1
|
Schwartz R, Warwick AN, Khawaja AP, Luben R, Khalid H, Phatak S, Jhingan M, de Vente C, Valmaggia P, Liakopoulos S, Olvera-Barrios A, Sánchez CI, Egan C, Bonelli R, Tufail A. Genetic Distinctions Between Reticular Pseudodrusen and Drusen: A Genome-Wide Association Study. Am J Ophthalmol 2025; 274:286-295. [PMID: 40064387 DOI: 10.1016/j.ajo.2025.03.007] [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: 01/17/2025] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 04/07/2025]
Abstract
OBJECTIVE To identify genetic determinants specific to reticular pseudodrusen (RPD) compared with drusen. DESIGN Genome-wide association study (GWAS) SUBJECTS: Participants with RPD, drusen, and controls from the UK Biobank (UKBB), a large, multisite, community-based cohort. METHODS Participants with RPD, drusen, and controls from the UK Biobank (UKBB), a large, multisite, community-based cohort, were included. A deep learning framework analyzed 169,370 optical coherence tomography (OCT) volumes to identify cases and controls within the UKBB. Five retina specialists validated the cohorts using OCT and color fundus photographs. Several GWAS were undertaken utilizing the quantity and presence of RPD and drusen. Genome-wide significance was defined as P < 5e-8. MAIN OUTCOMES MEASURES Genetic associations were examined with the number of RPD and drusen within 'pure' cases, where only RPD or drusen were present in either eye. A candidate approach assessed 46 previously known AMD loci. Secondary GWAS were conducted for number of RPD and drusen in mixed cases, and binary case-control analyses for pure RPD and pure drusen. RESULTS The study included 1787 participants: 1037 controls, 361 pure drusen, 66 pure RPD, and 323 mixed cases. The primary pure RPD GWAS identified four genome-wide significant loci: rs11200630 near ARMS2-HTRA1 (P = 1.9e-09), rs79641866 at PARD3B (P = 1.3e-08), rs143184903 near ITPR1 (P = 8.1e-09), and rs76377757 near SLN (P = 4.3e-08). The latter three are uncommon variants (minor allele frequency <5%). A significant association at the CFH locus was also observed using a candidate approach (P = 1.8e-04). For pure drusen, two loci reached genome-wide significance: rs10801555 at CFH (P = 6.0e-33) and rs61871744 at ARMS2-HTRA1 (P = 4.2e-20). CONCLUSIONS The study highlights a clear association between the ARMS2-HTRA1 locus and higher RPD load. Although the CFH locus association did not achieve genome-wide significance, a suggestive link was observed. Three novel associations unique to RPD were identified, albeit for uncommon genetic variants. Further studies with larger sample sizes are needed to explore these findings.
Collapse
Affiliation(s)
- Roy Schwartz
- From the Moorfields Eye Hospital NHS Foundation Trust (R.S., A.N.W., H.K., S.P., M.J., C.E., A.T.), London, UK; Institute of Health Informatics (R.S.), University College London, London, UK.
| | - Alasdair N Warwick
- From the Moorfields Eye Hospital NHS Foundation Trust (R.S., A.N.W., H.K., S.P., M.J., C.E., A.T.), London, UK; University College London Institute of Cardiovascular Science (A.N.W.), London, UK
| | - Anthony P Khawaja
- NIHR Biomedical Research Centre (A.P.K., R.L., P.V., A.O.B., C.E., A.T.), Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology, London, UK
| | - Robert Luben
- NIHR Biomedical Research Centre (A.P.K., R.L., P.V., A.O.B., C.E., A.T.), Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology, London, UK; MRC Epidemiology Unit (R.L.), University of Cambridge, Cambridge, UK
| | - Hagar Khalid
- From the Moorfields Eye Hospital NHS Foundation Trust (R.S., A.N.W., H.K., S.P., M.J., C.E., A.T.), London, UK
| | - Sumita Phatak
- From the Moorfields Eye Hospital NHS Foundation Trust (R.S., A.N.W., H.K., S.P., M.J., C.E., A.T.), London, UK
| | - Mahima Jhingan
- From the Moorfields Eye Hospital NHS Foundation Trust (R.S., A.N.W., H.K., S.P., M.J., C.E., A.T.), London, UK
| | - Coen de Vente
- Quantitative Healthcare Analysis (qurAI) Group (C.D.V., C.I.S.), Informatics Institute, University of Amsterdam, Amsterdam, Netherlands; Biomedical Engineering and Physics (C.D.V., C.I.S.), Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
| | - Philippe Valmaggia
- NIHR Biomedical Research Centre (A.P.K., R.L., P.V., A.O.B., C.E., A.T.), Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology, London, UK; Department of Biomedical Engineering (P.V.), University of Basel, Basel, Switzerland; Department of Ophthalmology (P.V.), University Hospital Basel, Basel, Switzerland
| | - Sandra Liakopoulos
- Cologne Image Reading Center (S.L.), Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Department of Ophthalmology (S.L.), Goethe University, Frankfurt, Germany
| | - Abraham Olvera-Barrios
- NIHR Biomedical Research Centre (A.P.K., R.L., P.V., A.O.B., C.E., A.T.), Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology, London, UK; Institute of Ophthalmology (A.O.B), University College London, London, UK
| | - Clara I Sánchez
- Quantitative Healthcare Analysis (qurAI) Group (C.D.V., C.I.S.), Informatics Institute, University of Amsterdam, Amsterdam, Netherlands; Biomedical Engineering and Physics (C.D.V., C.I.S.), Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
| | - Catherine Egan
- From the Moorfields Eye Hospital NHS Foundation Trust (R.S., A.N.W., H.K., S.P., M.J., C.E., A.T.), London, UK; NIHR Biomedical Research Centre (A.P.K., R.L., P.V., A.O.B., C.E., A.T.), Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology, London, UK
| | - Roberto Bonelli
- Lowy Medical Research Institute (R.B.), La Jolla, California, USA
| | - Adnan Tufail
- From the Moorfields Eye Hospital NHS Foundation Trust (R.S., A.N.W., H.K., S.P., M.J., C.E., A.T.), London, UK; NIHR Biomedical Research Centre (A.P.K., R.L., P.V., A.O.B., C.E., A.T.), Moorfields Eye Hospital NHS Foundation Trust & UCL Institute of Ophthalmology, London, UK
| |
Collapse
|
2
|
van der Meer D, Rahman Z, Ottas A, Parekh P, Kutrolli G, Stinson SE, Koromina M, Rokicki J, Sønderby IE, Parker N, Tesfaye M, Hindley G, Rødevand LN, Koch E, Estonian Biobank Research Team, Steen NE, Berg JP, O'Connell KS, Smeland OB, Frei O, Dale AM, Djurovic S, Lehto K, Alver M, Milani L, Shadrin AA, Andreassen OA. Pleiotropic and sex-specific genetic mechanisms of circulating metabolic markers. Nat Commun 2025; 16:4961. [PMID: 40436851 PMCID: PMC12119811 DOI: 10.1038/s41467-025-60058-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 05/14/2025] [Indexed: 06/01/2025] Open
Abstract
Metabolites in plasma form biosignatures of a range of common complex human diseases. Discovering variants with pleiotropic effects across metabolites can reveal underlying biological mechanisms. We therefore performed uni- and multivariate genome-wide association studies (GWAS) on 249 circulating metabolic markers across 328,006 UK Biobank and Estonian Biobank participants. We investigated rare variation through whole exome sequencing gene burden tests, analysed the role of body mass index through Mendelian randomization, and performed genome-wide interaction analyses with sex. We discovered 15,585 loci summed over the univariate GWAS, with high pleiotropy across markers, linked to a wide range of disorders. Findings from common and rare variant gene tests converged on lipid homeostasis pathways. 31 loci interacted with sex, mapped to genes involved in cholesterol processing. The findings offer insights into the genetic architecture of circulating metabolites, revealing pleiotropic loci, highlighting the role of rare variation, and uncovering sex-specific molecular mechanisms of lipid metabolism.
Collapse
Affiliation(s)
- Dennis van der Meer
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway.
| | - Zillur Rahman
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Aigar Ottas
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Riia 23b, Tartu, 51010, Estonia
| | - Pravesh Parekh
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Gleda Kutrolli
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Sara E Stinson
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Maria Koromina
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jaroslav Rokicki
- Centre of Research and Education in Forensic Psychiatry (SIFER), Oslo University Hospital, Oslo, Norway
| | - Ida E Sønderby
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Nadine Parker
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Markos Tesfaye
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Guy Hindley
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Psychosis Studies, Institute of Psychiatry, Psychology and Neurosciences, King's College London, London, UK
| | - Linn N Rødevand
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Elise Koch
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
| | | | - Nils Eiel Steen
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Jens Petter Berg
- Department of Medical Biochemistry, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kevin S O'Connell
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Olav B Smeland
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Oleksandr Frei
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Centre for Bioinformatics, Department of Informatics, University of Oslo, Oslo, Norway
| | - Anders M Dale
- Center for Multimodal Imaging and Genetics, University of California at San Diego, La Jolla, CA, 92037, USA
| | - Srdjan Djurovic
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Kelli Lehto
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Riia 23b, Tartu, 51010, Estonia
| | - Maris Alver
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Riia 23b, Tartu, 51010, Estonia
| | - Lili Milani
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Riia 23b, Tartu, 51010, Estonia
| | - Alexey A Shadrin
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Ole A Andreassen
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway.
- K.G. Jebsen Centre for Neurodevelopmental Disorders, University of Oslo and Oslo University Hospital, Oslo, Norway.
| |
Collapse
Collaborators
Andres Metspalu, Tõnu Esko, Reedik Mägi, Mait Metspalu, Mari Nelis, Georgi Hudjashov, Priit Palta, Nele Taba, Erik Abner, Jaanika Kronberg, Urmo Võsa,
Collapse
|
3
|
Yu X, Faggion S, Liu Y, Wang B, Zeng Q, Lu C, Hu J, Bargelloni L, Fang L, Bao Z. Role of multi-omics in aquaculture genetics and breeding: current status and future perspective. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2828-8. [PMID: 40448907 DOI: 10.1007/s11427-024-2828-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/26/2024] [Indexed: 06/02/2025]
Abstract
Aquaculture, a fast-growing sector, plays an important role in the supply of nutrient-rich food for humans. Selective breeding is a promising approach to ensure the development and sustainability of intensive aquaculture systems by achieving cumulative and permanent improvements in desirable traits. The advancement of omics technologies offers unprecedented opportunities for genetic improvement, especially in the prioritization of SNPs to be used in the genomic selection and editing of economically important traits. This review highlights novel breeding strategies in aquaculture, emphasizing how multi-omics data can be integrated into selective breeding programs. Specifically, we discuss the current achievements in integrating functional data into conventional genomic prediction models and highlight the potential of artificial intelligence to efficiently map genes and predict phenotypes or genetic merit using multi-omics data. Ultimately, we discuss genome editing methods for their potential to fix existing alleles, introduce alleles from wild populations or related species, and create de novo alleles, with the general goal of improving commercially important traits in aquaculture species.
