMP-PLP variants were twice as prevalent in patients with AF (UKB, 2.04%; 95% CI, 1.89%-2.20%; AllofUs, 2.52%; 95% CI, 2.25%-2.82%) and 5 times as prevalent in AF with onset before age 45 years (UKB, 4.99%; 95% CI, 3.07%-7.91%; AllofUs, 4.66%; 3.40%-6.32%). Cumulative incidence of CMP/HF was high in patients with AF (18%) compared with patients without AF (3%). Still, among patients with AF without prior CMP/HF (UKB, 20 226; AllofUs, 8330), carrying a CMP-PLP variant was associated with 1.6-fold risk of incident CMP/HF (meta-analysis, 95% CI, 1.32-1.90). Finally, CMP-PLP variants, a polygenic score, and clinical risk factors were independent estimators of CMP/HF. Conclusions and Relevance Results of this cohort study suggest that the prevalence of CMP-PLP variants was substantial in patients with early-onset AF. Patients with AF carrying a CMP-PLP variant had an associated increased risk of future CMP/HF, independent of clinical and polygenic risk. These results indicate that genetic testing in patients with AF may identify individuals at higher risk for developing CMP/HF.
Collapse
Affiliation(s)
- Leonoor F. J. M. Wijdeveld
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Physiology, Amsterdam UMC, location Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, the Netherlands
| | - Ezimamaka Ajufo
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Saketh P. Challa
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Joel T. Rämö
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Xin Wang
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Shinwan Kany
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Cardiology, University Heart and Vascular Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Jennifer L. Halford
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medicine, Massachusetts General Hospital, Boston
| | - Lu-Chen Weng
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Seung Hoan Choi
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Biostatistics, Boston University, Boston, Massachusetts
| | - Krishna G. Aragam
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Bianca J. J. M. Brundel
- Department of Physiology, Amsterdam UMC, location Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, the Netherlands
| | - Sean J. Jurgens
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Experimental Cardiology, Amsterdam UMC, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, the Netherlands
| | - Patrick T. Ellinor
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Heart and Vascular Institute, Mass General Brigham, Boston, Massachusetts
| |
Collapse
|
13
|
Iden JA, Ben-Califa N, Naim A, Liron T, Neumann D, Gabet Y. Immunomodulatory function of cannabinoid receptor 2 and its agonist osteogenic growth peptide in health and cancer: a study in mice and humans. Oncogene 2025:10.1038/s41388-025-03399-9. [PMID: 40307509 DOI: 10.1038/s41388-025-03399-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 03/13/2025] [Accepted: 04/03/2025] [Indexed: 05/02/2025]
Abstract
Colon carcinoma is among the most prevalent malignant tumors, with inflammation being the primary risk factor. Cannabinoid receptor 2 (CB2/CNR2) has complex immunomodulatory functions. Therefore, we investigated the role of osteogenic growth peptide (OGP), an endogenous selective CB2 agonist, in colon carcinogenesis and immune modulation in transgenic mice (ApcMin/+).We injected 8-week-old (progression phase) or five-week-old (initiation phase) ApcMin/+ mice with OGP or vehicle weekly for 8 weeks or 4 weeks, respectively. During the progression phase, OGP-treated mice displayed significantly fewer tumors in the large intestine and smaller tumors in the small intestine. During the initiation phase, OGP significantly attenuated adenomagenesis in both the small and large intestine, decreased IL-6 and IL-4 levels, increased splenic anti-tumor CD8+ T cells, and diminished populations of tumor-promoting myeloid-derived suppressor cells. Further, we used exomic analyses of UKBiobank patients to determine the relationship between CNR2 polymorphisms and tumor-associated myeloid cells in humans. We found that the common CNR2-Q63R polymorphism is associated with monocyte count. Our results suggest that CB2 activation via OGP attenuates tumorigenesis and adenoma growth by modulating immune cells, corroborated by a significant association between CNR2 polymorphisms and monocytopoiesis in humans.
Collapse
Affiliation(s)
- Jennifer Ana Iden
- Department of Anatomy and Anthropology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel.
| | - Nathalie Ben-Califa
- Department of Cell and Developmental Biology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Aaron Naim
- Department of Anatomy and Anthropology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Tamar Liron
- Department of Anatomy and Anthropology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Drorit Neumann
- Department of Cell and Developmental Biology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yankel Gabet
- Department of Anatomy and Anthropology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
14
|
Qi B, Zhao J, Meng C, Bu P, Pu LQ, Zhao W, Li C, Xu Y. Exploring the association between systemic lupus erythematosus and osteonecrosis by Mendelian randomization analysis. Sci Rep 2025; 15:15040. [PMID: 40301378 PMCID: PMC12041352 DOI: 10.1038/s41598-025-95467-z] [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: 06/06/2024] [Accepted: 03/21/2025] [Indexed: 05/01/2025] Open
Abstract
Patients with systemic lupus erythematosus (SLE) have been shown to have a high risk of osteonecrosis, but the potential causal relationship between genetic susceptibility and risk of osteonecrosis is unclear. In this study, we used Mendelian randomization to investigate the effects of SLE, gout and rheumatoid arthritis on osteonecrosis, and performed post-GWAS localization and functional analyses of GWAS studies related to osteonecrosis, with the aim of obtaining a more in-depth understanding of the mechanisms of osteonecrosis. In this study, a total of 45 single nucleotide polymorphisms (SNPS) data associated with SLE from publicly available genome-wide association study (GIS) datasets were selected for magnetic resonance estimation using inverse-variance weighting, MR-Eagle method and weighted median method. The Cochrane Q-test, MR-Egger interception, MR-multidirectional residual and outlier methods, entrance/exit analysis and funnel plot were applied for sensitivity analysis. Two-sample Mendelian randomization analysis of the 19 SNPs obtained from screening showed no significant causal effect of SLE and osteonecrosis, and IVW and MR-Egger heterogeneity analyses showed no significant heterogeneity between the instrumental variables (P > 0.05). Multi-phenotype MR analysis showed no significant causal effect between gout and rheumatoid arthritis and osteonecrosis (p > 0.05). The available evidence does not support a significant causal effect of gout and rheumatoid arthritis on osteonecrosis, and the causal effect of SLE on the increased risk of osteonecrosis is only supported by the IVW method, which is of insufficient evidence validity, but suggests a better theoretical basis for the study of heritability related to SLE.
Collapse
Affiliation(s)
- Baochuang Qi
- First Clinical Medical College of Yunnan University of Chinese Medicine / Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, 650500, Yunnan, China
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Jinglin Zhao
- Medical Laboratory, Kunming Children's Hospital, Kunming, 650000, Yunnan, China
| | - Chen Meng
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Pengfei Bu
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Lu Qiao Pu
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Wanqiu Zhao
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China
| | - Chuan Li
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China.
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| | - Yongqing Xu
- Department of Orthopaedic, 920th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Kunming, Yunnan, China.
| |
Collapse
|
15
|
Parlar SC, Senkevich K, Yu E, Ruskey JA, Ahmad J, Asayesh F, Spiegelman D, Waters C, Monchi O, Dauvilliers Y, Dupré N, Greenbaum L, Hassin-Baer S, Miliukhina I, Timofeeva A, Emelyanov A, Pchelina S, Alcalay RN, Fon EA, Trempe JF, Gan-Or Z. LRRK2 rare-variant per-domain genetic burden in Parkinson's Disease: association confined to the kinase domain. NPJ Parkinsons Dis 2025; 11:102. [PMID: 40301370 PMCID: PMC12041573 DOI: 10.1038/s41531-025-00934-z] [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: 10/13/2024] [Accepted: 04/02/2025] [Indexed: 05/01/2025] Open
Abstract
LRRK2 variants are key genetic risk factors for Parkinson's Disease (PD). We conducted a per-domain rare coding variant burden analysis, including 8,888 PD cases and 69,412 controls. In meta-analysis, the Kinase domain was strongly associated with PD (Exonic: PFDR = 1.61 × 10-22, Non-synonymous: PFDR = 1.54 × 10-23, CADD > 20: PFDR = 3.09 × 10-24). Excluding the p.G2019S variant nullified this effect. Nominal associations were found in the ANK and Roc-COR domains, with potentially protective variants, p.R793M and p.Q1353K.
Collapse
Affiliation(s)
- Sitki Cem Parlar
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Konstantin Senkevich
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Department of Specialized Medicine, Division of Medical Genetics, McGill University Health Centre, Montreal, QC, Canada
| | - Eric Yu
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Jennifer A Ruskey
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Jamil Ahmad
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Farnaz Asayesh
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Dan Spiegelman
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
| | - Cheryl Waters
- Department of Neurology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Oury Monchi
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Department of Clinical Neurosciences and Department of Radiology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, Calgary, AB, Canada
| | - Yves Dauvilliers
- National Reference Center for Narcolepsy, Sleep Unit, Department of Neurology, Gui-de-Chauliac Hospital, CHU Montpellier, University of Montpellier, Montpellier, France
| | - Nicolas Dupré
- Division of Neurosciences, CHU de Québec, Université Laval, Quebec City, QC, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Lior Greenbaum
- The Danek Gertner Institute of Human Genetics, Sheba Medical Center, Ramat Gan, Israel
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Ramat Gan, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Sharon Hassin-Baer
- Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
- The Movement Disorders Institute, Department of Neurology, Sheba Medical Center, Tel Aviv, Israel
| | | | - Alla Timofeeva
- First Pavlov State Medical University of St. Petersburg, Saint Petersburg, Russia
| | - Anton Emelyanov
- First Pavlov State Medical University of St. Petersburg, Saint Petersburg, Russia
| | - Sofya Pchelina
- First Pavlov State Medical University of St. Petersburg, Saint Petersburg, Russia
| | - Roy N Alcalay
- Department of Neurology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
- Division of Movement Disorders, Tel Aviv Sourasky Medical Center, Tel Aviv-Yafo, Israel
| | - Edward A Fon
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Early Drug Discovery Unit (EDDU), Montreal Neurological Institute-Hospital (The Neuro), Montreal, QC, Canada
| | - Jean-François Trempe
- Department of Pharmacology & Therapeutics and Centre de Recherche en Biologie Structurale, McGill University, Montréal, QC, Canada
| | - Ziv Gan-Or
- Department of Human Genetics, McGill University, Montréal, QC, Canada.
- The Neuro (Montreal Neurological Institute-Hospital), McGill University, Montréal, QC, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada.
| |
Collapse
|
16
|
Mörseburg A, Zhao Y, Kentistou KA, Perry JRB, Ong KK, Day FR. Genetic determinants of proteomic aging. NPJ AGING 2025; 11:30. [PMID: 40287427 PMCID: PMC12033249 DOI: 10.1038/s41514-025-00205-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 02/21/2025] [Indexed: 04/29/2025]
Abstract
Changes in the proteome and its dysregulation have long been known to be a hallmark of aging. We derived a proteomic aging trait using data on 1459 plasma proteins from 44,435 UK Biobank individuals measured using an antibody-based assay. This metric is strongly associated with four age-related disease outcomes, even after adjusting for chronological age. Survival analysis showed that one-year older proteomic age, relative to chronological age, increases all-cause mortality hazard by 13 percent. We performed a genome-wide association analysis of proteomic age acceleration (proteomic aging trait minus chronological age) to identify its biological determinants. Proteomic age acceleration showed modest genetic correlations with four epigenetic clocks (Rg = 0.17 to 0.19) and telomere length (Rg = -0.2). Once we removed associations that were explained by a single pQTL, we were left with three signals mapping to BRCA1, POLR2A and TET2 with apparent widespread effects on plasma proteomic aging. Genetic variation at these three loci has been shown to affect other omics-related aging measures. Mendelian randomisation analyses showed causal effects of higher BMI and type 2 diabetes on faster proteomic age acceleration. This supports the idea that obesity and other features of metabolic syndrome have an adverse effect on the processes of human aging.
Collapse
Affiliation(s)
- Alexander Mörseburg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
| | - Yajie Zhao
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Katherine A Kentistou
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - John R B Perry
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Ken K Ong
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Felix R Day
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
| |
Collapse
|
17
|
Telis N, McEwen L, Bolze A, Lipschutz JH, Sweer LW, Judge DP, Pawloski PA, Grzymski JJ, Hajek C, Schiabor Barrett KM, Washington NL, Cirulli ET. Hypertension increases PPV for polycystic kidney disease in PKD1 and PKD2 variant carriers. HGG ADVANCES 2025; 6:100444. [PMID: 40269498 DOI: 10.1016/j.xhgg.2025.100444] [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: 09/10/2024] [Revised: 04/17/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the leading genetic form of KD. Although rare causal variants in the PKD1 and PKD2 genes have been identified, their penetrance and the disease progression and outcome are known to vary, and treatment efficacy in these carriers lags compared to patients with other forms of chronic KD (CKD). To develop a population screening strategy with high sensitivity to individuals likely to develop disease, we characterize the presentation and progression of ADPKD in variant carriers, identified in a multi-center all-comers cohort, as well as the UK Biobank. We show that the positive predictive value of hypertension for future diagnosis of KD is extremely high: 74% and 66% for PKD1 and PKD2, respectively. It is also highly preemptive, with hypertension occurring an average of 11 years before a KD diagnosis. Using pre-disease time point measurements of kidney function prior to their ADPKD diagnosis, we find that PKD1 and PKD2 variant carriers show significantly decreased kidney function (EGFR) an average of 5 years before their clinical diagnosis. Unlike other CKD patients, 54% of variant carriers with hypertension meet the diagnostic threshold for CKD years prior to their disease diagnosis, and their EGFRs are statistically indistinguishable from variant carriers who have already been diagnosed. These findings suggest that a population screening strategy using a combination of targeted sequencing and routine monitoring could identify cases of ADPKD with high sensitivity and support initiating treatment years prior to the current standard of care.