Collapse
Affiliation(s)
- Xiaofei Yu
- Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Sara Faggion
- Department of Comparative Biomedicine and Food Science, University of Padova, Legnaro, 35020, Italy
| | - Yuxiang Liu
- Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Bo Wang
- Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Qifan Zeng
- Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Chunzhe Lu
- Groningen Biomolecular Sciences & Biotechnology Institute, University of Groningen, Groningen, 9747 AG, the Netherlands
| | - Jingjie Hu
- Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Luca Bargelloni
- Department of Comparative Biomedicine and Food Science, University of Padova, Legnaro, 35020, Italy
| | - Lingzhao Fang
- Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus, 8830, Denmark.
| | - Zhenmin Bao
- Ministry of Education Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
4
|
Jee YH, Wang Y, Jung KJ, Lee JY, Kimm H, Duan R, Price AL, Martin AR, Kraft P. Genome-wide association studies in a large Korean cohort identify quantitative trait loci for 36 traits and illuminate their genetic architectures. Nat Commun 2025; 16:4935. [PMID: 40436827 PMCID: PMC12120081 DOI: 10.1038/s41467-025-59950-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 05/08/2025] [Indexed: 06/01/2025] Open
Abstract
Genome-wide association studies (GWAS) have predominantly focused on European ancestry populations, limiting biological discoveries across diverse populations. Here we report GWAS findings from 153,950 individuals across 36 quantitative traits in the Korean Cancer Prevention Study-II (KCPS2) Biobank. We discovered 301 previously unreported genetic loci in KCPS2, including an association between thyroid-stimulating hormone and CD36. Meta-analysis with the Korean Genome and Epidemiology Study, Biobank Japan, Taiwan Biobank, and UK Biobank identified 4588 loci that were not significant in any contributing GWAS. We describe differences in genetic architectures across these East Asian and European samples. We also highlight East Asian specific associations, including a known pleiotropic missense variant in ALDH2, which fine-mapping identified as a likely causal variant for multiple traits. Our findings provide insights into the genetic architecture of complex traits in East Asian populations and highlight how broadening the population diversity of GWAS samples can aid discovery.
Collapse
Affiliation(s)
- Yon Ho Jee
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Ying Wang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Keum Ji Jung
- Institute for Health Promotion, Department of Epidemiology and Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea.
| | - Ji-Young Lee
- Institute for Health Promotion, Department of Epidemiology and Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| | - Heejin Kimm
- Institute for Health Promotion, Department of Epidemiology and Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| | - Rui Duan
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Alkes L Price
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alicia R Martin
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Trans-Divisional Research Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Boston, MD, USA.
| |
Collapse
|
5
|
Pullin JM, Wallace C. Variant-specific priors clarify colocalisation analysis. PLoS Genet 2025; 21:e1011697. [PMID: 40424384 DOI: 10.1371/journal.pgen.1011697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
Linking GWAS variants to their causal gene and context remains an ongoing challenge. A widely used method for performing this analysis is the coloc package for statistical colocalisation analysis, which can be used to link GWAS and eQTL associations. Currently, coloc assumes that all variants in a region are equally likely to be causal, despite the success of fine-mapping methods that use additional information to adjust their prior probabilities. In this paper we propose and implement an approach for specifying variant-specific prior probabilities in the coloc method. We describe and compare six source of information for specifying prior probabilities: non-coding constraint, enhancer-gene link scores, the output of the PolyFun method and three estimates of eQTL-TSS distance densities. Using simulations and analysis of ground-truth pQTL-eQTL colocalisations we show that variant-specific priors, particularly the eQTL-TSS distance density priors, can improve colocalisation performance. Furthermore, across GWAS-eQTL colocalisations variant-specific priors changed colocalisation significance in up to 14.1% of colocalisations, at some loci revealing the likely causal gene.
Collapse
Affiliation(s)
- Jeffrey M Pullin
- MRC Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom
| | - Chris Wallace
- MRC Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
6
|
Erdman V, Petintseva A, Timasheva Y, Tuktarova I, Nasibullin T, Korytina G. Genetic predictors of longevity and survival in cellular homeostasis genes: A case-control study. Gene 2025; 962:149576. [PMID: 40398646 DOI: 10.1016/j.gene.2025.149576] [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: 02/21/2025] [Revised: 05/05/2025] [Accepted: 05/17/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Longevity is defined by the ability to maintain both physical and mental health throughout a long life and may results from adaptive mechanisms that mitigate aging's detrimental effects. METHODS The 20-year follow-up study of 3,312 unrelated individuals aged 18-114 years from the Volga-Ural region of Eurasia investigated variants in cellular homeostasis genes IGF1, PIK3R1, AKT1, MTOR, NFE2L2, KEAP1, HIF1A, TP53, and SIRT1, to identify associations with clinical aging phenotypes and healthy longevity. RESULTS In men, KEAP1 (rs1048290) CC genotype was a longevity and survival marker (OR = 2.39, P = 3E-05, HR = 0.54, P = 2.4E-03). NFE2L2 (rs6721961) TT genotype was linked to higher mortality (HR = 1.77, P = 0.031), particularly combined with KEAP1 (rs1048290) G and AKT1 (rs3803304) C alleles (HR = 2.8, P = 0.023). In women, AKT1 (rs3803304) C allele interacted with NFE2L2 (rs6721961) TT genotype (SF = 0.13, P = 3.6E-03), and was linked to longevity (OR = 2.22, P = 6.3E-03) and protection against cerebrovascular diseases (OR = 0.62, P = 5.1E-03). AKT1 (rs3803304) GG genotype, along with HIF1A (rs11549465) T and SIRT1 (rs3758391) T alleles (SF = 2.52, P = 1.5E-03), promoted survival (HR = 0.71, P = 0.014). In men, HIF1A (rs11549465) TT genotype predicted cardiovascular mortality (HR = 7.5, P = 5.5E-03). SIRT1 (rs3758391) TT genotype was associated with improved survival in individuals with diabetes (HR = 0.4, P = 5.8E-03) and multimorbidity (HR = 0.48, P = 0.025). CONCLUSION Variants in NFE2L2, KEAP1, SIRT1, AKT1, and HIF1A, along with their interactions, were significantly associated with survival in age-related diseases and healthy longevity.
Collapse
Affiliation(s)
- Vera Erdman
- Institute of Biochemistry and Genetics, Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa 450054, Russia; Bashkir State Medical University, Ufa 450008, Russia.
| | - Anna Petintseva
- Institute of Biochemistry and Genetics, Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa 450054, Russia
| | - Yanina Timasheva
- Institute of Biochemistry and Genetics, Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa 450054, Russia; Bashkir State Medical University, Ufa 450008, Russia
| | - Ilsiar Tuktarova
- Institute of Biochemistry and Genetics, Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa 450054, Russia
| | - Timur Nasibullin
- Institute of Biochemistry and Genetics, Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa 450054, Russia
| | - Gulnaz Korytina
- Institute of Biochemistry and Genetics, Ufa Federal Research Centre of the Russian Academy of Sciences, Ufa 450054, Russia; Bashkir State Medical University, Ufa 450008, Russia
| |
Collapse
|
7
|
Zhou F, Astle WJ, Butterworth AS, Asimit JL. Improved genetic discovery and fine-mapping resolution through multivariate latent factor analysis of high-dimensional traits. CELL GENOMICS 2025; 5:100847. [PMID: 40220762 DOI: 10.1016/j.xgen.2025.100847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/23/2024] [Accepted: 03/14/2025] [Indexed: 04/14/2025]
Abstract
Genome-wide association studies (GWASs) of high-dimensional traits, such as blood cell or metabolic traits, often use univariate approaches, ignoring trait relationships. Biological mechanisms generating variation in high-dimensional traits can be captured parsimoniously through a GWAS of latent factors. Here, we introduce flashfmZero, a zero-correlation latent-factor-based multi-trait fine-mapping approach. In an application to 25 latent factors derived from 99 blood cell traits in the INTERVAL cohort, we show that latent factor GWASs enable the detection of signals generating sub-threshold associations with several blood cell traits. The 99% credible sets (CS99) from flashfmZero were equal to or smaller in size than those from univariate fine-mapping of blood cell traits in 87% of our comparisons. In all cases univariate latent factor CS99 contained those from flashfmZero. Our latent factor approaches can be applied to GWAS summary statistics and will enhance power for the discovery and fine-mapping of associations for many traits.
Collapse
Affiliation(s)
- Feng Zhou
- MRC Biostatistics Unit, University of Cambridge, Cambridge CB2 0SR, UK
| | - William J Astle
- MRC Biostatistics Unit, University of Cambridge, Cambridge CB2 0SR, UK; NHS Blood and Transplant, Cambridge CB2 0PT, UK; NIHR Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge CB2 0BB, UK
| | - Adam S Butterworth
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge CB2 0BB, UK; British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB2 0BB, UK; Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge CB2 0BB, UK; British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge CB2 0QQ, UK; Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge CB10 1SA, UK
| | - Jennifer L Asimit
- MRC Biostatistics Unit, University of Cambridge, Cambridge CB2 0SR, UK.
| |
Collapse
|
8
|
Sharma SD, Hum RM, Nair N, Marshall L, Storrie A, Bowes J, MacGregor A, Yates M, Morris AP, Verstappen S, Barton A, van Steenbergen H, Knevel R, van der Helm-van Mil A, Viatte S. Systematic review and independent validation of genetic factors of radiographic outcome in rheumatoid arthritis identifies a genome-wide association with CARD9. Ann Rheum Dis 2025:S0003-4967(25)00897-0. [PMID: 40345877 DOI: 10.1016/j.ard.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/26/2025] [Accepted: 04/05/2025] [Indexed: 05/11/2025]
Abstract
OBJECTIVES This study aimed to investigate non-HLA genetic mechanisms underlying radiographic severity in rheumatoid arthritis (RA). METHODS A systematic review of publications reporting non-HLA genetic associations with radiographic severity in RA across ancestries was undertaken. Experimental validation was performed in the Norfolk Arthritis Register, comprising 1407 patients with available genetic and treatment data followed prospectively for up to 10 years, with 2198 longitudinal radiographs. Genome-wide genotyping was performed with Illumina Human Core Exome Array. Radiographic outcomes (presence of erosions; Larsen score) were modelled longitudinally. Fine mapping and functional annotations to refine associations to potential causative loci were undertaken using FUMA, PolyPhen2, and RegulomeDB. RESULTS The systematic review identified 102 publications reporting 139 independent associations with radiographic outcome. Association with 15 independent polymorphisms were replicated in the Norfolk Arthritis Register data set, implicating adaptive immune processes (Th1, Th2, and Th17 pathways), cytokine regulation, and osteoclast differentiation. Notably, we refined the association of rs59902911 at the CARD9 locus to an intronic polymorphism within an active enhancer (rs78892335), achieving genome-wide significance and with an effect size exceeding the minimal clinically important difference for each copy of the minor allele (4.78 Larsen units/copy; 95% CI, 3.15-6.41; p = 9.01 × 10-9). This polymorphism is associated with the expression of CARD9 in immune cells, including B cells. CONCLUSIONS We provide a comprehensive list of validated genetic associations with RA outcome and demonstrate that non-HLA polymorphisms can associate with radiographic severity independently of disease susceptibility. This highlights the importance of dedicated genetic outcome studies for patient stratification in precision medicine for RA.