Collapse
Affiliation(s)
- Natalie Telis
- Helix, 101 South Ellsworth Avenue, Suite 350, San Mateo, CA 94401, USA.
| | - Lisa McEwen
- Helix, 101 South Ellsworth Avenue, Suite 350, San Mateo, CA 94401, USA
| | - Alexandre Bolze
- Helix, 101 South Ellsworth Avenue, Suite 350, San Mateo, CA 94401, USA
| | - Joshua H Lipschutz
- Division of Cardiology, Medical University of South Carolina, 30 Courtenay Drive, MSC 592, Charleston, SC 29425, USA
| | - Leon W Sweer
- Wellspan Pulmonary and Sleep Medicine, WellSpan Health, 157 North Reading Road, Ephrata, PA 17522, USA
| | - Daniel P Judge
- Division of Cardiology, Medical University of South Carolina, 30 Courtenay Drive, MSC 592, Charleston, SC 29425, USA; Center for Inherited Cardiovascular Diseases, WellSpan Health, 157 North Reading Road, Ephrata, PA 17522, USA
| | | | - Joseph J Grzymski
- Renown Institute for Health Innovation, Reno, NV 89512, USA; Center for Genomic Medicine, Desert Research Institute, 2215 Raggio Pkwy, Reno, NV 89512, USA
| | - Catherine Hajek
- Helix, 101 South Ellsworth Avenue, Suite 350, San Mateo, CA 94401, USA
| | | | | | | |
Collapse
|
18
|
He W, Shi J, Qian Y, Fan T, Cai X, Li H, Huang P, Shi Q. Evidence to shared genetic correlation of ischemic stroke and intracerebral hemorrhage and cardiovascular related traits. PLoS One 2025; 20:e0320479. [PMID: 40267100 PMCID: PMC12017486 DOI: 10.1371/journal.pone.0320479] [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: 06/19/2024] [Accepted: 02/20/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Previous studies have demonstrated the genetic basis of stroke and also revealed their genetic correlation with some cardiovascular related diseases or traits at the entire genome, which, however, would not give the answer which regions may mainly account for the genetic overlap. This study aims to identify specific genetic loci that contribute to the shared genetic basis between ischemic stroke subtypes and common cardiovascular traits. METHODS We used Local Analysis of [co]Variant Annotation (LAVA), a recent developed local genetic correlation method, to perform a system local genetic correlation analysis on GWAS summary data of two major subtypes of stroke, including any ischemic stroke (AIS) and intracerebral hemorrhage (ICH), and ten common cardiovascular related diseases or traits (CRTs). We further used colocalization analysis to explore potential shared causal genes in loci with significant local genetic correlation. In addition, we also performed Transcriptome-wide association (TWAS) analysis and fine-mapping for each phenotype to functionally annotate significant loci. RESULTS LAVA analysis identified a total of 3 significant local genetic correlations (Bonferroni-adjusted P < 0.05) across 3 chromosomes between AIS and systolic blood pressure (SBP), AIS and hypertension (HT), and ICH and body mass index (BMI), among which locus 7.24 explicated to harbor a shared causal variant for AIS and SBP. TWIST1 in locus 7.24 was defined to be nominally associated with SBP, but not for AIS. Fine-mapping analysis also only identified TWIST1 a credible causal gene for BMI. CONCLUSIONS Our study revealed the local genetic correlations between two stroke subtypes and ten common CRTs. Gene-level analyses indicated that biological explanations underlying these identified local genetic correlations may existed elsewhere beyond a common pattern of genetic-gene expression regulation.
Collapse
Affiliation(s)
- Wei He
- Department of Physical Medicine and Rehabilitation, The Affiliated Jiangyin People’s Hospital of Southeast University Medical College, Wuxi, China
| | - Jiajia Shi
- Department of Physical Medicine and Rehabilitation, Kunshan Rehabilitation Hospital, Suzhou, China
| | - Yiming Qian
- Department of Physical Medicine and Rehabilitation, The Affiliated Jiangyin People’s Hospital of Southeast University Medical College, Wuxi, China
| | - Tao Fan
- Department of Neurology, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Xuehong Cai
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Haochang Li
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Peng Huang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qin Shi
- Department of Neurology, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| |
Collapse
|
19
|
Tabata K, Sudo T, Nagata Y, Ihara K, Asada K, Kinoshita A, Tanaka Y, Yamauchi Y, Sasaki T, Hachiya H, Imai Y, Fujita H, Sasano T, Furukawa T, Iwata T, Tanaka T. Rare genetic variants involved in increased risk of paroxysmal atrial fibrillation in a Japanese population. Sci Rep 2025; 15:13216. [PMID: 40240483 PMCID: PMC12003908 DOI: 10.1038/s41598-025-97794-7] [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: 08/02/2024] [Accepted: 04/07/2025] [Indexed: 04/18/2025] Open
Abstract
Atrial fibrillation (AF) is the most prevalent arrhythmia in the world and can cause serious complications such as stroke or heart failure. Paroxysmal atrial fibrillation (PAF), a subtype of AF, accounts for approximately 25% of AF cases and is estimated to affect approximately 30 million people worldwide. Despite extensive genetic research on AF, the genetic factors involved in PAF in East Asian (EAS) populations remain unidentified. The aim of our study was to identify genetic factors associated with PAF in the Japanese population, contributing to our understanding of the genetic architecture of AF in Japanese populations. We conducted whole-exome sequencing on a cohort of 1176 PAF individuals and 1172 non-PAF control subjects in a Japanese population. We processed the sequencing data in accordance with the best practices outlined in the Genome Analysis Toolkit (GATK) and conducted gene-based association tests under three variant grouping strategies (masks) using the burden test, SKAT, and SKAT-O. We then performed a meta-analysis of the resulting P-values, which revealed that four genes-ZNF785, SMPD3, GFRA4, and LGALS1-were significantly associated with PAF, representing novel findings. These findings provide new insights into PAF pathogenesis and suggest potential biomarkers for early detection.
Collapse
Affiliation(s)
- Kanji Tabata
- Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo (Science Tokyo), 2-12-1 Ookayama, Meguro-ku, Tokyo, 152-8550, Japan
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
| | - Takeaki Sudo
- Department of Educational Media Development, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
| | - Yuki Nagata
- Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo (Science Tokyo), 2-12-1 Ookayama, Meguro-ku, Tokyo, 152-8550, Japan
- Bioresource Research Support Center, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
| | - Kensuke Ihara
- Department of Bio-Informational Pharmacology, Medicine Research Institute, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
- Department of Cardiovascular Medicine, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
| | - Ken Asada
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Atsuhiro Kinoshita
- Department of Educational Media Development, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
| | - Yasuaki Tanaka
- Department of Cardiology, Yokosuka Kyosai Hospital, Yokosuka, Japan
| | - Yasuteru Yamauchi
- Department of Cardiology, Yokohama City Minato Red Cross Hospital, Yokohama, Japan
| | - Takeshi Sasaki
- Department of Cardiology, Heart Rhythm Center, National Hospital Organization Disaster Medical Center, Tokyo, Japan
| | - Hitoshi Hachiya
- Cardiology Division, Cardiovascular Center, Tsuchiura Kyodo General Hospital, Ibaraki, Japan
| | - Yasushi Imai
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, Tochigi, Japan
| | - Hideo Fujita
- Division of Cardiovascular Medicine, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Tetsuo Sasano
- Department of Cardiovascular Medicine, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
| | - Tetsushi Furukawa
- Department of Bio-Informational Pharmacology, Medicine Research Institute, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
| | - Takanori Iwata
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan
| | - Toshihiro Tanaka
- Department of Human Genetics and Disease Diversity, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo (Science Tokyo), 2-12-1 Ookayama, Meguro-ku, Tokyo, 152-8550, Japan.
- Bioresource Research Support Center, Institute of Science Tokyo (Science Tokyo), Tokyo, Japan.
| |
Collapse
|
20
|
Wang YX, Fei CJ, Shen C, Ou YN, Liu WS, Yang L, Wu BS, Deng YT, Feng JF, Cheng W, Yu JT. Exome sequencing identifies protein-coding variants associated with loneliness and social isolation. J Affect Disord 2025; 375:192-204. [PMID: 39842675 DOI: 10.1016/j.jad.2025.01.096] [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: 05/24/2024] [Revised: 10/31/2024] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND Loneliness and social isolation are serious yet underappreciated public health problems, with their genetic underpinnings remaining largely unknown. We aimed to explore the role of protein-coding variants in the manifestation of loneliness and social isolation. METHODS We conducted the first exome-wide association analysis on loneliness and social isolation, utilizing 336,115 participants of white-British ancestry for loneliness and 346,115 for social isolation. Sensitivity analyses were performed to validate the genetic findings. We estimated the genetic burden heritability of loneliness and social isolation and provided biological insights into them. RESULTS We identified six novel risk genes (ANKRD12, RIPOR2, PTEN, ARL8B, NF1, and PIMREG) associated with loneliness and two (EDARADD and GIGYF1) with social isolation through analysis of rare coding variants. Brain-wide association analysis uncovered 47 associations between identified genes and brain structure phenotypes, many of which are critical for social processing and interaction. Phenome-wide association analysis established significant links between these genes and phenotypes across five categories, mostly blood biomarkers and cognitive measures. LIMITATIONS The measurements of loneliness and social isolation in UK Biobank are brief for these multi-layer social factors, some relevant aspects may be missed. CONCLUSIONS Our study revealed 13 risk genes associated with loneliness and 6 with social isolation, with the majority being novel discoveries. These findings advance our understanding of the genetic basis of these two traits. The study provides a foundation for future studies aimed at exploring the functional mechanisms of these genes and their potential implications for public health interventions targeting loneliness and social isolation.
Collapse
Affiliation(s)
- Yi-Xuan Wang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Chen-Jie Fei
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Chun Shen
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Wei-Shi Liu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Liu Yang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Bang-Sheng Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yue-Ting Deng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China; Department of Computer Science, University of Warwick, Coventry CV4 7AL, UK
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China; Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China; Department of Computer Science, University of Warwick, Coventry CV4 7AL, UK.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
21
|
Montano C, Timp W. Evolution of genome-wide methylation profiling technologies. Genome Res 2025; 35:572-582. [PMID: 40228903 PMCID: PMC12047278 DOI: 10.1101/gr.278407.123] [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] [Indexed: 04/16/2025]
Abstract
In this mini-review, we explore the advancements in genome-wide DNA methylation profiling, tracing the evolution from traditional methods such as methylation arrays and whole-genome bisulfite sequencing to the cutting-edge single-molecule profiling enabled by long-read sequencing (LRS) technologies. We highlight how LRS is transforming clinical and translational research, particularly by its ability to simultaneously measure genetic and epigenetic information, providing a more comprehensive understanding of complex disease mechanisms. We discuss current challenges and future directions in the field, emphasizing the need for innovative computational tools and robust, reproducible approaches to fully harness the capabilities of LRS in molecular diagnostics.
Collapse
Affiliation(s)
- Carolina Montano
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Winston Timp
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA;
| |
Collapse
|
22
|
Fiorica PN, Golmard L, Kim J, Bao R, Lin FY, Roy A, Pribnow A, Perrino MR, Masliah-Planchon J, Michalak-Provost S, Wong J, Filser M, Stoppa-Lyonnet D, Bourdeaut F, Brahimi A, Ingster O, Saulnier Sholler G, Jackson SA, Sasaki MM, Fowler T, Ng A, Corbett RJ, Kaufman RS, Haley JS, Carey DJ, Huang KL, Diskin SJ, Rokita JL, Al-Kateb H, McGee RB, Schiffman JD, Chen KS, Stewart DR, Williams Parsons D, Plon SE, Schultz KAP, Onel K. Germline Pathogenic DROSHA Variants Are Linked to Pineoblastoma and Wilms Tumor Predisposition. Clin Cancer Res 2025; 31:1491-1503. [PMID: 39992227 PMCID: PMC11995001 DOI: 10.1158/1078-0432.ccr-24-2785] [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: 08/27/2024] [Revised: 11/22/2024] [Accepted: 02/13/2025] [Indexed: 02/25/2025]
Abstract
PURPOSE DROSHA, DGCR8, and DICER1 regulate miRNA biogenesis and are commonly mutated in cancer. Although DGCR8 and DICER1 germline pathogenic variants (GPV) cause autosomal dominant tumor predisposition, no association between DROSHA GPVs and clinical phenotypes has been reported. EXPERIMENTAL DESIGN After obtaining informed consent, sequencing was performed on germline and tumor samples from all patients. The occurrence of germline DROSHA GPVs was investigated in large pediatric and adult cancer datasets. The population prevalence of DROSHA GPVs was investigated in the UK Biobank and Geisinger DiscovEHR cohorts. RESULTS We describe nine children from eight families with heterozygous DROSHA GPVs and a diagnosis of pineoblastoma (n = 8) or Wilms tumor (n = 1). A somatic second hit in DROSHA was detected in all eight tumors analyzed. All pineoblastoma tumors analyzed were classified as miRNA processing-altered 1 subtype. We estimate the population prevalence of germline DROSHA loss-of-function variants to be 1:3,875 to 1:4,843 but find no evidence for increased adult cancer risk. CONCLUSIONS This is the first report of DROSHA-related tumor predisposition. As pineoblastoma and Wilms tumor are also associated with DICER1 GPVs, our results suggest that the tissues of origin for these tumors are uniquely tolerant of general miRNA loss. The miRNA processing-altered 1 pineoblastoma subtype is associated with older age of diagnosis and better outcomes than other subtypes, suggesting DROSHA GPV status may have important clinical and prognostic significance. We suggest that genetic testing for DROSHA GPVs be considered for patients with pineoblastoma, Wilms tumor, or other DICER1-/DGCR8-related conditions and propose surveillance recommendations through research studies for individuals with DROSHA GPVs.