Collapse
Affiliation(s)
- Seema Devi Sharma
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Ryan Malcolm Hum
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Nisha Nair
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Lysette Marshall
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom
| | - Alice Storrie
- Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - John Bowes
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Alexander MacGregor
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom; Department of Rheumatology, Norfolk and Norwich University Hospital, United Kingdom
| | - Max Yates
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom; Department of Rheumatology, Norfolk and Norwich University Hospital, United Kingdom
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Suzanne Verstappen
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom; Centre for Epidemiology Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom
| | - Anne Barton
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Hanna van Steenbergen
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rachel Knevel
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Sebastien Viatte
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, United Kingdom; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom; Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
9
|
Laman Trip DS, van Oostrum M, Memon D, Frommelt F, Baptista D, Panneerselvam K, Bradley G, Licata L, Hermjakob H, Orchard S, Trynka G, McDonagh EM, Fossati A, Aebersold R, Gstaiger M, Wollscheid B, Beltrao P. A tissue-specific atlas of protein-protein associations enables prioritization of candidate disease genes. Nat Biotechnol 2025:10.1038/s41587-025-02659-z. [PMID: 40316700 DOI: 10.1038/s41587-025-02659-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 03/28/2025] [Indexed: 05/04/2025]
Abstract
Despite progress in mapping protein-protein interactions, their tissue specificity is understudied. Here, given that protein coabundance is predictive of functional association, we compiled and analyzed protein abundance data of 7,811 proteomic samples from 11 human tissues to produce an atlas of tissue-specific protein associations. We find that this method recapitulates known protein complexes and the larger structural organization of the cell. Interactions of stable protein complexes are well preserved across tissues, while cell-type-specific cellular structures, such as synaptic components, are found to represent a substantial driver of differences between tissues. Over 25% of associations are tissue specific, of which <7% are because of differences in gene expression. We validate protein associations for the brain through cofractionation experiments in synaptosomes, curation of brain-derived pulldown data and AlphaFold2 modeling. We also construct a network of brain interactions for schizophrenia-related genes, indicating that our approach can functionally prioritize candidate disease genes in loci linked to brain disorders.
Collapse
Affiliation(s)
- Diederik S Laman Trip
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
| | - Marc van Oostrum
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, Institute of Translational Medicine, ETH Zurich, Zurich, Switzerland
- Biozentrum, University of Basel, Basel, Switzerland
| | - Danish Memon
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
| | - Fabian Frommelt
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Delora Baptista
- Gulbenkian Institute for Molecular Medicine, Oeiras, Portugal
| | - Kalpana Panneerselvam
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
| | - Glyn Bradley
- Computational Biology, Functional Genomics, GSK, Stevenage, UK
| | - Luana Licata
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Henning Hermjakob
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
| | - Sandra Orchard
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
| | - Gosia Trynka
- Open Targets, Wellcome Genome Campus, Cambridge, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Ellen M McDonagh
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Andrea Fossati
- Science for Life Laboratory, Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Solna, Sweden
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Matthias Gstaiger
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Bernd Wollscheid
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, Institute of Translational Medicine, ETH Zurich, Zurich, Switzerland
| | - Pedro Beltrao
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, Switzerland.
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK.
- Open Targets, Wellcome Genome Campus, Cambridge, UK.
- Gulbenkian Institute for Molecular Medicine, Oeiras, Portugal.
| |
Collapse
|
10
|
Jiang L, Shen M, Zhang S, Zhang J, Shi Y, Gu Y, Yang T, Fu Q, Wang B, Chen Y, Xu K, Chen H. A regulatory variant rs9379874 in T1D risk region 6p22.2 affects BTN3A1 expression regulating T cell function. Acta Diabetol 2025; 62:695-706. [PMID: 39417845 DOI: 10.1007/s00592-024-02389-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVE Genome-wide association studies (GWAS) have identified that 6p22.2 region is associated with type 1 diabetes (T1D) risk in the Chinese Han population. This study aims to reveal associations between this risk region and T1D subgroups and related clinical features, and further identify causal variant(s) and target gene(s) in this region. METHODS 2608 T1D and 4814 healthy controls were recruited from East, Central, and South China. Baseline data and genotyping for rs4320356 were collected. The most likely causal variant and gene were identified by bioinformatics analysis, dual-luciferase reporter assays, expression quantitative trait loci (eQTL), and functional annotation of the non-coding region within the 6p22.2 region. RESULTS The leading variant rs4320356 in the 6p22.2 region was associated with T1D risk in the Chinese and Europeans. However, this variant was not significantly associated with islet function or autoimmunity. In silico analysis suggested rs9379874 was the most potential causal variant for T1D risk among thymus, spleen, and T cells, overlapping with the enhancer-related histone mark in multiple T cell subsets. Dual luciferase reporter assay and eQTL showed that the T allele of rs9379874 increased BTN3A1 expression by binding to FOXA1. Public single-cell RNA sequencing analysis indicated that BTN3A1 was related to T-cell activation, ATP metabolism, and cytokine metabolism pathways, which might contribute to T1D development. CONCLUSION This study indicates that a functional variant rs9379874 regulates BTN3A1 expression, expanding the genomic landscape of T1D risk and offering a potential target for developing novel therapies.
Collapse
Affiliation(s)
- Liying Jiang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Department of Rehabilitation Medicine, Lishui People's Hospital, Lishui, 323000, Zhejiang, China
| | - Min Shen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Saisai Zhang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jie Zhang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yun Shi
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yong Gu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Tao Yang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Qi Fu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Bingwei Wang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yang Chen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Kuanfeng Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Heng Chen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
11
|
Molinero E, Pena RN, Estany J, Ros-Freixedes R. Unravelling novel and closely linked association signals for fat-related traits in pigs using prioritised variants from whole-genome sequence data. Animal 2025; 19:101496. [PMID: 40250079 DOI: 10.1016/j.animal.2025.101496] [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: 10/15/2024] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/20/2025] Open
Abstract
For most production traits, the largest proportions of genetic variance remain unmapped. Dense whole-genome sequence (WGS) data enable the possibility of discovering novel associations as well as unravelling closely linked association signals with a resolution that marker arrays cannot reach. However, the identification of variants from WGS data that are causal of the variation of complex traits is hindered by the high dimensionality and linkage disequilibrium. Thus, at best, we can narrow the circle around the causal variants to prioritise a set of variants for their posterior validation. In this study, we assessed the utility of WGS data for uncovering associations of weaker effects using, as a model, fat content and composition traits in a Duroc pig population where we previously described major effects of the LEPR and SCD genes. We genotyped 971 pigs for a set of 182 variants from 154 candidate genes that were prioritised from amongst the WGS variants discovered in 205 sequenced individuals. These variants were prioritised conditional to LEPR and SCD. The association of the prioritised variants with the target traits was then tested in the confirmation set of 971 pigs. A total of 17 potentially independent quantitative trait loci (8.4% of the total number of studied genes) were significantly associated (q-value < 0.05) with at least one of the studied traits. We identified novel associations attributable to genes such as ABCC2, MOGAT2, or PLPP1 for backfat thickness, myristic acid content, and monounsaturated fatty acid content, respectively. Our results also revealed a finer granularity of weaker genetic effects in loci such as those around the DGAT2 and FADS2 genes, which may mask the effects of closely located genes like MOGAT2 and DAGLA, respectively. To refine the prioritisation of variants for validation studies, especially when targeting those of weaker effects, we recommend larger and more diverse discovery sets, more precise and complete functional gene annotation, and the integration of other omics data.
Collapse
Affiliation(s)
- E Molinero
- Department of Animal Science, University of Lleida, 191 Av. Alcalde Rovira Roure, 25198 Lleida, Spain; Agrotecnio-CERCA Center, 191 Av. Alcalde Rovira Roure, 25198 Lleida, Spain
| | - R N Pena
- Department of Animal Science, University of Lleida, 191 Av. Alcalde Rovira Roure, 25198 Lleida, Spain; Agrotecnio-CERCA Center, 191 Av. Alcalde Rovira Roure, 25198 Lleida, Spain
| | - J Estany
- Department of Animal Science, University of Lleida, 191 Av. Alcalde Rovira Roure, 25198 Lleida, Spain; Agrotecnio-CERCA Center, 191 Av. Alcalde Rovira Roure, 25198 Lleida, Spain
| | - R Ros-Freixedes
- Department of Animal Science, University of Lleida, 191 Av. Alcalde Rovira Roure, 25198 Lleida, Spain; Agrotecnio-CERCA Center, 191 Av. Alcalde Rovira Roure, 25198 Lleida, Spain.
| |
Collapse
|
12
|
Wood TWP, Henriques WS, Cullen HB, Romero M, Blengini CS, Sarathy S, Sorkin J, Bekele H, Jin C, Kim S, Wang X, Laureau R, Chemiakine A, Khondker RC, Isola JVV, Stout MB, Gennarino VA, Mogessie B, Jain D, Schindler K, Suh Y, Wiedenheft B, Berchowitz LE. The retrotransposon-derived capsid genes PNMA1 and PNMA4 maintain reproductive capacity. NATURE AGING 2025; 5:765-779. [PMID: 40263616 DOI: 10.1038/s43587-025-00852-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 03/05/2025] [Indexed: 04/24/2025]
Abstract
Almost half of the human genome consists of retrotransposons-'parasitic' sequences that insert themselves into the host genome via an RNA intermediate. Although most of these sequences are silenced or mutationally deactivated, they can present opportunities for evolutionary innovation: mutation of a deteriorating retrotransposon can result in a gene that provides a selective advantage to the host in a process termed 'domestication'1-3. The PNMA family of gag-like capsid genes was domesticated from an ancient vertebrate retrotransposon of the Metaviridae clade at least 100 million years ago4,5. PNMA1 and PNMA4 are positively regulated by the master germ cell transcription factors MYBL1 and STRA8, and their transcripts are bound by the translational regulator DAZL during gametogenesis6. This developmental regulation of PNMA1 and PNMA4 expression in gonadal tissue suggested to us that they might serve a reproductive function. Through the analysis of donated human ovaries, genome-wide association studies (GWASs) and mouse models, we found that PNMA1 and PNMA4 are necessary for the maintenance of a normal reproductive lifespan. These proteins self-assemble into capsid-like structures that exit human cells, and we observed large PNMA4 particles in mouse male gonadal tissue that contain RNA and are consistent with capsid formation.
Collapse
Affiliation(s)
- Thomas W P Wood
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - William S Henriques
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA
| | - Harrison B Cullen
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Mayra Romero
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Cecilia S Blengini
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Shreya Sarathy
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Julia Sorkin
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Hilina Bekele
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Chen Jin
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Seungsoo Kim
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Xifan Wang
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Raphaelle Laureau
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Alexei Chemiakine
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - Rishad C Khondker
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
| | - José V V Isola
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Michael B Stout
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Vincenzo A Gennarino
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Stem Cell Initiative, New York, NY, USA
- Initiative for Columbia Ataxia and Tremor, New York, NY, USA
| | - Binyam Mogessie
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Devanshi Jain
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Karen Schindler
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Yousin Suh
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Blake Wiedenheft
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA
| | - Luke E Berchowitz
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA.
- Taub Institute for Research on Alzheimer's and the Aging Brain, New York, NY, USA.
| |
Collapse
|
13
|
Yang Z, Wang C, Posadas-Garcia YS, Añorve-Garibay V, Vardarajan B, Estrada AM, Sohail M, Mayeux R, Ionita-Laza I. Fine-mapping in admixed populations using CARMA-X, with applications to Latin American studies. Am J Hum Genet 2025; 112:1215-1232. [PMID: 40147449 PMCID: PMC12120188 DOI: 10.1016/j.ajhg.2025.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 02/23/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025] Open
Abstract
Genome-wide association studies (GWASs) in ancestrally diverse populations are rapidly expanding, opening up unique opportunities for novel gene discoveries and increased utility of genetic findings in non-European individuals. A popular technique to identify putative causal variants at GWAS loci is via statistical fine-mapping. Despite tremendous efforts, fine-mapping remains a very challenging task, even in the relatively simple scenario of studies with a single, homogeneous population. For studies with admixed individuals, such as within Latin America and the Caribbean, methods for gene discovery are still limited. Here, we propose a Bayesian model for fine-mapping in admixed populations, CARMA-X, that addresses some of the unique challenges of admixed individuals. The proposed method includes an estimation method for the linkage disequilibrium (LD) matrix that accounts for small reference panels for admixed individuals, heterogeneity across populations and cross-ancestry LD, and a Bayesian hypothesis test that leads to robust fine-mapping when relying on external reference panels of modest size for LD estimation. Using simulations, we compare performance with recently proposed fine-mapping methods for multi-ancestry studies and show that the proposed model provides higher power while controlling false discoveries, especially when using an out-of-sample LD matrix. We further illustrate our approach through applications to two Latin American genetic studies, the Estudio Familiar de Influencia Genética en Alzheimer (EFIGA) study in the Dominican Republic and the Mexican Biobank, where we show the benefit of modeling ancestry-specific effects by prioritizing putative causal variants and genes, including several findings driven by ancestry-specific effects in the African and Native American ancestries.
Collapse
Affiliation(s)
- Zikun Yang
- Department of Biostatistics, Columbia University, New York, NY, USA.
| | - Chen Wang
- Department of Biostatistics, Columbia University, New York, NY, USA
| | | | | | - Badri Vardarajan
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Andrés Moreno Estrada
- Unidad de Genómica Avanzada (UGA-LANGEBIO), Centro de Investigación y Estudios Avanzados del IPN (Cinvestav), Irapuato, Mexico
| | - Mashaal Sohail
- Center for Genomic Sciences, National Autonomous University of Mexico, Mexico City, Mexico
| | - Richard Mayeux
- Department of Neurology, College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Iuliana Ionita-Laza
- Department of Biostatistics, Columbia University, New York, NY, USA; Department of Statistics, Lund University, Lund, Sweden.
| |
Collapse
|
14
|
Rokicki J, Campbell ML, van der Meer D, Sartorius AI, Tesli N, Jahołkowski P, Shadrin A, Andreassen O, Westlye LT, Quintana DS, Haukvik UK. Brain-based gene expression and corresponding behavioural relevance of risk genes for broad antisocial behaviour. Neuroimage 2025; 311:121198. [PMID: 40216214 DOI: 10.1016/j.neuroimage.2025.121198] [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: 12/03/2024] [Revised: 03/27/2025] [Accepted: 04/09/2025] [Indexed: 04/15/2025] Open
Abstract
Antisocial behaviour (ASB) involves persistent irresponsible, delinquent activities violating rights and safety of others. A meta-analysis of genome-wide association studies revealed significant genetic associations with ASB, yet their brain expression patterns and behavioural relevance remain unclear. Our investigation of fifteen genes associated with ASB examined their biological role and distribution across tissues, integrating post-mortem brain sample data from the Allen-Human-Brain Atlas and the Genotype-Tissue Expression project. We found that these genes were differentially expressed in the brain, particularly in regions like the cerebellum, putamen, and caudate, and were notably downregulated in the pancreas. Single cell type expression analysis revealed that ASB-associated genes had strong correlations with ductal and endothelial cells in the pancreas, indicating a possible metabolic influence on ASB. Certain genes like NTN1, SMAD5, NCAM2, and CDC42EP3 displayed specificity for cognitive terms including chronic pain, heart rate, and aphasia. These expression patterns aligned with neurocognitive domains related to thinking, and learning, distress, motor skills, as determined by fMRI analysis. This study connects specific brain gene expression with potential genetic and metabolic factors in ASB, offering novel insights into its biological basis and possible interdisciplinary approaches to understanding and addressing aggressive behaviours.
Collapse
Affiliation(s)
- Jaroslav Rokicki
- Centre of Research and Education in Forensic Psychiatry (SIFER), Oslo University Hospital, Oslo, Norway.
| | - Megan L Campbell
- SAMRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry and Neuroscience Institute, University of Cape Town, Cape Town, South Africa; Global Initiative for Neuropsychiatric Genetics Education in Research, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Dennis van der Meer
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway; School of Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, , Netherlands
| | - Alina I Sartorius
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway
| | - Natalia Tesli
- Centre of Research and Education in Forensic Psychiatry (SIFER), Oslo University Hospital, Oslo, Norway; Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Piotr Jahołkowski
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Alexey Shadrin
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ole Andreassen
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway; KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Lars T Westlye
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway
| | - Daniel S Quintana
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway; NevSom, Department of Rare Disorders and Disabilities, Oslo University Hospital, Oslo, Norway
| | - Unn K Haukvik
- Centre of Research and Education in Forensic Psychiatry (SIFER), Oslo University Hospital, Oslo, Norway; Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Mental health and addiction, Institute of Clinical Medicine, University of Oslo, Norway
| |
Collapse
|
15
|
Liu S, Yang J, Lin Y, Zhang L, Luo W. Exploring the comorbidity association and biological mechanisms of chronic rhinosinusitis and chronic obstructive pulmonary disease. Sci Rep 2025; 15:13855. [PMID: 40263405 PMCID: PMC12015216 DOI: 10.1038/s41598-025-98175-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/09/2025] [Indexed: 04/24/2025] Open
Abstract
Although chronic rhinosinusitis (CRS) and chronic obstructive pulmonary disease (COPD) are both common chronic inflammatory diseases, the interaction between their comorbidities remains poorly understood within the unified airway disease framework. This study, for the first time, integrated multi-omics analysis and large-scale epidemiological data to explore their common mechanisms and clinical significance. The NHANES database was used for analysis, and multivariate logistic regression was employed to assess the comorbidity risk of CRS and COPD. Machine-learning models (glmnet, ranger, and xgboost) were used to analyze the NHANES data to determine the best model. Subsequently, Mendelian randomization(MR) was applied to explore relevant associations. Additionally, CRS and COPD datasets from GEO were further analyzed to identify potential targets. The NHANES analysis showed a significant association between CRS and COPD, with MR results indicating that CRS significantly increased the risk of COPD. Multi-omics integration revealed that C3 and CD163 are core targets in CRS/COPD patients. The ranger model was identified as the most suitable in this study. This study provides new evidence that CRS is an independent risk factor for COPD and establishes a unified airway mechanism centered on C3-CD163-mediated inflammation. These findings advance the "one airway, one disease" paradigm and support a dual-target therapeutic strategy.
Collapse
MESH Headings
- Humans
- Pulmonary Disease, Chronic Obstructive/epidemiology
- Pulmonary Disease, Chronic Obstructive/genetics
- Sinusitis/epidemiology
- Sinusitis/genetics
- Comorbidity
- Rhinitis/epidemiology
- Rhinitis/genetics
- Chronic Disease
- Male
- Female
- Middle Aged
- Risk Factors
- Receptors, Cell Surface/metabolism
- Receptors, Cell Surface/genetics
- CD163 Antigen
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Mendelian Randomization Analysis
- Aged
- Machine Learning
- Adult
- Rhinosinusitis
Collapse
Affiliation(s)
- Shihan Liu
- Department of Otorhinolaryngology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jinxiong Yang
- Department of Otorhinolaryngology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiyi Lin
- Department of Otorhinolaryngology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lingli Zhang
- Department of Otorhinolaryngology, Central Hospital Affiliated to Chongqing University of Technology, Chongqing, China.
| | - Wenlong Luo
- Department of Otorhinolaryngology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
16
|
Shi X, Geng S, Hou J, Shi T, Qin M, Li W, Dai Z, Zhou Z, Zhang M, Lei Z. Unveiling Novel Genetic Loci and Superior Alleles for Nickel Accumulation in Wheat via Genome-Wide Association Study. PLANTS (BASEL, SWITZERLAND) 2025; 14:1262. [PMID: 40284150 PMCID: PMC12030696 DOI: 10.3390/plants14081262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
Nickel (Ni) pollution poses significant threats to human health and crop development through the food chain. This study aimed to identify the novel genomic regions and superior alleles associated with Ni accumulation in wheat (Triticum aestivum L.) grains using genome-wide association analysis (GWAS) with a diversity panel of 207 bread wheat varieties. In total, five unique genetic loci associated with Ni accumulation were identified and they explained, on average, 8.20-11.29% of the phenotypic variation. Among them, three unique genetic loci were mutually verified by different statistical models in at least two environments, indicating their stability across different environments. Moreover, the highest effect quantitative trait nucleotide (QTN) AX-111126872 with a quantitative trait locus (QTL) hotspot on chromosome 6B identified in this study was not reported previously. Three putative candidate genes linked to Ni accumulation were revealed from the stable genetic loci. Among them, one gene associated with the stable genetic locus on chromosome 6B (AX-111126872) encodes the glycine-rich proteins (GRPs) as a critical factor influencing Ni accumulation in wheat grains. This study increases our understanding of the genetic architecture of Ni accumulation in wheat grains, which is potentially helpful for breeding wheat varieties without Ni toxicity.
Collapse
Affiliation(s)
- Xia Shi
- Henan Institute of Crop Molecular Breeding, Henan Academy of Agricultural Sciences, Henan Key Laboratory of Wheat Biology, National Engineering Laboratory of Wheat Key Laboratory of Wheat Biology and Genetic Breeding in Central Huanghuai Area, Ministry of Agriculture, Zhengzhou 450002, China; (X.S.); (S.G.); (J.H.); (T.S.); (M.Q.); (W.L.); (Z.D.); (Z.Z.)
| | - Shenghui Geng
- Henan Institute of Crop Molecular Breeding, Henan Academy of Agricultural Sciences, Henan Key Laboratory of Wheat Biology, National Engineering Laboratory of Wheat Key Laboratory of Wheat Biology and Genetic Breeding in Central Huanghuai Area, Ministry of Agriculture, Zhengzhou 450002, China; (X.S.); (S.G.); (J.H.); (T.S.); (M.Q.); (W.L.); (Z.D.); (Z.Z.)
| | - Jinna Hou
- Henan Institute of Crop Molecular Breeding, Henan Academy of Agricultural Sciences, Henan Key Laboratory of Wheat Biology, National Engineering Laboratory of Wheat Key Laboratory of Wheat Biology and Genetic Breeding in Central Huanghuai Area, Ministry of Agriculture, Zhengzhou 450002, China; (X.S.); (S.G.); (J.H.); (T.S.); (M.Q.); (W.L.); (Z.D.); (Z.Z.)
| | - Taotao Shi
- Henan Institute of Crop Molecular Breeding, Henan Academy of Agricultural Sciences, Henan Key Laboratory of Wheat Biology, National Engineering Laboratory of Wheat Key Laboratory of Wheat Biology and Genetic Breeding in Central Huanghuai Area, Ministry of Agriculture, Zhengzhou 450002, China; (X.S.); (S.G.); (J.H.); (T.S.); (M.Q.); (W.L.); (Z.D.); (Z.Z.)
| | - Maomao Qin
- Henan Institute of Crop Molecular Breeding, Henan Academy of Agricultural Sciences, Henan Key Laboratory of Wheat Biology, National Engineering Laboratory of Wheat Key Laboratory of Wheat Biology and Genetic Breeding in Central Huanghuai Area, Ministry of Agriculture, Zhengzhou 450002, China; (X.S.); (S.G.); (J.H.); (T.S.); (M.Q.); (W.L.); (Z.D.); (Z.Z.)
| | - Wenxu Li
- Henan Institute of Crop Molecular Breeding, Henan Academy of Agricultural Sciences, Henan Key Laboratory of Wheat Biology, National Engineering Laboratory of Wheat Key Laboratory of Wheat Biology and Genetic Breeding in Central Huanghuai Area, Ministry of Agriculture, Zhengzhou 450002, China; (X.S.); (S.G.); (J.H.); (T.S.); (M.Q.); (W.L.); (Z.D.); (Z.Z.)
| | - Ziju Dai
- Henan Institute of Crop Molecular Breeding, Henan Academy of Agricultural Sciences, Henan Key Laboratory of Wheat Biology, National Engineering Laboratory of Wheat Key Laboratory of Wheat Biology and Genetic Breeding in Central Huanghuai Area, Ministry of Agriculture, Zhengzhou 450002, China; (X.S.); (S.G.); (J.H.); (T.S.); (M.Q.); (W.L.); (Z.D.); (Z.Z.)