Collapse
Affiliation(s)
- Peter N. Fiorica
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Lisa Golmard
- Department of Genetics, Institut Curie, Paris, France
- PSL Research University, Paris, France
| | - Jung Kim
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Riyue Bao
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Frank Y. Lin
- Department of Pediatrics, Baylor College of Medicine, Houston Texas
- Texas Children’s Cancer and Hematology Center, Texas Children’s Hospital, Houston, Texas
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Angshumoy Roy
- Department of Pediatrics, Baylor College of Medicine, Houston Texas
- Texas Children’s Cancer and Hematology Center, Texas Children’s Hospital, Houston, Texas
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
- Department of Pathology, Texas Children’s Hospital, Houston, Texas
| | - Allison Pribnow
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Lucile Packard Children’s Hospital, Stanford University School of Medicine, Stanford, California
| | - Melissa R. Perrino
- Division of Cancer Predisposition, Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | | | | | - Jennifer Wong
- Department of Genetics, Institut Curie, Paris, France
- PSL Research University, Paris, France
| | - Mathilde Filser
- Department of Genetics, Institut Curie, Paris, France
- PSL Research University, Paris, France
| | - Dominique Stoppa-Lyonnet
- Department of Genetics, Institut Curie, Paris, France
- Inserm U830, Institut Curie, Paris, France
- Paris Cité University, Paris, France
| | - Franck Bourdeaut
- PSL Research University, Paris, France
- Inserm U830, Institut Curie, Paris, France
- SIREDO Oncology Center (Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer), Institut Curie, Paris, France
| | - Afane Brahimi
- Department of Clinical Genetics, Lille University Hospital, Lille, France
| | - Olivier Ingster
- Department of Genetics, Centre Angers University Hospital, Angers, France
| | | | - Sarah A. Jackson
- Department of Pediatrics, The University of Chicago, Chicago, Illinois
| | - Mark M. Sasaki
- Department of Pediatrics, The University of Chicago, Chicago, Illinois
| | - Trent Fowler
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
- Peel Therapeutics, Inc., Salt Lake City, Utah
| | - Anita Ng
- Karches Center for Oncology Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York
| | - Ryan J. Corbett
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Rebecca S. Kaufman
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | | | | | - Kuan-lin Huang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sharon J. Diskin
- Department of Biomedical and Health Informatics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jo Lynne Rokita
- Center for Data-Driven Discovery in Biomedicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, District of Columbia
| | - Hussam Al-Kateb
- Department of Lab Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Rose B. McGee
- Division of Cancer Predisposition, Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Joshua D. Schiffman
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
- Peel Therapeutics, Inc., Salt Lake City, Utah
- Department of Pediatrics, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Kenneth S. Chen
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Douglas R. Stewart
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - D. Williams Parsons
- Department of Pediatrics, Baylor College of Medicine, Houston Texas
- Texas Children’s Cancer and Hematology Center, Texas Children’s Hospital, Houston, Texas
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Sharon E. Plon
- Department of Pediatrics, Baylor College of Medicine, Houston Texas
- Texas Children’s Cancer and Hematology Center, Texas Children’s Hospital, Houston, Texas
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Kris Ann P. Schultz
- International Pleuropulmonary Blastoma/DICER1 Registry, Minneapolis, Minnesota
| | - Kenan Onel
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, New York
- Department of Clinical Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, New York
- Center for Precision Oncology and Cancer Prevention, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| |
Collapse
|
23
|
Wang JN, Yu DH, Li ZY, Kong LY, Li NH, Wu YX, Wang TQ, Wang ZF, Li ZQ. A Mendelian randomization study: causal relationship between immune cells and the risks of social phobia, specific phobia, and agoraphobia. BMC Psychiatry 2025; 25:350. [PMID: 40200188 PMCID: PMC11980060 DOI: 10.1186/s12888-025-06794-4] [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: 02/27/2024] [Accepted: 03/28/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Although phobia is a common psychiatric disorder, the underlying biological mechanisms have not been fully elucidated. Complex immune-brain interactions that affect neural development, survival, and function may have causal and therapeutic implications in psychiatric illnesses. In this study, the relationships between immune cell traits and phobia were analysed using Mendelian randomization to explore the biological mechanisms. METHODS Based on publicly-available genetic data, a two-sample MR analysis was used to determine the causal relationship between 731 immune cell traits and the risk of developing phobias. Sensitivity analyses were conducted to verify the robustness, heterogeneity, and horizontal pleiotropy of the results. RESULTS After forward and reverse analyses, false discovery rate (FDR) corrections were performed. No significant associations between phobias and immune cell traits were identified. After adjusting the FDR threshold, social phobia affected two immune cell traits: CD39 on granulocytes (β = 9.0347, 95% confidence interval (CI) = 4.4802-13.5891, P = 0.0001, FDR = 0.0738), and CD11c on granulocytes (β = 7.7976, 95% CI = 3.4616-12.1336, P = 0.0004, FDR = 0.1547). Three immune cell traits affected the risk of specific phobias: CD4 + CD8dim T cell %leukocyte (odds ratio (OR) = 0.9985, 95% CI = 0.9976-0.9993, P = 0.0006, FDR = 0.1373), CD45 on CD33 + HLA DR + CD14dim (OR = 0.9977, 95% CI = 0.9964-0.9990, P = 0.0004, FDR = 0.1373), and CD8 on CD28 + CD45RA + CD8br (OR = 0.9990, 95% CI = 0.9985-0.9996, P = 0.0003, FDR = 0.1373). Two immune cell traits affected the risk of agoraphobia: CD3 on CD39 + resting regulatory T cells (Tregs) (OR = 1.0010, 95 CI%=1.0005-1.0015, P = 0.0001, FDR = 0.0596) and HLA DR on CD33br HLA DR + CD14dim (OR = 0.9993, 95 CI%=0.9990-0.9997, P = 0.0002, FDR = 0.0596). CONCLUSIONS Immune cell traits closely related to phobias were screened out through genomics, which provides a reference for the subsequent research on the immune system-phobia interaction.
Collapse
Affiliation(s)
- Jun-Neng Wang
- Brain Glioma Center & Department of Neurosurgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
| | - Dong-Hu Yu
- Brain Glioma Center & Department of Neurosurgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
| | - Zhi-Yu Li
- Brain Glioma Center & Department of Neurosurgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
| | - Ling-Yue Kong
- Brain Glioma Center & Department of Neurosurgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
| | - Nan-Hao Li
- Brain Glioma Center & Department of Neurosurgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
| | - You-Xian Wu
- Brain Glioma Center & Department of Neurosurgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
| | - Tian-Qing Wang
- Brain Glioma Center & Department of Neurosurgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
| | - Ze-Fen Wang
- Department of Physiology, Wuhan University School of Basic Medical Sciences, 115 Donghu Road, Wuhan, 430071, China.
| | - Zhi-Qiang Li
- Brain Glioma Center & Department of Neurosurgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China.
| |
Collapse
|
24
|
Kerr SM, Klaric L, Muckian MD, Johnston K, Drake C, Halachev M, Cowan E, Snadden L, Dean J, Zheng SL, Thami PK, Ware JS, Tzoneva G, Shuldiner AR, Miedzybrodzka Z, Wilson JF. Actionable genetic variants in 4,198 Scottish participants from the Orkney and Shetland founder populations and implementation of return of results. Am J Hum Genet 2025; 112:793-807. [PMID: 40088892 DOI: 10.1016/j.ajhg.2025.02.018] [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: 11/01/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/17/2025] Open
Abstract
The benefits of returning clinically actionable genetic results to participants in research cohorts are accruing, yet such a genome-first approach is challenging. Here, we describe the implementation of return of such results in two founder populations from Scotland. Between 2005 and 2015, we recruited >4,000 adults with grandparents from Orkney and Shetland into the Viking Genes research cohort. The return of genetic data was not offered at baseline, but in 2023, we sent invitations to participants for consent to return of actionable genetic findings. We generated exome sequence data from 4,198 participants and used the American College of Medical Genetics and Genomics (ACMG) v.3.2 list of 81 genes, ClinVar review, and pathogenicity status, plus manual curation, to develop a pipeline to identify potentially actionable variants. We identified 104 individuals (2.5%) with 108 actionable genotypes at 39 variants in 23 genes and validated these. Working with the NHS Clinical Genetics service, which provided genetic counseling and clinical verification of the research results, and after expert clinical review, we notified 64 consenting participants (or their next of kin) of their actionable genotypes. Ten actionable variants across seven genes (BRCA1, BRCA2, ATP7B, TTN, KCNH2, MUTYH, and GAA) have risen 50- to >3,000-fold in frequency through genetic drift in ancestral island localities. Viking Genes is one of the first UK research cohorts to return actionable findings, providing an ethical and logistical exemplar of return of results. The genetic structure in the Northern Isles of Scotland with multiple founder effects provides a unique opportunity for a tailored approach to disease prevention through genetic screening.
Collapse
Affiliation(s)
- Shona M Kerr
- MRC Human Genetics Unit, University of Edinburgh, Institute of Genetics and Cancer, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Lucija Klaric
- MRC Human Genetics Unit, University of Edinburgh, Institute of Genetics and Cancer, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Marisa D Muckian
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Teviot Place, Edinburgh EH8 9AG, UK; Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Kiera Johnston
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Teviot Place, Edinburgh EH8 9AG, UK
| | - Camilla Drake
- MRC Human Genetics Unit, University of Edinburgh, Institute of Genetics and Cancer, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Mihail Halachev
- MRC Human Genetics Unit, University of Edinburgh, Institute of Genetics and Cancer, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Emma Cowan
- Department of Medical Genetics, Ashgrove House, NHS Grampian, Aberdeen AB25 2ZA, UK; Medical Genetics Group, University of Aberdeen, Polwarth Building, Aberdeen AB25 2ZD, UK
| | - Lesley Snadden
- Department of Medical Genetics, Ashgrove House, NHS Grampian, Aberdeen AB25 2ZA, UK; Medical Genetics Group, University of Aberdeen, Polwarth Building, Aberdeen AB25 2ZD, UK
| | - John Dean
- Medical Genetics Group, University of Aberdeen, Polwarth Building, Aberdeen AB25 2ZD, UK
| | - Sean L Zheng
- National Heart and Lung Institute, Imperial College London, London, UK; MRC Laboratory of Medical Sciences, Imperial College London, London, UK; Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Prisca K Thami
- National Heart and Lung Institute, Imperial College London, London, UK; MRC Laboratory of Medical Sciences, Imperial College London, London, UK
| | - James S Ware
- National Heart and Lung Institute, Imperial College London, London, UK; MRC Laboratory of Medical Sciences, Imperial College London, London, UK; Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | | | | | - Zosia Miedzybrodzka
- Department of Medical Genetics, Ashgrove House, NHS Grampian, Aberdeen AB25 2ZA, UK; Medical Genetics Group, University of Aberdeen, Polwarth Building, Aberdeen AB25 2ZD, UK
| | - James F Wilson
- MRC Human Genetics Unit, University of Edinburgh, Institute of Genetics and Cancer, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK; Centre for Global Health Research, Usher Institute, University of Edinburgh, Teviot Place, Edinburgh EH8 9AG, UK; Centre for Genomic and Experimental Medicine, University of Edinburgh, Institute of Genetics and Cancer, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK.
| |
Collapse
|
25
|
Wilcox N, Tyrer JP, Dennis J, Yang X, Perry JRB, Gardner EJ, Easton DF. The contribution of coding variants to the heritability of multiple cancer types using UK Biobank whole-exome sequencing data. Am J Hum Genet 2025; 112:903-912. [PMID: 40073867 DOI: 10.1016/j.ajhg.2025.02.013] [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: 09/27/2024] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Genome-wide association studies have been highly successful at identifying common variants associated with cancer; however, they do not explain all the inherited risks of cancer. Family-based studies, targeted sequencing, and, more recently, exome-wide association studies have identified rare coding variants in some genes associated with cancer risk, but the overall contribution of these variants to the heritability of cancer is less clear. Here, we describe a method to estimate the genome-wide contribution of rare coding variants to heritability that fits models to the burden effect sizes using an empirical Bayesian approach. We apply this method to the burden of protein-truncating variants in over 15,000 genes for 11 cancers in the UK Biobank using whole-exome sequencing data on over 400,000 individuals. We extend the method to consider the overlap of genes contributing to pairs of cancers. We found ovarian cancer to have the greatest proportion of heritability attributable to protein-truncating variants in genes (46%). The joint cancer models highlight significant clustering of cancer types, including a near-complete overlap in susceptibility genes for breast, ovarian, prostate, and pancreatic cancer. Our results provide insights into the contribution of rare coding variants to the heritability of cancer and identify additional genes with strong evidence of susceptibility to multiple cancer types.
Collapse
Affiliation(s)
- Naomi Wilcox
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
| | - Jonathan P Tyrer
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Joe Dennis
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Xin Yang
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - John R B Perry
- Metabolic Research Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK; MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Eugene J Gardner
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK; Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| |
Collapse
|
26
|
Du X, Mendez-Lara K, Hu S, Diao R, Bhavimani G, Hernandez R, Glass K, De Arruda Saldanha C, Flannick J, Heinz S, Majithia AR. An Alternatively Translated Isoform of PPARG Suggests AF-1 Domain Inhibition as an Insulin Sensitization Target. Diabetes 2025; 74:651-663. [PMID: 39854214 PMCID: PMC11926277 DOI: 10.2337/db24-0497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 01/21/2025] [Indexed: 01/26/2025]
Abstract
ARTICLE HIGHLIGHTS Genetic screens were performed across PPARG to study how disruptive mutations across the full coding sequence affect function. An alternative translational start site in PPARG generates a truncated isoform, peroxisome proliferator-activated receptor γ (PPARγ) M135, which lacks the N-terminal activation function 1 (AF-1) domain and shows increased agonist-induced transactivation of target genes. In human carriers of rare PPARG variants, AF-1 domain-disrupting genetic variants increase agonist-induced PPARγ activity and decrease metabolic syndrome severity. Targeting the AF-1 domain is a potential therapeutic strategy for insulin sensitization.