| | - Zhengfu Zhou
- Henan Institute of Crop Molecular Breeding, Henan Academy of Agricultural Sciences, Henan Key Laboratory of Wheat Biology, National Engineering Laboratory of Wheat Key Laboratory of Wheat Biology and Genetic Breeding in Central Huanghuai Area, Ministry of Agriculture, Zhengzhou 450002, China; (X.S.); (S.G.); (J.H.); (T.S.); (M.Q.); (W.L.); (Z.D.); (Z.Z.)
| | | | - Zhensheng Lei
- Henan Institute of Crop Molecular Breeding, Henan Academy of Agricultural Sciences, Henan Key Laboratory of Wheat Biology, National Engineering Laboratory of Wheat Key Laboratory of Wheat Biology and Genetic Breeding in Central Huanghuai Area, Ministry of Agriculture, Zhengzhou 450002, China; (X.S.); (S.G.); (J.H.); (T.S.); (M.Q.); (W.L.); (Z.D.); (Z.Z.)
| |
Collapse
|
17
|
Mhatre S, Dutta D, Iyer A, Gholap S, Mishra A, Krishnatreya M, George GS, Doibale PN, Dun Y, Wang Z, Jahagirdar O, Chaturvedi P, Rajaraman P, Wang CP, Chaturvedi A, Kar S, Dikshit R, Chatterjee N. A Genome-wide Association study of Buccal Mucosa Cancer in India and Multi-ancestry Meta-analysis Identifies Novel Risk Loci and Gene-environment Interactions. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.16.25325815. [PMID: 40321259 PMCID: PMC12047951 DOI: 10.1101/2025.04.16.25325815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Genome-wide association studies (GWAS) of oral cancers (OC) to date have focused predominantly on European Ancestry (EA) populations. India faces an excess burden of OC, but the most common site of occurrence is the cancer of the buccal mucosa, which is relatively rare in EA populations. We conducted a GWAS of buccal mucosa cancer (BMC) comprising 2,160 BMC cases and 2,325 controls from different geographical locations in India. Single-SNP association tests detected one novel locus (6q27) and one novel signal within the known OC risk locus 5p13.33, at the genome-wide significance level (P-value<5 × 10-8). We additionally conducted a GWAS of 397 BMC cases and 439 controls from Taiwan and performed multi-ancestry GWAS meta-analysis of OC on 5255 cases and 8748 controls across EA, Indian and Taiwanese populations. We identified a novel risk locus harbouring the tumour suppressor gene NOTCH1 through a gene-level analysis of the multi-ancestry GWAS data. Pathway analysis suggested that PD-1 signalling, and Interferon Gamma Signalling may be important in the aetiology of BMC. Within data from the Indian BMC GWAS, we further identified statistically significant evidence of both multiplicative interactions (P-value=0.026) indicating stronger polygenic risk of BMC among individuals with history of chewing tobacco compared to those without. Our study provides insights into the etiologies of BMC in India, highlighting both its similarities and differences with other types of oral cavity cancers, as well as the interactions between polygenic gene score and tobacco chewing.
Collapse
Affiliation(s)
- Sharayu Mhatre
- Division of Molecular Epidemiology and Population Genomics, Centre for Cancer Epidemiology, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Diptavo Dutta
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
| | - Anand Iyer
- Division of Molecular Epidemiology and Population Genomics, Centre for Cancer Epidemiology, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Shruti Gholap
- Division of Molecular Epidemiology and Population Genomics, Centre for Cancer Epidemiology, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Aseem Mishra
- Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
- Department of Head and Neck Surgical Oncology, Mahamana Pandit Madan Mohan Malviya Cancer Centre, Varanasi, Uttar Pradesh, India
| | - Manigreeva Krishnatreya
- Department of Cancer Registry and Epidemiology, Dr. Bhubaneswar Borooah Cancer Institute, Tata Memorial Centre, Guwahati, Assam, India
| | - Grace Sarah George
- Division of Molecular Epidemiology and Population Genomics, Centre for Cancer Epidemiology, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Pravin Narayanrao Doibale
- Division of Molecular Epidemiology and Population Genomics, Centre for Cancer Epidemiology, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Yuzheng Dun
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ziqiao Wang
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Om Jahagirdar
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
| | - Pankaj Chaturvedi
- Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
- Department of Head and Neck Oncology, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Preetha Rajaraman
- Radiation Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
| | | | - Anil Chaturvedi
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland, USA
| | - Siddhatha Kar
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Rajesh Dikshit
- Division of Molecular Epidemiology and Population Genomics, Centre for Cancer Epidemiology, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Nilanjan Chatterjee
- Department of Head and Neck Oncology, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
18
|
Lange LM, Cerquera-Cleves C, Schipper M, Panagiotaropoulou G, Braun A, Kraft J, Awasthi S, Bell N, Posthuma D, Ripke S, Blauwendraat C, Heilbron K. Prioritizing Parkinson's disease risk genes in genome-wide association loci. NPJ Parkinsons Dis 2025; 11:77. [PMID: 40240380 PMCID: PMC12003903 DOI: 10.1038/s41531-025-00933-0] [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: 01/03/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Many drug targets in ongoing Parkinson's disease (PD) clinical trials have strong genetic links. While genome-wide association studies (GWAS) nominate regions associated with disease, pinpointing causal genes is challenging. Our aim was to prioritize additional druggable genes underlying PD GWAS signals. The polygenic priority score (PoPS) integrates genome-wide information from MAGMA gene-level associations and over 57,000 gene-level features. We applied PoPS to East Asian and European PD GWAS data and prioritized genes based on PoPS, distance to the GWAS signal, and non-synonymous credible set variants. We prioritized 46 genes, including well-established PD genes (SNCA, LRRK2, GBA1, TMEM175, VPS13C), genes with strong literature evidence supporting a mechanistic link to PD (RIT2, BAG3, SCARB2, FYN, DYRK1A, NOD2, CTSB, SV2C, ITPKB), and genes relatively unexplored in PD. Many hold potential for drug repurposing or development. We prioritized high-confidence genes with strong links to PD pathogenesis that may represent our next-best candidates for developing disease-modifying therapeutics.
Collapse
Affiliation(s)
- Lara M Lange
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Institute of Neurogenetics, University of Luebeck, Luebeck, Germany
| | - Catalina Cerquera-Cleves
- Neurology Unit, Department of Neurosciences, Hospital Universitario San Ignacio, Bogotá, Colombia
- Centre de recherche du Centre Hospitalier Universitaire de Québec, Axe Neurosciences, Département de Psychiatrie et Neurosciences, Laval University, Québec, QC, Canada
| | | | - Georgia Panagiotaropoulou
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Alice Braun
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Julia Kraft
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Swapnil Awasthi
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Nathaniel Bell
- Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Danielle Posthuma
- Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Child and Adolescent Psychiatry and Pediatric Psychology, Section Complex Trait Genetics, Amsterdam Neuroscience, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | - Stephan Ripke
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany
| | - Cornelis Blauwendraat
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Center for Alzheimer's and Related Dementias, National Institute on Aging and National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Karl Heilbron
- Department of Psychiatry and Psychotherapy, Charité-Universitätsmedizin Berlin, Berlin, Germany.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- German Center for Mental Health (DZPG), partner site Berlin/Potsdam, Berlin, Germany.
- Bayer AG, Research & Development, Pharmaceuticals, Berlin, Germany.
| |
Collapse
|
19
|
Venkatesh SS, Wittemans LBL, Palmer DS, Baya NA, Ferreira T, Hill B, Lassen FH, Parker MJ, Reibe S, Elhakeem A, Banasik K, Bruun MT, Erikstrup C, Aagard Jensen B, Juul A, Mikkelsen C, Nielsen HS, Ostrowski SR, Pedersen OB, Rohde PD, Sørensen E, Ullum H, Westergaard D, Haraldsson A, Holm H, Jonsdottir I, Olafsson I, Steingrimsdottir T, Steinthorsdottir V, Thorleifsson G, Figueredo J, Karjalainen MK, Pasanen A, Jacobs BM, Kalantzis G, Hubers N, Lippincott M, Fraser A, Lawlor DA, Timpson NJ, Nyegaard M, Stefansson K, Magi R, Laivuori H, van Heel DA, Boomsma DI, Balasubramanian R, Seminara SB, Chan YM, Laisk T, Lindgren CM. Genome-wide analyses identify 25 infertility loci and relationships with reproductive traits across the allele frequency spectrum. Nat Genet 2025:10.1038/s41588-025-02156-8. [PMID: 40229599 DOI: 10.1038/s41588-025-02156-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 03/07/2025] [Indexed: 04/16/2025]
Abstract
Genome-wide association studies (GWASs) may help inform the etiology of infertility. Here, we perform GWAS meta-analyses across seven cohorts in up to 42,629 cases and 740,619 controls and identify 25 genetic risk loci for male and female infertility. We additionally identify up to 269 genetic loci associated with follicle-stimulating hormone, luteinizing hormone, estradiol and testosterone through sex-specific GWAS meta-analyses (n = 6,095-246,862). Exome sequencing analyses reveal that women carrying testosterone-lowering rare variants in some genes are at risk of infertility. However, we find no local or genome-wide genetic correlation between female infertility and reproductive hormones. While infertility is genetically correlated with endometriosis and polycystic ovary syndrome, we find limited genetic overlap between infertility and obesity. Finally, we show that the evolutionary persistence of infertility-risk alleles may be explained by directional selection. Taken together, we provide a comprehensive view of the genetic determinants of infertility across multiple diagnostic criteria.
Collapse
Affiliation(s)
- Samvida S Venkatesh
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK.
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Laura B L Wittemans
- Novo Nordisk Research Centre Oxford, Oxford, UK
- Nuffield Department of Women's and Reproductive Health, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Duncan S Palmer
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Nuffield Department of Population Health, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Nikolas A Baya
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Teresa Ferreira
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Barney Hill
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Nuffield Department of Population Health, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Frederik Heymann Lassen
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Melody J Parker
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Nuffield Department of Clinical Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Saskia Reibe
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Nuffield Department of Population Health, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Ahmed Elhakeem
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
- Department of Obstetrics and Gynecology, Copenhagen University Hospital, Hvidovre, Copenhagen, Denmark
| | - Mie T Bruun
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Health, Aarhus University, Aarhus, Denmark
| | - Bitten Aagard Jensen
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | - Anders Juul
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Growth and Reproduction, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Christina Mikkelsen
- Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Science, Copenhagen University, Copenhagen, Denmark
| | - Henriette S Nielsen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Obstetrics and Gynecology, The Fertility Clinic, Hvidovre University Hospital, Copenhagen, Denmark
| | - Sisse R Ostrowski
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Ole B Pedersen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Immunology, Zealand University Hospital-Køge, Køge, Denmark
| | - Palle Duun Rohde
- Genomic Medicine, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Erik Sørensen
- Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | | | - David Westergaard
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
- Department of Obstetrics and Gynecology, Copenhagen University Hospital, Hvidovre, Copenhagen, Denmark
| | - Asgeir Haraldsson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Children's Hospital Iceland, Landspitali University Hospital, Reykjavik, Iceland
| | - Hilma Holm
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
| | - Ingileif Jonsdottir
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
| | - Isleifur Olafsson
- Department of Clinical Biochemistry, Landspitali University Hospital, Reykjavik, Iceland
| | - Thora Steingrimsdottir
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Department of Obstetrics and Gynecology, Landspitali University Hospital, Reykjavik, Iceland
| | | | | | - Jessica Figueredo
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Minna K Karjalainen
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Northern Finland Birth Cohorts, Arctic Biobank, Infrastructure for Population Studies, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Anu Pasanen
- Research Unit of Clinical Medicine, Medical Research Center Oulu, University of Oulu, and Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| | - Benjamin M Jacobs
- Centre for Preventive Neurology, Wolfson Institute of Population Health, Queen Mary University London, London, UK
| | | | - Nikki Hubers
- Department of Biological Psychology, Netherlands Twin Register, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Institute, Amsterdam, The Netherlands
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, The Netherlands
| | - Margaret Lippincott
- Reproductive Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Abigail Fraser
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Mette Nyegaard
- Genomic Medicine, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Statens Serum Institut, Copenhagen, Denmark
| | - Kari Stefansson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
| | - Reedik Magi
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Hannele Laivuori
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital, The Wellbeing Services County of Pirkanmaa, Tampere, Finland
- Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | | | - Dorret I Boomsma
- Amsterdam Reproduction and Development Institute, Amsterdam, The Netherlands
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, The Netherlands
| | - Ravikumar Balasubramanian
- Reproductive Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Stephanie B Seminara
- Reproductive Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yee-Ming Chan
- Harvard Medical School, Boston, MA, USA
- Division of Endocrinology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - Triin Laisk
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Cecilia M Lindgren
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK.