Collapse
Affiliation(s)
- Xiaomi Du
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
- Bioinformatics and Systems Biology Graduate Program, University of California San Diego, La Jolla, CA
| | - Karen Mendez-Lara
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Siqi Hu
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Rachel Diao
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Guru Bhavimani
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Ruben Hernandez
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Kimberly Glass
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Camila De Arruda Saldanha
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Jason Flannick
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Pediatrics, Boston Children’s Hospital, Boston, MA
| | - Sven Heinz
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Amit R. Majithia
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| |
Collapse
|
27
|
Zhang Y, Guo J, Zhang N. Mendelian randomization study investigating the relationship between nutrients and polycystic ovary syndrome. Int J Gynaecol Obstet 2025; 169:198-205. [PMID: 39569763 DOI: 10.1002/ijgo.16029] [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: 06/28/2024] [Revised: 10/22/2024] [Accepted: 10/29/2024] [Indexed: 11/22/2024]
Abstract
OBJECTIVE Polycystic ovary syndrome (PCOS) affects many women and is linked to nutrient intake, but the causal role of nutrients is unclear. Our study used Mendelian randomization (MR) to explore this relationship. METHODS We employed two-sample MR to investigate the causal effects of four macronutrients (carbohydrates, fats, sugars, proteins) and 12 micronutrients (vitamin A [retinol], folate, vitamin B6, vitamin B12, vitamin C [ascorbic acid], vitamin D, vitamin E [alpha-tocopherol], magnesium, calcium, iron, selenium, and zinc) on the risk of PCOS based on their circulating levels. We conducted analyses using the inverse variance-weighted method (IVW), MR-Egger regression, weighted median method, and simple model approach, with IVW as the primary method. To correct for multiple testing, the false discovery rate (FDR) method was applied. The reliability and potential bias of the results were assessed by sensitivity analysis and F-statistics. RESULTS From a genetic standpoint, higher circulating vitamin A (retinol) levels were found to be a protective factor for PCOS and closely associated with the risk of PCOS, consistent with recent research findings. The inverse variance-weighted (IVW) estimation indicated a protective effect of vitamin A (retinol) on PCOS (IVWOR = 0.97, 95% CI: 0.95-0.99, P = 0.002, FDR_P = 0.03). However, no causal relationship was found between other nutrients and PCOS. In addition, MR-PRESSO and MR-Egger regression showed that our results were not affected by horizontal pleiotropy (P > 0.05). Finally, leave-one-out sensitivity analysis confirmed the robustness of MR Results. CONCLUSION Our findings suggest that circulating vitamin A (retinol) levels influence the risk of PCOS, but further research is needed.
Collapse
Affiliation(s)
- Yujie Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Jiawen Guo
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Ning Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| |
Collapse
|
28
|
Wei J, Yu Y, Wu H, Li Y, Wang N, Tan X. Clonal Hematopoiesis of Indeterminate Potential and Risk of Microvascular Complications Among Individuals With Type 2 Diabetes: A Cohort Study. Diabetes 2025; 74:585-595. [PMID: 39804667 DOI: 10.2337/db24-0841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 01/08/2025] [Indexed: 03/22/2025]
Abstract
ARTICLE HIGHLIGHTS Clonal hematopoiesis of indeterminate potential (CHIP) is an age-related disorder that is associated with macrovascular diseases, such as coronary artery disease and stroke. However, the effects of CHIP on microvascular complication have not been explored in individuals with type 2 diabetes. We wanted to determine whether CHIP is associated with diabetic microvascular complications (DMCs). CHIP was associated with a high risk of DMCs, specifically, diabetic retinopathy and diabetic kidney disease, but not diabetic neuropathy. Gene-specific analyses suggested that some driver genes were associated with risk of developing DMCs. These findings indicated that CHIP may represent a novel risk factor for DMCs among individuals with type 2 diabetes, distinct from traditional risk factors, which may have implications for prevention and management of DMCs.
Collapse
Affiliation(s)
- Jiahe Wei
- Department of Big Data in Health Science, Zhejiang University School of Public Health and Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
| | - Yuefeng Yu
- Department of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hanzhang Wu
- Department of Big Data in Health Science, Zhejiang University School of Public Health and Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
| | - Yingjun Li
- Department of Epidemiology and Health Statistics, School of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ningjian Wang
- Department of Endocrinology and Metabolism, Institute of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Tan
- Department of Big Data in Health Science, Zhejiang University School of Public Health and Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
29
|
Zhao J, Xiong Y, Liu Y, Ling J, Liu S, Wang W. Endothelium Piezo1 deletion alleviates experimental varicose veins by attenuating perivenous inflammation. Mol Cell Biochem 2025; 480:2423-2435. [PMID: 39249600 DOI: 10.1007/s11010-024-05115-9] [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: 03/28/2024] [Accepted: 08/31/2024] [Indexed: 09/10/2024]
Abstract
Previous large-scale genetic studies have prioritized the causal genes piezo type mechanosensitive ion channel component 1 (PIEZO1) and castor zinc finger 1 (CASZ1) associated with varicose veins (VVs). This study aims to evaluate their roles in both clinical and experimental VVs. In this study, we investigated abundance of PIEZO1 and CASZ1 in both varicose and normal veins from the same patients. Yoda1 (a selective PIEZO1 agonist, 2.6 mg/kg/day) or vehicle was administered intraperitoneally for 3 weeks to evaluate the effect of PIEZO1 activation on experimental VVs. Subsequently, endothelial Piezo1 deletion mice (Piezo1iΔEC mice) were generated to explored the role of endothelial PIEZO1 on VVs. Laser speckle imaging, flow cytometry, cell tracing with Evans blue or rhodamine-6G, and histopathological staining were utilized to evaluate the pathophysiology of VVs. Our results showed that mRNA expression of PIEZO1, but not CASZ1, was abundant and increased in clinical VVs. The Piezo1tP1-td mice revealed endothelium-specific expression of PIEZO1 in mice veins. By establishing iliac vein ligation-induced VVs in mice, Yoda1 exacerbated experimental VVs with increased inflammatory cell infiltration. Subsequently, endothelial Piezo1 deletion (Piezo1iΔEC mice) alleviated experimental VVs and vascular remodeling by directly reducing vascular permeability and leukocyte-endothelium interactions compared to the control (Piezo1fl/fl mice). PIEZO1 is highly expressed in clinical VVs, meanwhile, activation or inhibition of PIEZO1 exerts a remarkable effect on experimental VVs. Furthermore, Piezo1 may constitute a potential therapeutic approach for the medical treatment of VVs.
Collapse
Affiliation(s)
- Jiani Zhao
- Department of General and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yacheng Xiong
- Department of General and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yu Liu
- Department of General and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jin Ling
- Department of General and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Shuai Liu
- Department of General and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Wei Wang
- Department of General and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
30
|
Lee DSM, Cardone KM, Zhang DY, Tsao NL, Abramowitz S, Sharma P, DePaolo JS, Conery M, Aragam KG, Biddinger K, Dilitikas O, Hoffman-Andrews L, Judy RL, Khan A, Kullo IJ, Puckelwartz MJ, Reza N, Satterfield BA, Singhal P, Arany Z, Cappola TP, Carruth ED, Day SM, Do R, Haggerty CM, Joseph J, McNally EM, Nadkarni G, Owens AT, Rader DJ, Ritchie MD, Sun YV, Voight BF, Levin MG, Damrauer SM. Common-variant and rare-variant genetic architecture of heart failure across the allele-frequency spectrum. Nat Genet 2025; 57:829-838. [PMID: 40195560 PMCID: PMC12049093 DOI: 10.1038/s41588-025-02140-2] [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: 04/25/2024] [Accepted: 02/21/2025] [Indexed: 04/09/2025]
Abstract
Heart failure is a complex trait, influenced by environmental and genetic factors, affecting over 30 million individuals worldwide. Here we report common-variant and rare-variant association studies of all-cause heart failure and examine how different classes of genetic variation impact its heritability. We identify 176 common-variant risk loci at genome-wide significance in 2,358,556 individuals and cluster these signals into five broad modules based on pleiotropic associations with anthropomorphic traits/obesity, blood pressure/renal function, atherosclerosis/lipids, immune activity and arrhythmias. In parallel, we uncover exome-wide significant associations for heart failure and rare predicted loss-of-function variants in TTN, MYBPC3, FLNC and BAG3 using exome sequencing of 376,334 individuals. We find that total burden heritability of rare coding variants is highly concentrated in a small set of Mendelian cardiomyopathy genes, while common-variant heritability is diffusely spread throughout the genome. Finally, we show that common-variant background modifies heart failure risk among carriers of rare pathogenic truncating variants in TTN. Together, these findings discern genetic links between dysregulated metabolism and heart failure and highlight a polygenic component to heart failure not captured by current clinical genetic testing.
Collapse
Affiliation(s)
- David S M Lee
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kathleen M Cardone
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - David Y Zhang
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Noah L Tsao
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sarah Abramowitz
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Pranav Sharma
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - John S DePaolo
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mitchell Conery
- Genomics and Computational Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Krishna G Aragam
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kiran Biddinger
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ozan Dilitikas
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Lily Hoffman-Andrews
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Renae L Judy
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Atlas Khan
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York City, NY, USA
| | - Iftikhar J Kullo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Megan J Puckelwartz
- Department of Pharmacology, Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Nosheen Reza
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Pankhuri Singhal
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Zoltan Arany
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Thomas P Cappola
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Eric D Carruth
- Department of Genomic Health, Geisinger, Danville, PA, USA
| | - Sharlene M Day
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ron Do
- The Charles Bronfman Institute for Personalized Medicine, Mount Sinai Icahn School of Medicine, New York City, NY, USA
- BioMe Phenomics Center, Mount Sinai Icahn School of Medicine, New York City, NY, USA
- Department of Genetics and Genomic Sciences, Mount Sinai Icahn School of Medicine, New York City, NY, USA
| | | | - Jacob Joseph
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Massachusetts Veterans Epidemiology Research and Information Center, VA Boston Healthcare System, Boston, MA, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Bluhm Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Girish Nadkarni
- Division of Nephrology, Department of Medicine, Mount Sinai Icahn School of Medicine, New York City, NY, USA
| | - Anjali T Owens
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel J Rader
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Translational Medicine and Human Genetics, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Marylyn D Ritchie
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yan V Sun
- Atlanta VA Health Care System, Decatur, GA, USA
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Benjamin F Voight
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael G Levin
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA.
| | - Scott M Damrauer
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| |
Collapse
|
31
|
Khan TG, Bragazzi Cunha J, Raut C, Burroughs M, Vyas HS, Leix K, Goonewardena SN, Smrcka AV, Speliotes EK, Emmer BT. Functional interrogation of cellular Lp(a) uptake by genome-scale CRISPR screening. Atherosclerosis 2025; 403:119174. [PMID: 40174266 PMCID: PMC12011201 DOI: 10.1016/j.atherosclerosis.2025.119174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 02/04/2025] [Accepted: 03/07/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND AND AIMS An elevated level of lipoprotein(a), or Lp(a), in the bloodstream has been causally linked to the development of atherosclerotic cardiovascular disease and calcific aortic valve stenosis. Steady state levels of circulating lipoproteins are modulated by their rate of clearance, but the identity of the Lp(a) uptake receptor(s) has been controversial. METHODS We performed a genome-scale CRISPR screen to functionally interrogate all potential Lp(a) uptake regulators in HuH7 cells. Screen validation was performed by single gene disruption and overexpression. Direct binding between purified lipoproteins and recombinant protein was tested using biolayer interferometry. An association between human genetic variants and circulating Lp(a) levels was analyzed in the UK Biobank cohort. RESULTS The top positive and negative regulators of Lp(a) uptake in our screen were LDLR and MYLIP, encoding the LDL receptor and its ubiquitin ligase IDOL, respectively. We also found a significant correlation for other genes with established roles in LDLR regulation. No other gene products, including those previously proposed as Lp(a) receptors, exhibited a significant effect on Lp(a) uptake in our screen. We validated the functional influence of LDLR expression on HuH7 Lp(a) uptake, confirmed in vitro binding between the LDLR extracellular domain and purified Lp(a), and detected an association between loss-of-function LDLR variants and increased circulating Lp(a) levels in the UK Biobank cohort. CONCLUSIONS Our findings support a central role for the LDL receptor in mediating Lp(a) uptake by hepatocytes.
Collapse
Affiliation(s)
- Taslima G Khan
- Program in Chemical Biology, University of Michigan, Ann Arbor, MI, USA
| | - Juliana Bragazzi Cunha
- Division of Hospital Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Chinmay Raut
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Michael Burroughs
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Hitarthi S Vyas
- Division of Hospital Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Kyle Leix
- Division of Hospital Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Sascha N Goonewardena
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, USA
| | - Alan V Smrcka
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Elizabeth K Speliotes
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Brian T Emmer
- Division of Hospital Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
32
|
Patin E, Quintana-Murci L. Tracing the Evolution of Human Immunity Through Ancient DNA. Annu Rev Immunol 2025; 43:57-82. [PMID: 39705165 DOI: 10.1146/annurev-immunol-082323-024638] [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: 12/22/2024]
Abstract
Infections have imposed strong selection pressures throughout human evolution, making the study of natural selection's effects on immunity genes highly complementary to disease-focused research. This review discusses how ancient DNA studies, which have revolutionized evolutionary genetics, increase our understanding of the evolution of human immunity. These studies have shown that interbreeding between modern humans and Neanderthals or Denisovans has influenced present-day immune responses, particularly to viruses. Additionally, ancient genomics enables the tracking of how human immunity has evolved across cultural transitions, highlighting strong selection since the Bronze Age in Europe (<4,500 years) and potential genetic adaptations to epidemics raging during the Middle Ages and the European colonization of the Americas. Furthermore, ancient genomic studies suggest that the genetic risk for noninfectious immune disorders has gradually increased over millennia because alleles associated with increased risk for autoimmunity and inflammation once conferred resistance to infections. The challenge now is to extend these findings to diverse, non-European populations and to provide a more global understanding of the evolution of human immunity.
Collapse
Affiliation(s)
- Etienne Patin
- Institut Pasteur, Université Paris Cité, CNRS UMR 2000, Human Evolutionary Genetics Unit, Paris, France;
| | - Lluis Quintana-Murci
- Human Genomics and Evolution, Collège de France, Paris, France
- Institut Pasteur, Université Paris Cité, CNRS UMR 2000, Human Evolutionary Genetics Unit, Paris, France;
| |
Collapse
|
33
|
Yang H, Jin C, Li J, Zhang Z, Zhao K, Yin X, Wang Z, Zhu G, Yan X, Jiang Z, Qi Y, Ma X, Wang K. Causal relationship between bladder cancer and gut microbiota contributes to the gut-bladder axis: A two-sample Mendelian randomization study. Urol Oncol 2025; 43:267.e9-267.e18. [PMID: 39489648 DOI: 10.1016/j.urolonc.2024.10.014] [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: 05/05/2024] [Revised: 09/22/2024] [Accepted: 10/14/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Recent studies have underscored a potential link between gut microbiota and urological tumors, yet the causal relationship with bladder cancer (BCa) and the role of metabolic pathways remain unclear. METHODS Instrumental variables (IVs) for gut microbiota were obtained from genome-wide association studies (GWAS) conducted by the MiBioGen consortium (n = 18,340). GWAS data for BCa were sourced from a comprehensive genome-wide meta-analysis encompassing 23 cohorts. Mendelian randomization (MR) was employed to investigate the causal relationship between gut microbiota and BCa, utilizing inverse variance weighted (IVW) as the primary MR method. Additionally, metabolic pathways associated with these microbiota were analyzed to understand their functional roles in BCa pathogenesis. Sensitivity analyses were conducted to validate all MR results. RESULTS The MR analysis identified five gut microbiota taxa with a causal association with BCa, with the genus Bilophila notably promoting BCa. Metabolic pathway analysis revealed significant associations between specific pathways and BCa, suggesting that changes in amino acid and NAD metabolism might influence BCa development. Sensitivity analyses indicated no significant heterogeneity or horizontal pleiotropy among the IVs. CONCLUSION This study revealed the significant causal relationship between gut microbiota and BCa, particularly identifying Bilophila as a key pathogenic initiator. These findings elucidated the potential impact of metabolic pathways, especially amino acid and NAD metabolism, on the pathogenesis of BCa. They not only laid the foundation for innovative therapeutic strategies but also highlighted the immense potential of microbiota-based interventions in the prevention and treatment of BCa, paving the way for new directions in precision medicine.
Collapse
Affiliation(s)
- Han Yang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chen Jin
- Department of Pediatric Surgery, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jie Li
- Department of Oncology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, China
| | - Zongliang Zhang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Kai Zhao
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xinbao Yin
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhenlin Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guanqun Zhu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xuechuan Yan
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zaiqing Jiang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yixin Qi
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xuezhen Ma
- Department of Oncology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao, China.
| | - Ke Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
34
|
Loughrey PB, Mothojakan NB, Iacovazzo D, Arni A, Aflorei ED, Arnaldi G, Barlier A, Beckers A, Bizzi MF, Chanson P, Dal J, Daly AF, Dang MN, David A, Andrade MDO, Else T, Elston MS, Evans A, Ferrau F, Fica S, Flanagan D, Gadelha MR, Grossman AB, Kapur S, Khoo B, Kumar AV, Kumar-Sinha C, Lechan RM, Ludman M, Metherell LA, Miljic D, Mourougavelou V, Musat M, Occhi G, Owens M, Pascanu I, Pinheiro SVB, Radian S, Ribeiro-Oliveira A, Schöfl C, Patel KA, Hernández-Ramírez LC, Korbonits M. Reassessing the role of the p.(Arg304Gln) missense AIP variant in pituitary tumorigenesis. Eur J Endocrinol 2025; 192:385-397. [PMID: 40070360 PMCID: PMC11962913 DOI: 10.1093/ejendo/lvaf044] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/02/2024] [Accepted: 03/10/2025] [Indexed: 04/03/2025]
Abstract
OBJECTIVE Heterozygous germline loss-of-function variants in AIP are associated with young-onset growth hormone and/or prolactin-secreting pituitary tumours. However, the pathogenic role of the c.911G > A; p.(Arg304Gln) (R304Q) AIP variant has been controversial. Recent data from public exome/genome databases show this variant is not infrequent. The objective of this work was to reassess the pathogenicity of R304Q based on clinical, genomic, and functional assay data. DESIGN Data were collected on published R304Q pituitary neuroendocrine tumour cases and from International Familial Isolated Pituitary Adenoma Consortium R304Q cases (n = 38, R304Q cohort). Clinical features, population cohort frequency, computational analyses, prediction models, presence of loss-of-heterozygosity, and in vitro/in vivo functional studies were assessed and compared with data from pathogenic/likely pathogenic AIP variant patients (AIPmut cohort, n = 184). RESULTS Of 38 R304Q patients, 61% (23/38) had growth hormone excess, in contrast to 80% of AIPmut cohort (147/184, P < .001). R304Q cohort was older at disease onset and diagnosis than the AIPmut cohort (median [quartiles] onset: 25 y [16-35] vs 16 y [14-23], P < .001; median [quartiles] diagnosis: 36 y [24-44] vs 21 y [15-29], P < .001). R304Q is present in gnomADv2.1 (0.31%) and UK Biobank (0.16%), including three persons with homozygous R304Q. No loss-of-heterozygosity was detected in four R304Q pituitary neuroendocrine tumour samples. In silico predictions and experimental data were conflicting. CONCLUSIONS Evidence suggests that R304Q is not pathogenic for pituitary neuroendocrine tumour. We recommend changing this variant classification to likely benign and do not recommend pre-symptomatic genetic testing of family members or follow-up of already identified unaffected individuals with the R304Q variant.
Collapse
Affiliation(s)
- Paul Benjamin Loughrey
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7AE, United Kingdom
| | - Nadira B Mothojakan
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Donato Iacovazzo
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Ankit Arni
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, EX1 2HZ, United Kingdom
| | - Elena D Aflorei
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Giorgio Arnaldi
- Section of Endocrinology, PROMISE, University of Palermo, Palermo 90127, Italy
- Unità Operativa Complessa of Endocrine Diseases, A.O.U.P. Paolo Giaccone of Palermo, Palermo 90127, Italy
| | - Anne Barlier
- Aix Marseille Univ APHM, INSERM, UMR1251 MMG, Laboratory of Molecular Biology GEnOPé, Biogénopôle, Hôpital de la Timone, Marseille 13385, France
| | - Albert Beckers
- Department of Endocrinology, Centre Hospitalier Universitaire de Liège, University of Liège, Liège 4000, Belgium
| | - Mariana F Bizzi
- Department of Internal Medicine, Federal University of Minas Gerais, Belo Horizonte/Minas Gerais, 30130-100, Brazil
| | - Philippe Chanson
- Université Paris-Saclay, Inserm, Physiologie et Physiopathologie Endocriniennes, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Service d’Endocrinologie et des Maladies de la Reproduction, Centre de Référence des Maladies Rares de l’Hypophyse HYPO, Le Kremlin-Bicêtre, 94275, France
| | - Jakob Dal
- Department of Endocrinology, Aalborg University Hospital, Aalborg 9000, Denmark
| | - Adrian F Daly
- Department of Endocrinology, Centre Hospitalier Universitaire de Liège, University of Liège, Liège 4000, Belgium
| | - Mary N Dang
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Alessia David
- Centre for Bioinformatics, Department of Life Sciences, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Matheus de Oliveira Andrade
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
- Faculty of Medicine, University of Brasilia, Brasilia 70910-900, Brazil
| | - Tobias Else
- MEND/Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI 48109, United States
| | - Marianne S Elston
- Waikato Clinical Campus, The University of Auckland, Hamilton 3216, New Zealand
| | - Amy Evans
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Francesco Ferrau
- Department of Human Pathology of Adulthood and Childhood ‘G. Barresi’, University of Messina, Messina 98125, Italy
| | - Simona Fica
- Endocrinology and Diabetes Department, Elias Hospital, University of Medicine and Pharmacy Carol Davila Bucharest, Bucharest 011461, Romania
| | - Daniel Flanagan
- Department of Endocrinology, University Hospitals Plymouth NHS Trust, Plymouth, PL6 8DH, United Kingdom
| | - Monica R Gadelha
- Endocrinology Unit, Department of Internal Medicine, Universidade Federal do Rio de Janeiro, Rio de Janeiro, 21941-853, Brazil
| | - Ashley B Grossman
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Sonal Kapur
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Bernard Khoo
- Division of Medicine, University College London, Royal Free Campus, London, NW3 2PS, United Kingdom
| | - Ajith V Kumar
- North East Thames Regional Genetics Service, Great Ormond Street Hospital for Children, London, WC1N 3BH, United Kingdom
| | - Chandan Kumar-Sinha
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109-0940, United States
| | - Ronald M Lechan
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, MA 02111, United States
| | - Mark Ludman
- Institute of Genetics, Meir Medical Center, Kfar Saba, 4428164, Israel
| | - Louise A Metherell
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Dragana Miljic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Center of Serbia, Belgrade 11000, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Vishnou Mourougavelou
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Madalina Musat
- National Institute of Endocrinology, University of Medicine and Pharmacy Carol Davila Bucharest, Bucharest 050474, Romania
| | - Gianluca Occhi
- Department of Biology, University of Padua, Padua 35128, Italy
| | - Martina Owens
- Exeter Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, EX2 5DW, United Kingdom
| | - Ionela Pascanu
- Department of Endocrinology, George Emil Palade University of Medicine Pharmacy Science and Technology of Targu Mures, Targu Mures 540139, Romania
| | - Sergio V B Pinheiro
- Department of Pediatrics, Federal University of Minas Gerais, Belo Horizonte/Minas Gerais 30130-100, Brazil
| | - Serban Radian
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| | - Antonio Ribeiro-Oliveira
- Department of Internal Medicine, Federal University of Minas Gerais, Belo Horizonte/Minas Gerais, 30130-100, Brazil
| | - Christof Schöfl
- Center of Endocrinology and Metabolism, Bamberg and Erlangen, Obstmarkt 1, Bamberg 96047, Germany
| | - Kashyap A Patel
- Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, EX1 2HZ, United Kingdom
| | - Laura C Hernández-Ramírez
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
- Red de Apoyo a la Investigación, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City 14080, Mexico
| | - Márta Korbonits
- Centre for Endocrinology, Barts and The London School of Medicine, Queen Mary University of London, London, EC1M 6BQ, United Kingdom
| |
Collapse
|
35
|
Lawson LP, Parameswaran S, Panganiban RA, Constantine GM, Weirauch MT, Kottyan LC. Update on the genetics of allergic diseases. J Allergy Clin Immunol 2025:S0091-6749(25)00327-6. [PMID: 40139464 DOI: 10.1016/j.jaci.2025.03.012] [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: 10/21/2024] [Revised: 02/24/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025]
Abstract
The field of genetic etiology of allergic diseases has advanced significantly in recent years. Shared risk loci reflect the contribution of genetic factors to the sequential development of allergic conditions across the atopic march, while unique risk loci provide opportunities to understand tissue specific manifestations of allergic disease. Most identified risk variants are noncoding, indicating that they likely influence gene expression through gene regulatory mechanisms. Despite recent advances, challenges persist, particularly regarding the need for increased ancestral diversity in research populations. Further, while polygenic risk scores show promise for identifying individuals at higher genetic risk for allergic diseases, their predictive accuracy varies across different ancestries and can be difficult to translate to an individual's absolute risk of developing a disease. Methodologies, including "nearest gene," 3D chromatin interaction analysis, expression quantitative trait locus analysis, experimental screens, and integrative bioinformatic models, have established connections between genetic variants and their regulatory targets, enhancing our understanding of disease risk and phenotypic variability. In this review, we focus on the state of knowledge of allergic sensitization and 5 allergic diseases: asthma, atopic dermatitis, allergic rhinitis, food allergy, and eosinophilic esophagitis. We summarize recent progress and highlight opportunities for advancing our understanding of their genetic etiology.