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Nuffield Department of Women's and Reproductive Health, Medical Sciences Division, University of Oxford, Oxford, UK.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
20
|
Petty LE, Chen HH, Frankel EG, Zhu W, Downie CG, Graff M, Lin P, Sharma P, Zhang X, Scartozzi AC, Roshani R, Landman JM, Boehnke M, Bowden DW, Chambers JC, Mahajan A, McCarthy MI, Ng MCY, Sim X, Spracklen CN, Zhang W, Preuss M, Bottinger EP, Nadkarni GN, Loos RJF, Chen YDI, Tan J, Ipp E, Genter P, Emery LS, Louie T, Sofer T, Stilp AM, Taylor KD, Xiang AH, Buchanan TA, Roll K, Gao C, Palmer ND, Norris JM, Wagenknecht LE, Nousome D, Varma R, McKean-Cowdin R, Guo X, Hai Y, Hsueh W, Sandow K, Parra EJ, Cruz M, Valladares-Salgado A, Wacher-Rodarte N, Rotter JI, Goodarzi MO, Rich SS, Bertoni A, Raffel LJ, Nadler JL, Kandeel FR, Duggirala R, Blangero J, Lehman DM, DeFronzo RA, Thameem F, Wang Y, Gahagan S, Blanco E, Burrows R, Huerta-Chagoya A, Florez JC, Tusie-Luna T, González-Villalpando C, Orozco L, Haiman CA, Hanis CL, Rohde R, Whitsel EA, Reiner AP, Kooperberg C, Li Y, Duan Q, Lee M, Correa-Burrows P, Fried SK, North KE, McCormick JB, Fisher-Hoch SP, Gamazon ER, Morris AP, Mercader JM, Highland HM, Below JE. Large-scale multi-omics analyses in Hispanic/Latino populations identify genes for cardiometabolic traits. Nat Commun 2025; 16:3438. [PMID: 40210677 PMCID: PMC11985957 DOI: 10.1038/s41467-025-58574-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: 03/28/2024] [Accepted: 03/21/2025] [Indexed: 04/12/2025] Open
Abstract
Here, we present a multi-omics study of type 2 diabetes and quantitative blood lipid and lipoprotein traits conducted to date in Hispanic/Latino populations (nmax = 63,184). We conduct a meta-analysis of 16 type 2 diabetes and 19 lipid trait GWAS, identifying 20 genome-wide significant loci for type 2 diabetes, including one novel locus and novel signals at two known loci, based on fine-mapping. We also identify sixty-one genome-wide significant loci across the lipid/lipoprotein traits, including nine novel loci, and novel signals at 19 known loci through fine-mapping. Next, we analyze genetically regulated expression, perform Mendelian randomization, and analyze association with transcriptomic and proteomic measure using multi-omics data from a Hispanic/Latino population. Using this approach, we identify genes linked to type 2 diabetes and lipid/lipoprotein traits, including TMEM205 and NEDD9 for HDL cholesterol, TREH for triglycerides, and ANXA4 for type 2 diabetes.
Collapse
Affiliation(s)
- Lauren E Petty
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hung-Hsin Chen
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
| | - Elizabeth G Frankel
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wanying Zhu
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Carolina G Downie
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mariaelisa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Phillip Lin
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Priya Sharma
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xinruo Zhang
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alyssa C Scartozzi
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rashedeh Roshani
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joshua M Landman
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Donald W Bowden
- Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - John C Chambers
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, London North West Healthcare NHS Trust, London, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, Imperial College London, London, UK
| | - Anubha Mahajan
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mark I McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Maggie C Y Ng
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xueling Sim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Cassandra N Spracklen
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Epidemiology and Biostatistics, University of Massachusetts-Amherst, Amherst, MA, USA
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, London North West Healthcare NHS Trust, London, UK
| | - Michael Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Erwin P Bottinger
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Girish N Nadkarni
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Digital Health Center, Hasso-Plattner-Institut, Potsdam, Germany
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yii-Der Ida Chen
- 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
| | - Jingyi Tan
- 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
| | - Eli Ipp
- Division of Endocrinology & Metabolism, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Pauline Genter
- Division of Endocrinology & Metabolism, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Leslie S Emery
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Tin Louie
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Tamar Sofer
- Departments of Medicine and Biostatistics, Harvard University, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Adrienne M Stilp
- Department of Biostatistics, University of Washington, Seattle, WA, 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
| | - Anny H Xiang
- Department of Research & Evaluation, Division of Biostatistics Research, Kaiser Permanente of Southern California, Pasadena, CA, USA
| | - Thomas A Buchanan
- Department of Medicine, Division of Endocrine, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Kathryn Roll
- 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
| | - Chuan Gao
- Molecular Genetics and Genomics Program, Wake Forest School of Medicine, Winston-, Salem, NC, USA
| | - Nicholette D Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jill M Norris
- Department of Epidemiology, University of Colorado Denver, Aurora, CO, USA
| | - Lynne E Wagenknecht
- Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Darryl Nousome
- Department of Preventative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Rohit Varma
- Southern California Eye Institute, CHA Hollywood Presbyterian Medical Center, Los Angeles, CA, USA
| | - Roberta McKean-Cowdin
- Department of Preventative Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, 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
| | - Yang Hai
- 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
| | - Willa Hsueh
- Department of Internal Medicine, Diabetes and Metabolism Research Center, Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Kevin Sandow
- 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
| | - Esteban J Parra
- Department of Anthropology, University of Toronto at Mississauga, Mississauga, ON, Canada
| | - Miguel Cruz
- Unidad de Investigación Médica en Bioquimica, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, IMSS, Mexico City, Mexico
| | - Adan Valladares-Salgado
- Unidad de Investigación Médica en Bioquimica, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, IMSS, Mexico City, Mexico
| | - Niels Wacher-Rodarte
- Unidad de Investigación Médica en Epidemiologia Clinica, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, IMSS, Mexico City, Mexico
| | - 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
| | - Mark O Goodarzi
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Alain Bertoni
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Leslie J Raffel
- Department of Pediatrics, Division of Medical Genetics, UCI Irvine School of Medicine, Irvine, CA, USA
| | - Jerry L Nadler
- Department of Medicine and Pharmacology, New York Medical College, Valhalla, NY, USA
| | - Fouad R Kandeel
- Departments of Clinical Diabetes, Endocrinology & Metabolism and Translational Research and Cellular Therapeutics, City of Hope, Duarte, CA, USA
| | | | - John Blangero
- Human Genetics and STDOI, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Donna M Lehman
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Ralph A DeFronzo
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Farook Thameem
- Department of Biochemistry, Faculty of Medicine, Health Science Center, Kuwait University, Safat, Kuwait
| | - Yujie Wang
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sheila Gahagan
- Division of Academic General Pediatrics, Child Development and Community Health, University of California at San Diego, San Diego, CA, USA
| | - Estela Blanco
- College y Escuela de Salud Pública, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Raquel Burrows
- Department of Public Health Nutrition, Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| | - Alicia Huerta-Chagoya
- Consejo Nacional de Ciencia y Tecnología (CONACYT), Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jose C Florez
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Teresa Tusie-Luna
- Unidad de Biología Molecular y Medicina Genómica, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México/Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Clicerio González-Villalpando
- Centro de Estudios en Diabetes, Unidad de Investigacion en Diabetes y Riesgo Cardiovascular, Centro de Investigacion en Salud Poblacional, Instituto Nacional de Salud Pública, Mexico City, Mexico
| | - Lorena Orozco
- Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Christopher A Haiman
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Craig L Hanis
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Rebecca Rohde
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Eric A Whitsel
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexander P Reiner
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Yun Li
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Qing Duan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Miryoung Lee
- Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Brownsville Regional Campus, Brownsville, TX, USA
| | - Paulina Correa-Burrows
- Department of Public Health Nutrition, Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| | - Susan K Fried
- Diabetes, Obesity Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joseph B McCormick
- Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Brownsville Regional Campus, Brownsville, TX, USA
| | - Susan P Fisher-Hoch
- Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Brownsville Regional Campus, Brownsville, TX, USA
| | - Eric R Gamazon
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Clare Hall & MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, University of Manchester, Manchester, UK
| | - Josep M Mercader
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Heather M Highland
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jennifer E Below
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
21
|
Tan D, Chen Y, Ilboudo Y, Liang KYH, Butler-Laporte G, Richards JB. Caution when using network partners for target identification in drug discovery. HGG ADVANCES 2025; 6:100409. [PMID: 39861949 PMCID: PMC11850190 DOI: 10.1016/j.xhgg.2025.100409] [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: 08/28/2024] [Revised: 01/21/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025] Open
Abstract
Identifying novel, high-yield drug targets is challenging and often results in a high failure rate. However, recent data indicate that leveraging human genetic evidence to identify and validate these targets significantly increases the likelihood of success in drug development. Two recent papers from Open Targets claimed that around half of US Food and Drug Administration-approved drugs had targets with direct human genetic evidence. By expanding target identification to include protein network partners-molecules in physical contact-the proportion of drug targets with genetic evidence support increased to two-thirds. However, the efficacy of using these network partners for target identification was not formally tested. To address this, we tested the approach on a list of robust positive control genes. We used the IntAct database to find physically interacting proteins of genes identified by exome-wide association studies (ExWASs), genome-wide association studies (GWASs) combined with a locus-to-gene mapping algorithm called the Effector Index, and Genetic Priority Score (GPS), which integrated eight genetic features with drug indications from the Open Targets and SIDER databases. We assessed how accurately including interacting genes with the ExWAS-, Effector Index-, and GPS-selected genes identified positive controls, focusing on precision, sensitivity, and specificity. Our results indicated that although molecular interactions led to higher sensitivity in identifying positive control genes, their practical application is limited by low precision. Expanding genetically identified targets to include network partners using IntAct did not increase the likelihood of identifying drug targets across the 412 tested traits, suggesting that such results should be interpreted with caution.