Collapse
Affiliation(s)
- Lucinda P Lawson
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Sreeja Parameswaran
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Ronald A Panganiban
- Asthma Research, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Gregory M Constantine
- Human Eosinophil Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, Md
| | - Matthew T Weirauch
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Leah C Kottyan
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| |
Collapse
|
36
|
Georgakis MK, Malik R, Bounkari OE, Hasbani NR, Li J, Huffman JE, Shakt G, Tack RWP, Kimball TN, Asare Y, Morrison AC, Tsao NL, Judy R, Mitchell BD, Xu H, Montasser ME, Do R, Kenny EE, Loos RJF, Terry JG, Carr JJ, Bis JC, Psaty BM, Longstreth WT, Young KA, Lutz SM, Cho MH, Broome J, Khan AT, Wang FF, Heard-Costa N, Seshadri S, Vasan RS, Palmer ND, Freedman BI, Bowden DW, Yanek LR, Kral BG, Becker LC, Peyser PA, Bielak LF, Ammous F, Carson AP, Hall ME, Raffield LM, Rich SS, Post WS, Tracy RP, Taylor KD, Guo X, Mahaney MC, Curran JE, Blangero J, Clarke SL, Haessler JW, Hu Y, Assimes TL, Kooperberg C, Bernhagen J, Anderson CD, Damrauer SM, Zand R, Rotter JI, de Vries PS, Dichgans M. Rare damaging CCR2 variants are associated with lower lifetime cardiovascular risk. Genome Med 2025; 17:27. [PMID: 40119478 PMCID: PMC11929344 DOI: 10.1186/s13073-025-01456-2] [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: 06/27/2024] [Accepted: 03/11/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND Previous work has shown a role of CCL2, a key chemokine governing monocyte trafficking, in atherosclerosis. However, it remains unknown whether targeting CCR2, the cognate receptor of CCL2, provides protection against human atherosclerotic cardiovascular disease. METHODS Computationally predicted damaging or loss-of-function (REVEL > 0.5) variants within CCR2 were detected in whole-exome-sequencing data from 454,775 UK Biobank participants and tested for association with cardiovascular endpoints in gene-burden tests. Given the key role of CCR2 in monocyte mobilization, variants associated with lower monocyte count were prioritized for experimental validation. The response to CCL2 of human cells transfected with these variants was tested in migration and cAMP assays. Validated damaging variants were tested for association with cardiovascular endpoints, atherosclerosis burden, and vascular risk factors. Significant associations were replicated in six independent datasets (n = 1,062,595). RESULTS Carriers of 45 predicted damaging or loss-of-function CCR2 variants (n = 787 individuals) were at lower risk of myocardial infarction and coronary artery disease. One of these variants (M249K, n = 585, 0.15% of European ancestry individuals) was associated with lower monocyte count and with both decreased downstream signaling and chemoattraction in response to CCL2. While M249K showed no association with conventional vascular risk factors, it was consistently associated with a lower risk of myocardial infarction (odds ratio [OR]: 0.66, 95% confidence interval [CI]: 0.54-0.81, p = 6.1 × 10-5) and coronary artery disease (OR: 0.74, 95%CI: 0.63-0.87, p = 2.9 × 10-4) in the UK Biobank and in six replication cohorts. In a phenome-wide association study, there was no evidence of a higher risk of infections among M249K carriers. CONCLUSIONS Carriers of an experimentally confirmed damaging CCR2 variant are at a lower lifetime risk of myocardial infarction and coronary artery disease without carrying a higher risk of infections. Our findings provide genetic support for the translational potential of CCR2-targeting as an atheroprotective approach.
Collapse
Affiliation(s)
- Marios K Georgakis
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University (LMU), Feodor-Lynen-Str. 17, 81377, Munich, Germany.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| | - Rainer Malik
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University (LMU), Feodor-Lynen-Str. 17, 81377, Munich, Germany
| | - Omar El Bounkari
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University (LMU), Feodor-Lynen-Str. 17, 81377, Munich, Germany
| | - Natalie R Hasbani
- Human Genetics Center, Department of Epidemiology, School of Public Health, Human Genetics, and Environmental Sciences, the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jiang Li
- Department of Molecular and Functional Genomics, Geisinger Health System, Danville, PA, USA
| | | | - Gabrielle Shakt
- Department of Surgery, Perelman School of Medicineat Theaq , University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Reinier W P Tack
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Tamara N Kimball
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Yaw Asare
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University (LMU), Feodor-Lynen-Str. 17, 81377, Munich, Germany
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, School of Public Health, Human Genetics, and Environmental Sciences, the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Noah L Tsao
- Department of Surgery, Perelman School of Medicineat Theaq , University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Renae Judy
- Department of Surgery, Perelman School of Medicineat Theaq , University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Braxton D Mitchell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Geriatrics Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - Huichun Xu
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - May E Montasser
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ron Do
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eimear E Kenny
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Center for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James G Terry
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John Jeffrey Carr
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joshua C Bis
- Department of Medicine, Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
| | - Bruce M Psaty
- Department of Medicine, Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - W T Longstreth
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Kendra A Young
- Department of Epidemiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sharon M Lutz
- Department of Population Medicine, PRecisiOn Medicine Translational Research (PROMoTeR) Center, Harvard Pilgrim Health Careand, Harvard Medical School , Boston, MA, USA
- Department of Biostatistics, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jai Broome
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Alyna T Khan
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Fei Fei Wang
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Nancy Heard-Costa
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- Boston University and National Heart, Blood Institute'S Framingham Heart Study, Lung, and Framingham, MA, USA
| | - Sudha Seshadri
- Bigg'S Institute for Alzheimer'S Disease and Neurodegenerative Disorders, University of Texas Health Science Center, San Antonio, TX, USA
| | - Ramachandran S Vasan
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- Boston University and National Heart, Blood Institute'S Framingham Heart Study, Lung, and Framingham, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Nicholette D Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Barry I Freedman
- Section on Nephrology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Donald W Bowden
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Lisa R Yanek
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brian G Kral
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lewis C Becker
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Patricia A Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Lawrence F Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Farah Ammous
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - April P Carson
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Michael E Hall
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Wendy S Post
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Russel P Tracy
- Departments of Pathology & Laboratory Medicine, and Biochemistry, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Michael C Mahaney
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - Joanne E Curran
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - John Blangero
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - Shoa L Clarke
- Department of Medicine (Division of Cardiovascular Medicine), Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford, CA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Jeffrey W Haessler
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Yao Hu
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Themistocles L Assimes
- Department of Medicine (Division of Cardiovascular Medicine), Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford, CA, USA
- VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Jürgen Bernhagen
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University (LMU), Feodor-Lynen-Str. 17, 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Centre for Cardiovascular Research (DZHK, Munich), Partner Site Munich Heart Alliance, Munich, Germany
| | - Christopher D Anderson
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Scott M Damrauer
- Department of Surgery, Perelman School of Medicineat Theaq , University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicineat the , University of Pennsylvania, Philadelphia, PA, USA
| | - Ramin Zand
- Department of Neurology, Pennsylvania State University, Hershey, PA, USA
- Department of Neurology, Neuroscience Institute, Geisinger Health System, Danville, PA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, School of Public Health, Human Genetics, and Environmental Sciences, the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-University (LMU), Feodor-Lynen-Str. 17, 81377, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
- German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany.
- German Centre for Cardiovascular Research (DZHK, Munich), Partner Site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
37
|
Blair DR, Risch N. Reduced Penetrance is Common Among Predicted Loss-of-Function Variants and is Likely Driven by Residual Allelic Activity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.09.23.24314008. [PMID: 39399029 PMCID: PMC11469360 DOI: 10.1101/2024.09.23.24314008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Loss-of-function genetic variants (LoFs) often result in severe phenotypes, including autosomal dominant diseases driven by haploinsufficiency. Due to low carrier frequencies, their penetrance is generally unknown but typically variable. Here, we investigate the penetrance of >6,000 predicted LoFs (pLoFs) linked to 91 haploinsufficient diseases using a cohort of ≈24,000 carriers with linked electronic health record data. We find evidence for widespread reduced penetrance, which persisted after accounting for variant annotation artifacts, missed diagnoses, and incomplete clinical data. We thus hypothesized that many pLoFs have incomplete penetrance, which may be driven by residual allelic activity. To test this, we trained machine learning models to predict pLoF penetrance using variant-specific genomic features that may correlate with incomplete loss-of-function. The models were predictive of pLoF penetrance across a range of diseases and variant types, including those with prior clinical evidence for pathogenicity. This suggests that many pLoFs have incomplete penetrance due to residual allelic activity, complicating disease prognostication in asymptomatic carriers.
Collapse
|
38
|
Prizment A, Standafer A, Qu C, Beutel KM, Wang S, Huang WY, Lindblom A, Pearlman R, Van Guelpen B, Wolk A, Buchanan DD, Grant RC, Schmit SL, Platz EA, Joshu CE, Couper DJ, Peters U, Starr TK, Scott P, Pankratz N. Functional variants in the cystic fibrosis transmembrane conductance regulator (CFTR) gene are associated with increased risk of colorectal cancer. Hum Mol Genet 2025; 34:617-625. [PMID: 39825500 PMCID: PMC11924186 DOI: 10.1093/hmg/ddaf007] [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: 07/06/2024] [Revised: 12/17/2024] [Accepted: 01/14/2025] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Individuals with cystic fibrosis (CF; a recessive disorder) have an increased risk of colorectal cancer (CRC). Evidence suggests individuals with a single CFTR variant may also have increased CRC risk. METHODS Using population-based studies (GECCO, CORECT, CCFR, and ARIC; 53 785 CRC cases and 58 010 controls), we tested for an association between the most common CFTR variant (Phe508del) and CRC risk. For replication, we used whole exome sequencing data from UK Biobank (UKB; 5126 cases and 20 504 controls matched 4:1 based on genetic distance, age, and sex), and extended our analyses to all other heterozygous CFTR variants annotated as CF-causing. RESULTS In our meta-analysis of GECCO-CORECT-CCFR-ARIC, the odds ratio (OR) for CRC risk associated with Phe508del was 1.11 (P = 0.010). In our UKB replication, the OR for CRC risk associated with Phe508del was 1.28 (P = 0.002). The sequencing data from UKB also revealed an association between the presence of any other single CF-causing variant (excluding Phe508del) and CRC risk (OR = 1.33; P = 0.030). When stratifying CFTR variants by functional class, class I variants (no protein produced) had a stronger association (OR = 1.77; p = 0.002), while class II variants (misfolding and retention of the protein in the endoplasmic reticulum) other than Phe508del (OR = 1.75; p = 0.107) had similar effect size as Phe508del, and variants in classes III-VI had non-significant ORs less than 1.0 and/or were not present in cases. CONCLUSIONS CF-causing heterozygous variants, especially class I variants, are associated with a modest but statistically significant increased CRC risk. More research is needed to explain the biology underlying these associations.
Collapse
Affiliation(s)
- Anna Prizment
- Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
- Masonic Cancer Center, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Abby Standafer
- Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Conghui Qu
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, 98019, USA
| | - Kathleen M Beutel
- Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Shuo Wang
- Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Wen-Yi Huang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, MSC 9776, Bethesda, MD, 20892, USA
| | - Annika Lindblom
- Department of Molecular Medicine and Surgery, Karolinska Institutet, K1 Molekylär medicin och kirurgi, K1 MMK Klinisk genetik, 171 76 Stockholm, Sweden
- Department of Clinical Genetics, Karolinska University Hospital, Eugeniavägen 3, 171 64 Solna, Sweden
| | - Rachel Pearlman
- Department of Internal Medicine, Division of Human Genetics, The Ohio State University Comprehensive Cancer Center, 2012 Kenny Rd, Columbus, OH, 43221, USA
| | - Bethany Van Guelpen
- Department of Radiation Sciences, Oncology Unit, 27C, Målpunkt QC11, NUS, Norrlands universitetssjukhus, Umeå University, 901 85 Umeå, Sweden
| | - Alicja Wolk
- Institute of Environmental Medicine, Karolinska Institutet, C6 Institutet för miljömedicin, C6 CVD-NUT-EPI Wolk, 171 77 Stockholm, Sweden
| | - Daniel D Buchanan
- Department of Clinical Pathology, University of Melbourne Center for Cancer Research, University of Melbourne, 305 Grattan Street, Melbourne, Victoria, 3010, Australia
| | - Robert C Grant
- UHN-Princess Margaret Cancer Centre, University of Toronto, 7-811 700 University Ave, Toronto, Ontario, M5G 1X6, Canada
| | - Stephanie L Schmit
- Genomic Medicine Institute, Cleveland Clinic, 9500 Euclid Avenue, Mail Code NE50, Cleveland, OH, 44195, USA
- Population and Cancer Prevention Program, Case Comprehensive Cancer Center, 10900 Euclid Ave, Cleveland, OH, 44106, USA
| | - Elizabeth A Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe Street, Baltimore, MD, 21205, USA
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, 615 N Wolfe Street, Baltimore, MD, 21205, USA
| | - Corinne E Joshu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe Street, Baltimore, MD, 21205, USA
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, 615 N Wolfe Street, Baltimore, MD, 21205, USA
| | - David J Couper
- Department of Biostatistics, University of North Carolina at Chapel Hill, 123 W Franklin Street, Suite 450, CB #8030, Chapel Hill, NC, 27516, USA
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, 98019, USA
| | - Timothy K Starr
- Masonic Cancer Center, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
- Department of Obstetrics and Gynecology, Medical School, University of Minnesota, 515 Delaware St SE, Minneapolis, MN, 55455, USA
| | - Patricia Scott
- Masonic Cancer Center, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
- Department of Biomedical Sciences, University of Minnesota Medical School Duluth, 1035 University Drive, Duluth, MN, 55812, USA
| | - Nathan Pankratz
- Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| |
Collapse
|
39
|
Mehra R, Patra V, Dhillan R, Cvnm D, Singh H, Gupta L, Rastogi G, Sharma I, Sharma V. Interactive and evolutionary effect of CASZ1 gene variants on varicose veins susceptibility in South Asian Indians. Biol Res 2025; 58:17. [PMID: 40108671 PMCID: PMC11921479 DOI: 10.1186/s40659-025-00599-1] [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: 11/18/2024] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Varicose veins (VV) are spectrum of common vascular diseases having complex genetic etiology. The Castor Zinc Finger 1 (CASZ1) gene has been involved in vascular development and its variant has shown association with VV in various ethnicities, but CASZ1 susceptibility to VV risk is unexplored in the South Asian Indian population. The objective of this study was to estimate the association of the CASZ1 gene variations and VV susceptibility in the South Asian Indians, and to examine the evolutionary patterns of these variants compared to other populations. METHODOLOGY Population based case control analysis was conducted on all CASZ1 variants present in the Global Screening Array, including the established VV variant rs11121615 with a focus on validating and identification of both novel and established genetic markers to capture a full spectrum of population-specific genetic markers unique to studied population group. Linkage disequilibrium patterns and cumulative variant effects were also analyzed, followed by selection pressure assessment using neutrality tests. RESULTS Three CASZ1 variants rs72860191 (OR 1.58, 95% CI 1.07-2.32, p = 0.01), rs7519604 (OR 1.43, 95% CI 1.05-1.94, p = 0.01), and rs11121615 (OR 0.69, 95% CI 0.50-0.95, p = 0.02) were observed to be significantly associated with VV. Haplotype analysis identified unique haplotype structure of South Asian Indians compared to other global populations. Moreover, the cumulative OR was observed to be higher than the independently estimated values (OR = 2.41, 95% CI 1.48-3.94), indicating genotypic epistasis of VV associated variants. The neutrality tests revealed balancing selection within CASZ1 in the studied population compared to other populations, CONCLUSION: The present study identified CASZ1 variants and their epistatic interactions is associated with VV susceptibility supported with evidence of balancing selection, provides crucial insights into the genetic architecture of VV in studied group, highlighting the impact of evolutionary forces on disease susceptibility.
Collapse
Affiliation(s)
- Rohit Mehra
- Department of Vascular and Endovascular Surgery, Command Hospital (Southern Command), Pune, India.
| | - Vikram Patra
- Department of Vascular and Endovascular Surgery, 92-Base Hospital, Jammu, Kashmir, India
| | - Rishi Dhillan
- Department of Vascular and Endovascular Surgery, Army Hospital (Research and Referral), New Delhi, India
| | - Dattatraya Cvnm
- Department of Vascular and Endovascular Surgery, Army Hospital (Research and Referral), New Delhi, India
| | | | - Love Gupta
- NMC Genetics India Pvt. Ltd, Gurugram, Haryana, India
| | | | - Indu Sharma
- Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Varun Sharma
- NMC Genetics India Pvt. Ltd, Gurugram, Haryana, India.
- Head Research and Development Division, NMC Genetics India Pvt. Ltd. Gurugram, Gurugram, India.
| |
Collapse
|
40
|
Wang G, Yi X. Hyperopia may exert a protective effect against senile cataracts: Evidence from a Mendelian randomization study. Medicine (Baltimore) 2025; 104:e41794. [PMID: 40101038 PMCID: PMC11922449 DOI: 10.1097/md.0000000000041794] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 02/19/2025] [Indexed: 03/20/2025] Open
Abstract
Myopia has been extensively documented as a significant risk factor for cataracts; however, the relationship between hyperopia and cataract development remains poorly understood. Given the distinct refractive profiles of myopia and hyperopia, hyperopia may confer a protective effect against cataracts. In this study, we employed Mendelian randomization (MR) to examine the causal association between hyperopia and cataracts. A 2-sample MR framework was utilized to examine the causal relationship between hyperopia and cataracts, with multivariable MR implemented to account for potential confounding variables. The inverse variance weighted (IVW) method served as the primary analytical tool, complemented by multiple sensitivity analyses to ensure the robustness and reliability of the findings. Enrichment analyses were conducted to elucidate the underlying biological pathways, while Bayesian colocalization analysis pinpointed shared genetic loci that influence both hyperopia and cataracts. In our study, we found that hyperopia may exert a protective effect against cataracts (IVW odds ratio, 0.920 [95% confidence interval, 0.872-0.972]; P = .0029) and cataract surgery (IVW odds ratio, 0.811 [95% confidence interval, 0.754-0.873]; P < .0001). Multivariable MR, adjusting for confounding factors such as smoking, glaucoma, and diabetes, confirmed hyperopia's protective association with cataracts. Bayesian colocalization identified rs12193446 as a high-probability shared causal variant, while enrichment analyses revealed potential biological mechanisms linking hyperopia to cataract development. Genetic evidence suggests that higher levels of hyperopia are associated with a reduced risk of age-related nuclear cataracts, cataract extraction, and lens implants. Given the opposite refractive states of myopia and hyperopia and their opposite effects on cataracts, these findings provide new insights into the pathogenesis of age-related cataracts.
Collapse
Affiliation(s)
- Guoqing Wang
- Department of Ophthalmology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Xinjiang Medical University, Urumqi, China
| | - Xianglong Yi
- Department of Ophthalmology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Xinjiang Medical University, Urumqi, China
| |
Collapse
|
41
|
Bech Vilaseca Q, Vad OB, Paludan-Müller C, Andreasen L, Olesen MS, Svendsen JH, Lundegaard PR. Prevalence of deleterious cardiomyopathy variants in early-onset atrial fibrillation. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.12.25323872. [PMID: 40162242 PMCID: PMC11952602 DOI: 10.1101/2025.03.12.25323872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Background Atrial fibrillation (AF) is a common cardiac arrhythmia associated with an increased risk of stroke, heart failure, and death. Recent studies suggests that individuals with early onset of AF could be at increased risk of developing heart failure and dilated cardiomyopathy. This study aimed to identifying genetic variants in a broad panel of cardiomyopathy genes among early-onset AF individuals. Methods We conducted targeted genetic sequencing of 29 cardiomyopathy-associated genes in 478 individuals with AF onset below 45 years of age from a Danish cohort. Additionally, we analyzed whole exome sequencing data in 374,289 individuals from the UK Biobank, including 29,108 individuals with AF. The cohort was stratified by age at AF diagnosis, and individuals with pre-existing cardiomyopathy were excluded. We focused on rare, truncating variants predicted to lead to loss of function, and potentially deleterious missense variants in the UK Biobank. Results In the Danish cohort, 42 (8.8%) individuals with early-onset AF had truncating genetic variants in known cardiomyopathy genes. The UK Biobank analysis showed an inverse dose-response-like relationship between age of AF onset and prevalence of truncating variants, ranging from 3.8% in the AF onset <45 years group to 1.4% in the group without AF diagnosis. The prevalence of rare missense variants showed a similar pattern. Conclusions We identified a high prevalence of deleterious variants in cardiomyopathy-associated genes among individuals with early-onset AF. This supports recent guideline suggestions and indicates that genetic testing and surveillance for cardiomyopathy could be relevant in selected individuals with an early AF diagnosis.
Collapse
|
42
|
Deng N, Agila R, He Q, You C, Zheng S. Comprehensive causal analysis between autoimmune diseases and glioma: A Mendelian randomization study. Medicine (Baltimore) 2025; 104:e41815. [PMID: 40068088 PMCID: PMC11902947 DOI: 10.1097/md.0000000000041815] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
The causal association between the autoimmune disease and the development of glioma and its subtypes remains unclear. We performed a comprehensive Mendelian randomization (MR) to clarify their causal association from genetic perspective. We obtained the summary-level datasets for autoimmune diseases from recently published genome-wide association studies in the UK Biobank (UKB) and the FinnGen consortium. Additionally, we collected summary statistics datasets related to glioma and its subtypes from a comprehensive meta-analysis genome-wide association study, which included 12,488 cases and 18,169 controls. We primarily used inverse variance weighting method, supplemented by Bonferroni correction to account for multiple tests to reduce the probability of false positive results. We also performed sensitivity analyses to address potential pleiotropy and strengthen the reliability of the results. After meta-analysis, pernicious anemia may decrease the risk of glioblastoma (GBM) (UKB: odds ratio (OR) = 0.01, 95% confidence interval (CI) = 0.01-0.02, P = 1.01E-12; FinnGen: OR = 0.86, 95% CI = 0.79-0.93, P = .0002; Meta: OR = 0.04, 95% CI = 0.03-0.04). In reverse MR analysis, GBM decreased the risk of celiac disease (UKB: OR = 0.96, 95% CI = 0.95-0.98, P = .0000; FinnGen: OR = 0.89, 95% CI = 0.84-0.94, P = .0001; Meta: OR = 0.95, 95% CI = 0.94-0.97). Heterogeneity and pleiotropy analyses, and reverse analysis, confirmed the robustness of these results. From the genetic perspective, our MR study uncovered that pernicious anemia may decrease the risk of GBM. Conversely, GBM appeared to mitigate the risk of celiac disease. Future studies are required to validate the causal association and illuminate the underlying mechanisms.
Collapse
Affiliation(s)
- Ni Deng
- Department of Respiratory Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Rafeq Agila
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiang He
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chao You
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Songping Zheng
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
43
|
Lehmann B, Lee H, Anderson-Trocmé L, Kelleher J, Gorjanc G, Ralph PL. On ARGs, pedigrees, and genetic relatedness matrices. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.03.641310. [PMID: 40093116 PMCID: PMC11908205 DOI: 10.1101/2025.03.03.641310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Genetic relatedness is a central concept in genetics, underpinning studies of population and quantitative genetics in human, animal, and plant settings. It is typically stored as a genetic relatedness matrix (GRM), whose elements are pairwise relatedness values between individuals. This relatedness has been defined in various contexts based on pedigree, genotype, phylogeny, coalescent times, and, recently, ancestral recombination graph (ARG). ARG-based GRMs have been found to better capture the structure of a population and improve association studies relative to the genotype GRM. However, calculating GRMs and further operations with them is fundamentally challenging due to inherent quadratic time and space complexity. Here, we first discuss the different definitions of relatedness in a unifying context, making use of the additive model of a quantitative trait to provide a definition of "branch relatedness" and the corresponding "branch GRM". We explore the relationship between branch relatedness and pedigree relatedness through a case study of French-Canadian individuals that have a known pedigree. Through the tree sequence encoding of an ARG, we then derive an efficient algorithm for computing products between the branch GRM and a general vector, without explicitly forming the branch GRM. This algorithm leverages the sparse encoding of genomes with the tree sequence and hence enables large-scale computations with the branch GRM. We demonstrate the power of this algorithm by developing a randomized principal components algorithm for tree sequences that easily scales to millions of genomes. All algorithms are implemented in the open source tskit Python package. Taken together, this work consolidates the different notions of relatedness as branch relatedness and by leveraging the tree sequence encoding of an ARG it provides efficient algorithms that enable computations with the branch GRM that scale to mega-scale genomic datasets.
Collapse
Affiliation(s)
- Brieuc Lehmann
- Department of Statistical Science, University College London, WC1E 7HB, UK
| | - Hanbin Lee
- Department of Statistics, University of Michigan, Ann Arbor MI 48109, USA
| | | | - Jerome Kelleher
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, OX3 7LF, UK
| | - Gregor Gorjanc
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, UK
| | - Peter L Ralph
- Institute of Ecology and Evolution, University of Oregon, Eugene OR 97402, USA
- Department of Data Science, University of Oregon, Eugene OR 97402, USA
| |
Collapse
|
44
|
Yang Q, Li M, Chen P, Dou N, Liu M, Lu P, Yu C. Systematic Evaluation of the Impact of a Wide Range of Dietary Habits on Myocardial Infarction: A Two-Sample Mendelian Randomization Analysis. J Am Heart Assoc 2025; 14:e035936. [PMID: 40008582 DOI: 10.1161/jaha.124.035936] [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: 07/01/2024] [Accepted: 11/19/2024] [Indexed: 02/27/2025]
Abstract
BACKGROUND Myocardial infarction is a cardiovascular disease that significantly contributes to global morbidity and disability. Given the significant role of diet in the pathogenesis and prevention of cardiovascular diseases, this study rigorously investigates the causal relationship between dietary habits and myocardial infarction. METHODS AND RESULTS This study used large-scale genome-wide association studies with pooled UK Biobank data to explore associations between 9 dietary categories (83 types) and myocardial infarction. A 2-sample Mendelian randomization approach was applied to assess these associations, while multivariate Mendelian randomization and mediation analyses investigated the role of lipids in mediating the effects of diet on myocardial infarction. Univariate Mendelian analyses revealed genetic associations among 9 categories of dietary habits (83 types) and myocardial infarction. Notably, robust evidence indicates the "tablespoons of cooked vegetables per day" as the most significant risk factor for myocardial infarction development. "Coffee consumption(cups per day)" and "frequency of adding salt to food" were also identified as supplementary risk factors. In contrast, "overall alcohol intake" showed a protective effect, potentially by increasing high-density lipoprotein cholesterol (4.48% mediation) and reducing triglycerides (6.24% mediation). Cereal category, particularly "cereal consumption (bowls per week)" was associated with reduced myocardial infarction risk, contributing by raising high-density lipoprotein cholesterol (3.69% mediation) and lowering total cholesterol (8.33% mediation). Additionally, "overall cheese consumption" was also protective against myocardial infarction. CONCLUSIONS Our findings elucidate the influence of dietary habits on myocardial infarction, showing underlying genetic mechanisms and emphasizing the regulatory role of lipids as an intermediate.