Collapse
Affiliation(s)
- Dandan Tan
- Quantitative Life Sciences Program, McGill University, Montréal, QC, Canada; Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, QC, Canada
| | - Yiheng Chen
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, QC, Canada; Department of Human Genetics, McGill University, Montréal, QC, Canada; 5Prime Sciences, Montréal, QC, Canada
| | - Yann Ilboudo
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, QC, Canada
| | - Kevin Y H Liang
- Quantitative Life Sciences Program, McGill University, Montréal, QC, Canada; Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, QC, Canada
| | - Guillaume Butler-Laporte
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, QC, Canada; Division of Infectious Diseases, McGill University, Montréal, QC, Canada
| | - J Brent Richards
- Lady Davis Institute, Jewish General Hospital, McGill University, Montréal, QC, Canada; Department of Human Genetics, McGill University, Montréal, QC, Canada; 5Prime Sciences, Montréal, QC, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, QC, Canada; Department of Medicine, McGill University, Montréal, QC, Canada; Department of Twin Research, King's College London, London, UK.
| |
Collapse
|
22
|
Mechelli R, Umeton R, Bellucci G, Bigi R, Rinaldi V, Angelini DF, Guerrera G, Pignalosa FC, Ilari S, Patrone M, Srinivasan S, Cerono G, Romano S, Buscarinu MC, Martire S, Malucchi S, Landi D, Lorefice L, Pizzolato Umeton R, Anastasiadou E, Trivedi P, Fornasiero A, Ferraldeschi M, IMSGC WTCCC2, Di Sapio A, Marfia G, Cocco E, Centonze D, Uccelli A, Di Silvestre D, Mauri P, de Candia P, D’Alfonso S, Battistini L, Farina C, Magliozzi R, Reynolds R, Baranzini SE, Matarese G, Salvetti M, Ristori G. A disease-specific convergence of host and Epstein-Barr virus genetics in multiple sclerosis. Proc Natl Acad Sci U S A 2025; 122:e2418783122. [PMID: 40184175 PMCID: PMC12002260 DOI: 10.1073/pnas.2418783122] [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: 09/14/2024] [Accepted: 01/27/2025] [Indexed: 04/05/2025] Open
Abstract
Recent sero-epidemiological studies have strengthened the hypothesis that Epstein-Barr virus (EBV) may be a causal factor in multiple sclerosis (MS). Given the complexity of the EBV-host interaction, various mechanisms may be responsible for the disease pathogenesis. Furthermore, it remains unclear whether this is a disease-specific process. Here, we showed that genes encoding EBV interactors are enriched in loci associated with MS but not with other diseases and in prioritized therapeutic targets. Analyses of MS blood and brain transcriptomes confirmed a dysregulation of MS-associated EBV interactors affecting the CD40 pathway. Such interactors were strongly enriched in binding sites for the EBV nuclear antigen 2 (EBNA2) viral transcriptional regulator, often in colocalization with CCCTC binding factor (CTCF) and RNA Polymerase II Subunit A (POLR2A). EBNA2 was expressed in the MS brain. The 1.2 EBNA2 allele downregulated the expression of the CD40 MS-associated gene analogously to the CD40 MS-risk variant. Finally, we showed that the 1.2 EBNA2 allele associates with the risk of MS. This study delineates how host and viral genetic variability converge in MS-specific pathogenetic mechanisms.
Collapse
Affiliation(s)
- Rosella Mechelli
- Department for the Promotion of Human Sciences and Quality of Life, San Raffaele Roma University, Rome, Italy
- Istituto Ricovero e Cura a Carattere Scientifico San Raffaele, Rome00166, Italy
| | - Renato Umeton
- Weill Cornell Medicine, New York, NY10021
- Massachusetts Institute of Technology, Cambridge, MA02139
- Harvard T. H. Chan School of Public Health, Boston, MA02115
| | - Gianmarco Bellucci
- Centre for Experimental Neurological Therapies, Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University, Rome00189, Italy
| | - Rachele Bigi
- Centre for Experimental Neurological Therapies, Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University, Rome00189, Italy
| | - Virginia Rinaldi
- Centre for Experimental Neurological Therapies, Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University, Rome00189, Italy
| | - Daniela F. Angelini
- Neuroimmunology Unit, Istituto Ricovero e Cura a Carattere Scientifico Fondazione Santa Lucia, Rome00179, Italy
| | - Gisella Guerrera
- Neuroimmunology Unit, Istituto Ricovero e Cura a Carattere Scientifico Fondazione Santa Lucia, Rome00179, Italy
| | - Francesca C. Pignalosa
- Department for the Promotion of Human Sciences and Quality of Life, San Raffaele Roma University, Rome, Italy
- Istituto Ricovero e Cura a Carattere Scientifico San Raffaele, Rome00166, Italy
| | - Sara Ilari
- Istituto Ricovero e Cura a Carattere Scientifico San Raffaele, Rome00166, Italy
| | - Marco Patrone
- Biocrystallography Unit, Division of Immunology, Transplantation, and Infectious Diseases, Istituto Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan20132, Italy
| | - Sundararajan Srinivasan
- Institute of Experimental Neurology & Division of Neuroscience, Istituto Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan20132, Italy
| | - Gabriel Cerono
- Department of Neurology, University of California San Francisco Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA94158
| | - Silvia Romano
- Centre for Experimental Neurological Therapies, Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University, Rome00189, Italy
| | - Maria C. Buscarinu
- Centre for Experimental Neurological Therapies, Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University, Rome00189, Italy
| | - Serena Martire
- Clinical Neurobiology Unit, Neuroscience Institute Cavalieri Ottolenghi, Orbassano10043, Italy
- Centri di Riferimento Regionale Sclerosi Multipla Biobank, University Hospital San Luigi Gonzaga, Orbassano10043, Italy
- Department of Neuroscience “Rita Levi Montalcini”, University of Turin, Turin10043, Italy
| | - Simona Malucchi
- Department of Neurology and Centri di Riferimento Regionale Sclerosi Multipla, University Hospital San Luigi Gonzaga, Orbassano10043, Italy
| | - Doriana Landi
- Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Tor Vergata University, Rome00133, Italy
| | - Lorena Lorefice
- Multiple Sclerosis Center, Binaghi Hospital, Azienda Sanitaria Locale Cagliari, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari09126, Italy
| | - Raffaella Pizzolato Umeton
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA01655
- Department of Neurology, Massachusetts General Hospital, Boston, MA02114
- Harvard Medical School, Boston, MA02115
| | - Eleni Anastasiadou
- Department of Clinical and Molecular Medicine, Sapienza University, Rome00189, Italy
| | - Pankaj Trivedi
- Department of Experimental Medicine, Sapienza University, Rome00161, Italy
| | - Arianna Fornasiero
- Centre for Experimental Neurological Therapies, Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University, Rome00189, Italy
| | - Michela Ferraldeschi
- Centre for Experimental Neurological Therapies, Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University, Rome00189, Italy
| | - IMSGC WTCCC2
- The International Multiple Sclerosis Genetics Consortium and the Wellcome Trust Case Control Consortium 2, Oxford OX1 2JD, United Kingdom
| | - Alessia Di Sapio
- Clinical Neurobiology Unit, Neuroscience Institute Cavalieri Ottolenghi, Orbassano10043, Italy
- Centri di Riferimento Regionale Sclerosi Multipla Biobank, University Hospital San Luigi Gonzaga, Orbassano10043, Italy
- Department of Neurology and Centri di Riferimento Regionale Sclerosi Multipla, University Hospital San Luigi Gonzaga, Orbassano10043, Italy
| | - Gerolama Marfia
- Multiple Sclerosis Clinical and Research Unit, Department of Systems Medicine, Tor Vergata University, Rome00133, Italy
| | - Eleonora Cocco
- Multiple Sclerosis Center, Binaghi Hospital, Azienda Sanitaria Locale Cagliari, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari09126, Italy
| | - Diego Centonze
- Department of Systems Medicine, Tor Vergata University, Rome00133, Italy
- Istituto Ricovero e Cura a Carattere Scientifico Istituto Neurologico Mediterraneo Neuromed, Pozzilli-86077, Italy
| | - Antonio Uccelli
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health and Centre of Excellence for Biomedical Research, University of Genova, Genoa16132, Italy
- Ospedale Policlinico San Martino, Istituto Ricovero e Cura a Carattere Scientifico, Genoa16132, Italy
| | - Dario Di Silvestre
- Elixir Proteomics Laboratory, Istituto di Tecnologie Biomediche, Consiglio Nazionale delle Ricerche, Segrate, Milan20054, Italy
| | - Pierluigi Mauri
- Elixir Proteomics Laboratory, Istituto di Tecnologie Biomediche, Consiglio Nazionale delle Ricerche, Segrate, Milan20054, Italy
| | - Paola de Candia
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli “Federico II”, Napoli80131, Italy
| | - Sandra D’Alfonso
- Department of Health Sciences, University of Eastern Piedmont, Interdisciplinary Research Center of Autoimmune Diseases, Novara28100, Italy
| | - Luca Battistini
- Neuroimmunology Unit, Istituto Ricovero e Cura a Carattere Scientifico Fondazione Santa Lucia, Rome00179, Italy
| | - Cinthia Farina
- Institute of Experimental Neurology & Division of Neuroscience, Istituto Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan20132, Italy
| | - Roberta Magliozzi
- Neurology B, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona37134, Italy
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, LondonW12 ONN, United Kingdom
| | - Richard Reynolds
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, LondonW12 ONN, United Kingdom
| | - Sergio E. Baranzini
- Department of Neurology, University of California San Francisco Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA94158
| | - Giuseppe Matarese
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli “Federico II”, Napoli80131, Italy
- Laboratorio di Immunologia, Istituto di Endocrinologia e Oncologia Sperimentale, Consiglio Nazionale delle Ricerche, Naples80131, Italy
| | - Marco Salvetti
- Centre for Experimental Neurological Therapies, Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University, Rome00189, Italy
- Istituto Ricovero e Cura a Carattere Scientifico Istituto Neurologico Mediterraneo Neuromed, Pozzilli-86077, Italy
| | - Giovanni Ristori
- Centre for Experimental Neurological Therapies, Department of Neurosciences, Mental Health and Sensory Organs, Sapienza University, Rome00189, Italy
| |
Collapse
|
23
|
Liu S, Zhang L, Luo W. Warfarin use and vestibular dysfunction insights from NHANES data, network pharmacology, Mendelian randomization, and molecular docking. Sci Rep 2025; 15:11748. [PMID: 40189685 PMCID: PMC11973179 DOI: 10.1038/s41598-025-96681-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 03/31/2025] [Indexed: 04/09/2025] Open
Abstract
Despite numerous anticoagulants available, warfarin is widely used due to its efficacy and cost-effectiveness in treating thromboembolic diseases. However, its potential impact on vestibular function remains unexplored. This study investigates the association between warfarin use and vestibular dysfunction using data from the NHANES database and examines underlying mechanisms through network pharmacology, Mendelian randomization (MR), and molecular docking. We conducted a cross-sectional analysis of NHANES data (1999-2004) to evaluate the prevalence of vestibular dysfunction among warfarin users. Network pharmacology identified overlapping genes between warfarin targets and vestibular dysfunction-related genes. MR analysis assessed the causal relationship, and molecular docking examined interactions between warfarin and significant genes. The study included 1681 participants, revealing a higher prevalence of vestibular dysfunction in warfarin users. Multiple regression analysis confirmed a significant association between warfarin use and vestibular dysfunction. Network pharmacology identified 31 overlapping genes, with MAPK8 emerging as a key gene through MR analysis. Molecular docking showed a strong binding affinity between warfarin and MAPK8. Findings suggest that warfarin use is significantly associated with vestibular dysfunction, potentially through interactions with MAPK8. This highlights the importance of monitoring vestibular function in patients on warfarin therapy and considering genetic factors to personalize treatment. Future research should explore these mechanisms further and validate findings in broader populations.