Collapse
Affiliation(s)
- Qian Yang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Shandong Institute of Endocrine and Metabolic Diseases Jinan Shandong China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases Jinan Shandong China
| | - Man Li
- Department of Geratology Qilu Hospital of Shandong University Jinan Shandong China
| | - Pengcheng Chen
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Shandong Institute of Endocrine and Metabolic Diseases Jinan Shandong China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases Jinan Shandong China
| | - Naixin Dou
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Shandong Institute of Endocrine and Metabolic Diseases Jinan Shandong China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases Jinan Shandong China
| | - Mei Liu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Shandong Institute of Endocrine and Metabolic Diseases Jinan Shandong China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases Jinan Shandong China
| | - Peng Lu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Shandong Institute of Endocrine and Metabolic Diseases Jinan Shandong China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases Jinan Shandong China
| | - Chunxiao Yu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan Shandong China
- Shandong Institute of Endocrine and Metabolic Diseases Jinan Shandong China
- Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases Jinan Shandong China
| |
Collapse
|
45
|
Pivirotto A, Peles N, Hey J. Allele age estimators designed for whole genome datasets show only a moderate reduction in performance when applied to whole exome datasets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.02.01.578465. [PMID: 38370640 PMCID: PMC10871225 DOI: 10.1101/2024.02.01.578465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Personalized genomics in the healthcare system is becoming increasingly accessible as the costs of sequencing decreases. With the increase in the number of genomes, larger numbers of rare variants are being discovered, leading to important initiatives in identifying the functional impacts in relation to disease phenotypes. One way to characterize these variants is to estimate the time the mutation entered the population. However, allele age estimators such as those implemented in the programs Relate, Genealogical Estimator of Variant Age (GEVA), and Runtc, were developed based on the assumption that datasets include the entire genome. We examined the performance of each of these estimators on simulated exome data under a neutral constant population size model, as well as under population expansion and background selection models. We found that each provides usable estimates of allele age from whole-exome datasets. Relate performs the best amongst all three estimators with Pearson coefficients of 0.83 and 0.73 (with respect to true simulated values, for neutral constant and expansion population model, respectively) with a 12 percent and 20 percent decrease in correlation between whole genome and whole exome estimations. Of the three estimators, Relate is best able to parallelize to yield quick results with little resources, however, Relate is currently only able to scale to thousands of samples making it unable to match the hundreds of thousands of samples being currently released. While more work is needed to expand the capabilities of current methods of estimating allele age, these methods show a modest decrease in performance in the estimation of the age of mutations.
Collapse
Affiliation(s)
- Alyssa Pivirotto
- Center for Computational Genetics and Genomics, Temple University, Philadelphia, PA USA
| | - Noah Peles
- Center for Computational Genetics and Genomics, Temple University, Philadelphia, PA USA
| | - Jody Hey
- Center for Computational Genetics and Genomics, Temple University, Philadelphia, PA USA
| |
Collapse
|
46
|
Dominy J, Bai J, Koch C, Zaman Khan M, Khalid S, Chung JH, Panditrao M, Liu L, Zhang Q, Jahanzaib M, Mian MR, Liaqat MB, Raza SS, Sultana R, Jalal A, Saeed MH, Abbas S, Memon FR, Ishaq M, Saleheen K, Rasheed A, Gurtan A, Saleheen D. Human CD33 deficiency is associated with mild alteration of circulating white blood cell counts. PLoS Genet 2025; 21:e1011600. [PMID: 40043053 PMCID: PMC11927914 DOI: 10.1371/journal.pgen.1011600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 03/21/2025] [Accepted: 01/29/2025] [Indexed: 03/22/2025] Open
Abstract
The single pass transmembrane protein CD33 is enriched in phagocytic and hematopoietic cell types, such as monocytes. CD33 is thought to be associated with immune cell function, susceptibility to Alzheimer's disease, and rare leukemias. Antagonism or genetic ablation of CD33 has been proposed to treat Alzheimer's disease, hematological cancers, and as a selection mechanism for enriching genetically altered blood cells. To understand the impact of chronic CD33 loss or ablation, we describe individuals who we confirmed to be missing CD33 due to germline loss of function variants. Through PheWAS-based approaches using existing whole exome biobanks and bespoke phenotyping using recall-by-genotype (RBG) studies, we show that CD33 loss of function alters circulating white blood cell counts and distributions, albeit mildly and with no overt clinical pathology. These findings indicate that chronic CD33 antagonism/ablation is likely to be safe in humans.
Collapse
Grants
- RC2 HL101834 NHLBI NIH HHS
- R01 HL133339 NHLBI NIH HHS
- RC1 TW008485 FIC NIH HHS
- R01 HL145437 NHLBI NIH HHS
- R01 HG010689 NHGRI NIH HHS
- NIH
- Regeneron Pharmaceuticals, Eli Lilly & Company, Merck, Astra Zeneca, NGM Biopharmaceuticals Inc., GSK, Astellas Pharma Inc., and Novo Nordisk
- Novartis
- Regeneron, Novartis, AstraZeneca, and Astellas
Collapse
Affiliation(s)
- John Dominy
- Biomedical Research at Novartis, Cambridge, Massachusetts, United States of America
| | - Jirong Bai
- Biomedical Research at Novartis, Cambridge, Massachusetts, United States of America
| | - Christopher Koch
- Biomedical Research at Novartis, Cambridge, Massachusetts, United States of America
| | | | - Shareef Khalid
- Center for Non-Communicable Diseases, Karachi, Sindh, Pakistan
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Jonathan H. Chung
- Biomedical Research at Novartis, Cambridge, Massachusetts, United States of America
| | - Madhura Panditrao
- Biomedical Research at Novartis, Cambridge, Massachusetts, United States of America
| | - Lulu Liu
- Biomedical Research at Novartis, Cambridge, Massachusetts, United States of America
| | - Qi Zhang
- Biomedical Research at Novartis, Cambridge, Massachusetts, United States of America
| | | | | | | | | | - Riffat Sultana
- Karachi Institute of Heart Diseases, Karachi, Sindh, Pakistan
| | - Anjum Jalal
- Punjab Institute of Cardiology, Lahore, Punjab, Pakistan
| | | | - Shahid Abbas
- Faisalabad Institute of Cardiology, Faisalabad, Punjab, Pakistan
| | | | - Mohammad Ishaq
- Karachi Institute of Heart Diseases, Karachi, Sindh, Pakistan
| | - Kashif Saleheen
- Center for Non-Communicable Diseases, Karachi, Sindh, Pakistan
| | - Asif Rasheed
- Center for Non-Communicable Diseases, Karachi, Sindh, Pakistan
| | - Allan Gurtan
- Biomedical Research at Novartis, Cambridge, Massachusetts, United States of America
| | - Danish Saleheen
- Center for Non-Communicable Diseases, Karachi, Sindh, Pakistan
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Cardiology, Columbia University Irving Medical Center, New York, New York, United States of America
| |
Collapse
|
47
|
Jahangiri Esfahani S, Ao X, Oveisi A, Diatchenko L. Rare variant association studies: Significance, methods, and applications in chronic pain studies. Osteoarthritis Cartilage 2025; 33:313-321. [PMID: 39725155 DOI: 10.1016/j.joca.2024.12.006] [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: 06/21/2024] [Revised: 11/27/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024]
Abstract
Rare genetic variants, characterized by their low frequency in a population, have emerged as essential components in the study of complex disease genetics. The biology of rare variants underscores their significance, as they can exert profound effects on phenotypic variation and disease susceptibility. Recent advancements in sequencing technologies have yielded the availability of large-scale sequencing data such as the UK Biobank whole-exome sequencing (WES) cohort empowered researchers to conduct rare variant association studies (RVASs). This review paper discusses the significance of rare variants, available methodologies, and applications. We provide an overview of RVASs, emphasizing their relevance in unraveling the genetic architecture of complex diseases with special focus on chronic pain and Arthritis. Additionally, we discuss the strengths and limitations of various rare variant association testing methods, outlining a typical pipeline for conducting rare variant association. This pipeline encompasses crucial steps such as quality control of WES data, rare variant annotation, and association testing. It serves as a comprehensive guide for researchers in the field of chronic pain diseases interested in rare variant association studies in large-scale sequencing datasets like the UK Biobank WES cohort. Lastly, we discuss how the identified variants can be further investigated through detailed experimental studies in animal models to elucidate their functional impact and underlying mechanisms.
Collapse
Affiliation(s)
- Sahel Jahangiri Esfahani
- Faculty of Medicine and Health Sciences, Department of Human Genetics, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Xiang Ao
- Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Anahita Oveisi
- Department of Neuroscience, Faculty of Science, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Luda Diatchenko
- Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada.
| |
Collapse
|
48
|
Harris L, McDonagh EM, Zhang X, Fawcett K, Foreman A, Daneck P, Sergouniotis PI, Parkinson H, Mazzarotto F, Inouye M, Hollox EJ, Birney E, Fitzgerald T. Genome-wide association testing beyond SNPs. Nat Rev Genet 2025; 26:156-170. [PMID: 39375560 DOI: 10.1038/s41576-024-00778-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2024] [Indexed: 10/09/2024]
Abstract
Decades of genetic association testing in human cohorts have provided important insights into the genetic architecture and biological underpinnings of complex traits and diseases. However, for certain traits, genome-wide association studies (GWAS) for common SNPs are approaching signal saturation, which underscores the need to explore other types of genetic variation to understand the genetic basis of traits and diseases. Copy number variation (CNV) is an important source of heritability that is well known to functionally affect human traits. Recent technological and computational advances enable the large-scale, genome-wide evaluation of CNVs, with implications for downstream applications such as polygenic risk scoring and drug target identification. Here, we review the current state of CNV-GWAS, discuss current limitations in resource infrastructure that need to be overcome to enable the wider uptake of CNV-GWAS results, highlight emerging opportunities and suggest guidelines and standards for future GWAS for genetic variation beyond SNPs at scale.
Collapse
Affiliation(s)
- Laura Harris
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Ellen M McDonagh
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Xiaolei Zhang
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Katherine Fawcett
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
- Department of Population Health Sciences, University of Leicester, Leicester, UK
| | - Amy Foreman
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Petr Daneck
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Panagiotis I Sergouniotis
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
- Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Helen Parkinson
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Francesco Mazzarotto
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Michael Inouye
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Edward J Hollox
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Ewan Birney
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Tomas Fitzgerald
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK.
| |
Collapse
|
49
|
Charras A, Hiraki LT, Lewandowski L, Hedrich CM. Genetic and epigenetic factors shape phenotypes and outcomes in systemic lupus erythematosus - focus on juvenile-onset systemic lupus erythematosus. Curr Opin Rheumatol 2025; 37:149-163. [PMID: 39660463 PMCID: PMC11789615 DOI: 10.1097/bor.0000000000001072] [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: 12/12/2024]
Abstract
PURPOSE OF REVIEW Systemic lupus erythematosus (SLE) is a severe autoimmune/inflammatory disease. Patients with juvenile disease-onset and those of non-European ancestry are most severely affected. While the exact pathophysiology remains unknown, common and rare gene variants in the context of environmental exposure and epigenetic alterations are involved. This manuscript summarizes the current understanding of genetic and epigenetic contributors to SLE risk, manifestations and outcomes. RECENT FINDINGS Though SLE is a mechanistically complex disease, we are beginning to understand the impact of rare and common gene variants on disease expression and associated outcomes. Recent trans -ancestral and multigenerational studies suggest that differential genetic and environmental impacts shape phenotypic variability between age-groups and ancestries. High genetic burden associates with young age at disease-onset, organ involvement, and severity. Additional epigenetic impact contributes to disease-onset and severity, including SLE-phenotypes caused by rare single gene variants. Studies aiming to identify predictors of organ involvement and disease outcomes promise future patient stratification towards individualized treatment and care. SUMMARY An improved understanding of genetic variation and epigenetic marks explain phenotypic differences between age-groups and ancestries, promising their future exploitation for diagnostic, prognostic and therapeutic considerations.
Collapse
Affiliation(s)
- Amandine Charras
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Linda T. Hiraki
- Genetics & Genome Biology, Research Institute, and Division of Rheumatology, The Hospital for Sick Children, & Division of Rheumatology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Laura Lewandowski
- National Institute of Arthritis and Musculoskeletal and Skin diseases, NIH, Bethesda, Maryland, USA
| | - Christian M. Hedrich
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Department of Rheumatology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| |
Collapse
|
50
|
Liang Y, Ma D, Li M, Wang Z, Hao C, Sun Y, Hao X, Zuo C, Li S, Feng Y, Qi S, Wang Y, Sun S, Xu YM, Andreassen OA, Shi C. Exome sequencing identifies novel genes associated with cerebellar volume and microstructure. Commun Biol 2025; 8:344. [PMID: 40025133 PMCID: PMC11873060 DOI: 10.1038/s42003-025-07797-3] [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: 09/28/2024] [Accepted: 02/21/2025] [Indexed: 03/04/2025] Open
Abstract
Proteins encoded by exons are critical for cellular functions, and mutations in these genes often result in significant phenotypic effects. The cerebellum is linked to various heritable human disease phenotypes, yet genome-wide association studies have struggled to capture the effects of rare variants on cerebellar traits. This study conducts a large-scale exome association analysis using data from approximately 35,000 UK Biobank participants, examining seven cerebellar traits, including total cerebellar volume and white matter microstructure. We identify 90 genes associated with cerebellar traits, 60 of which were previously unreported in genome-wide association studies. Notable findings include the discovery of genes like PRKRA and TTK, as well as RASGRP3, linked to cerebellar volume and white matter microstructure. Gene enrichment analysis reveals associations with non-coding RNA processing, cognitive function, neurodegenerative diseases, and mental disorders, suggesting shared biological mechanisms between cerebellar phenotypes and neuropsychiatric diseases.
Collapse
Affiliation(s)
- Yuanyuan Liang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Dongrui Ma
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Mengjie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Zhiyun Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Chenwei Hao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yuemeng Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Xiaoyan Hao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Chunyan Zuo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Shuangjie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yanmei Feng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Shasha Qi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yunpeng Wang
- NORMENT, K.G. Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Shilei Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- NHC Key Laboratory of Prevention and treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Yu-Ming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- NHC Key Laboratory of Prevention and treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, Henan, China.
| | - Ole A Andreassen
- NORMENT, K.G. Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- NHC Key Laboratory of Prevention and treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, Henan, China.
| |
Collapse
|