Collapse
Affiliation(s)
- Shihan Liu
- Department of Otorhinolaryngology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lingli Zhang
- Department of Otorhinolaryngology, Central Hospital Affiliated to Chongqing University of Technology, Chongqing, China.
| | - Wenlong Luo
- Department of Otorhinolaryngology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
24
|
Elgaeva EE, Zorkoltseva IV, Nostaeva AV, Verzun DA, Tiys ES, Timoshchuk AN, Kirichenko AV, Svishcheva GR, Freidin MB, Williams FMK, Suri P, Aulchenko YS, Axenovich TI, Tsepilov YA. Decomposing the genetic background of chronic back pain. Hum Mol Genet 2025; 34:711-725. [PMID: 39895344 DOI: 10.1093/hmg/ddae195] [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: 07/04/2024] [Revised: 10/09/2024] [Accepted: 12/16/2024] [Indexed: 02/04/2025] Open
Abstract
Chronic back pain (CBP) is a disabling condition with a lifetime prevalence of 40% and a substantial socioeconomic burden. Because of the high heterogeneity of CBP, subphenotyping may help to improve prediction and support personalized treatment of CBP. To investigate CBP subphenotypes, we decomposed its genetic background into a shared one common to other chronic pain conditions (back, neck, hip, knee, stomach, and head pain) and unshared genetic background specific to CBP. We identified and replicated 18 genes with shared impact across different chronic pain conditions and two genes that were specific for CBP. Among people with CBP, we demonstrated that polygenic risk scores accounting for the shared and unshared genetic backgrounds of CBP may underpin different CBP subphenotypes. These subphenotypes are characterized by varying genetic predisposition to diverse medical conditions and interventions such as diabetes mellitus, myocardial infarction, diagnostic endoscopic procedures, and surgery involving muscles, bones, and joints.
Collapse
Affiliation(s)
- Elizaveta E Elgaeva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10, Ac. Lavrentieva ave., 630090, Novosibirsk, Russia
- Novosibirsk State University, 1, Pirogova str., 630090, Novosibirsk, Russia
| | - Irina V Zorkoltseva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10, Ac. Lavrentieva ave., 630090, Novosibirsk, Russia
| | - Arina V Nostaeva
- Novosibirsk State University, 1, Pirogova str., 630090, Novosibirsk, Russia
| | - Dmitrii A Verzun
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10, Ac. Lavrentieva ave., 630090, Novosibirsk, Russia
- Novosibirsk State University, 1, Pirogova str., 630090, Novosibirsk, Russia
| | - Evgeny S Tiys
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10, Ac. Lavrentieva ave., 630090, Novosibirsk, Russia
| | - Anna N Timoshchuk
- MSU Institute for Artificial Intelligence, Lomonosov Moscow State University, 27, building 1, Lomonosovsky ave., 119991, Moscow, Russia
- Moscow Institute of Physics and Technology, 9, Institutsky lane, 141700, Dolgoprudny, Russia
| | - Anatoliy V Kirichenko
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10, Ac. Lavrentieva ave., 630090, Novosibirsk, Russia
| | - Gulnara R Svishcheva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10, Ac. Lavrentieva ave., 630090, Novosibirsk, Russia
- Vavilov Institute of General Genetics, RAS, 3, Gubkin str., 119991, Moscow, Russia
| | - Maxim B Freidin
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King's College London, Westminster Bridge Rd., SE1 7EH, London, UK
| | - Frances M K Williams
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King's College London, Westminster Bridge Rd., SE1 7EH, London, UK
| | - Pradeep Suri
- Department of Rehabilitation Medicine, University of Washington, 325, Ninth ave., WA 98104, Seattle, USA
- VA Puget Sound Health Care System, 1660, South Columbian Way, WA 98108, Seattle, USA
| | - Yurii S Aulchenko
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10, Ac. Lavrentieva ave., 630090, Novosibirsk, Russia
- PolyOmica, 61, Het Vlaggeschip, 5237 PA, 's-Hertogenbosch, The Netherlands
| | - Tatiana I Axenovich
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10, Ac. Lavrentieva ave., 630090, Novosibirsk, Russia
| | - Yakov A Tsepilov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10, Ac. Lavrentieva ave., 630090, Novosibirsk, Russia
| |
Collapse
|
25
|
Rios Coronado PE, Zhou J, Fan X, Zanetti D, Naftaly JA, Prabala P, Martínez Jaimes AM, Farah EN, Kundu S, Deshpande SS, Evergreen I, Kho PF, Ma Q, Hilliard AT, Abramowitz S, Pyarajan S, Dochtermann D, Damrauer SM, Chang KM, Levin MG, Winn VD, Paşca AM, Plomondon ME, Waldo SW, Tsao PS, Kundaje A, Chi NC, Clarke SL, Red-Horse K, Assimes TL. CXCL12 drives natural variation in coronary artery anatomy across diverse populations. Cell 2025; 188:1784-1806.e22. [PMID: 40049164 PMCID: PMC12029448 DOI: 10.1016/j.cell.2025.02.005] [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: 07/11/2024] [Revised: 10/22/2024] [Accepted: 02/06/2025] [Indexed: 03/12/2025]
Abstract
Coronary arteries have a specific branching pattern crucial for oxygenating heart muscle. Among humans, there is natural variation in coronary anatomy with respect to perfusion of the inferior/posterior left heart, which can branch from either the right arterial tree, the left, or both-a phenotype known as coronary dominance. Using angiographic data for >60,000 US veterans of diverse ancestry, we conducted a genome-wide association study of coronary dominance, revealing moderate heritability and identifying ten significant loci. The strongest association occurred near CXCL12 in both European- and African-ancestry cohorts, with downstream analyses implicating effects on CXCL12 expression. We show that CXCL12 is expressed in human fetal hearts at the time dominance is established. Reducing Cxcl12 in mice altered coronary dominance and caused septal arteries to develop away from Cxcl12 expression domains. These findings indicate that CXCL12 patterns human coronary arteries, paving the way for "medical revascularization" through targeting developmental pathways.
Collapse
Affiliation(s)
| | - Jiayan Zhou
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Xiaochen Fan
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Daniela Zanetti
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; VA Palo Alto Health Care System, Palo Alto, CA, USA; Institute of Genetic and Biomedical Research, National Research Council, Cagliari, Sardinia, Italy
| | | | - Pratima Prabala
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Azalia M Martínez Jaimes
- Department of Biology, Stanford University, Stanford, CA, USA; Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Elie N Farah
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA, USA
| | - Soumya Kundu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Salil S Deshpande
- Institute for Computational and Mathematical Engineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Ivy Evergreen
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Pik Fang Kho
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Qixuan Ma
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA, USA
| | | | - Sarah Abramowitz
- Department of Medicine, Division of Cardiovascular Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Sarnoff Cardiovascular Research Foundation, McLean, VA, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Saiju Pyarajan
- Center for Data and Computational Sciences, VA Boston Healthcare System, Boston, MA, USA
| | - Daniel Dochtermann
- Center for Data and Computational Sciences, VA Boston Healthcare System, Boston, MA, USA
| | - Scott M Damrauer
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA; Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kyong-Mi Chang
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA; Department of Medicine, Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael G Levin
- Department of Medicine, Division of Cardiovascular Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Virginia D Winn
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Anca M Paşca
- Department of Pediatrics, Neonatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Mary E Plomondon
- Department of Medicine, Rocky Mountain Regional VA Medical Center, Aurora, CO, USA; CART Program, VHA Office of Quality and Patient Safety, Washington, DC, USA
| | - Stephen W Waldo
- Department of Medicine, Rocky Mountain Regional VA Medical Center, Aurora, CO, USA; CART Program, VHA Office of Quality and Patient Safety, Washington, DC, USA; Division of Cardiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Philip S Tsao
- VA Palo Alto Health Care System, Palo Alto, CA, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Anshul Kundaje
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Neil C Chi
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA, USA
| | - Shoa L Clarke
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; VA Palo Alto Health Care System, Palo Alto, CA, USA; Department of Medicine, Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| | - Themistocles L Assimes
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; VA Palo Alto Health Care System, Palo Alto, CA, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
26
|
Long E, Williams J, Zhang H, Choi J. An evolving understanding of multiple causal variants underlying genetic association signals. Am J Hum Genet 2025; 112:741-750. [PMID: 39965570 DOI: 10.1016/j.ajhg.2025.01.018] [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: 09/22/2024] [Revised: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 02/20/2025] Open
Abstract
Understanding how genetic variation contributes to phenotypic variation is a fundamental question in genetics. Genome-wide association studies (GWASs) have discovered numerous genetic associations with various human phenotypes, most of which contain co-inherited variants in strong linkage disequilibrium (LD) with indistinguishable statistical significance. The experimental and analytical difficulty in identifying the "causal variant" among the co-inherited variants has traditionally led mechanistic studies to focus on relatively simple loci, where a single functional variant is presumed to explain most of the association signal and affect a target gene. The notion that a single causal variant is responsible for an association signal, while other variants in LD are merely correlated, has often been assumed in functional studies. However, emerging evidence powered by high-throughput experimental tools and context-specific functional databases argues that even a single independent signal may involve multiple functional variants in strong LD, each contributing to the observed genetic association. In this perspective, we articulate this evolving understanding of causal variants through examples from both traditional locus-by-locus approaches and more recent high-throughput functional studies. We then discuss the implications and prospects of this notion in understanding the genetic architecture of complex traits and interpreting the variant-level causality in GWAS follow-up studies.
Collapse
Affiliation(s)
- Erping Long
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jacob Williams
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Haoyu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jiyeon Choi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
27
|
Linder J, Srivastava D, Yuan H, Agarwal V, Kelley DR. Predicting RNA-seq coverage from DNA sequence as a unifying model of gene regulation. Nat Genet 2025; 57:949-961. [PMID: 39779956 PMCID: PMC11985352 DOI: 10.1038/s41588-024-02053-6] [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: 08/28/2023] [Accepted: 12/04/2024] [Indexed: 01/11/2025]
Abstract
Sequence-based machine-learning models trained on genomics data improve genetic variant interpretation by providing functional predictions describing their impact on the cis-regulatory code. However, current tools do not predict RNA-seq expression profiles because of modeling challenges. Here, we introduce Borzoi, a model that learns to predict cell-type-specific and tissue-specific RNA-seq coverage from DNA sequence. Using statistics derived from Borzoi's predicted coverage, we isolate and accurately score DNA variant effects across multiple layers of regulation, including transcription, splicing and polyadenylation. Evaluated on quantitative trait loci, Borzoi is competitive with and often outperforms state-of-the-art models trained on individual regulatory functions. By applying attribution methods to the derived statistics, we extract cis-regulatory motifs driving RNA expression and post-transcriptional regulation in normal tissues. The wide availability of RNA-seq data across species, conditions and assays profiling specific aspects of regulation emphasizes the potential of this approach to decipher the mapping from DNA sequence to regulatory function.
Collapse
Affiliation(s)
| | | | - Han Yuan
- Calico Life Sciences LLC, South San Francisco, CA, USA
| | - Vikram Agarwal
- mRNA Center of Excellence, Sanofi Pasteur Inc., Cambridge, MA, USA
| | | |
Collapse
|
28
|
Lu T, Manousaki D, Sun L, Paterson AD. Integrative Proteogenomic Analyses Provide Novel Interpretations of Type 1 Diabetes Risk Loci Through Circulating Proteins. Diabetes 2025; 74:630-639. [PMID: 39761376 DOI: 10.2337/db24-0380] [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: 05/06/2024] [Accepted: 01/01/2025] [Indexed: 06/01/2025]
Abstract
ARTICLE HIGHLIGHTS Identification of circulating proteins that may play a role in the pathogenesis of type 1 diabetes can provide promising targets for biomarker and drug target identification. Supported by multiple lines of evidence, circulating abundances of CTSH, IL27RA, SIRPG, and PGM1 were associated with the risk of type 1 diabetes. Tissues and cell types with enrichment of target protein-coding gene expression were identified. CTSH, IL27RA, SIRPG, and PGM1 may be explored as biomarkers or drug targets for type 1 diabetes.
Collapse
Affiliation(s)
- Tianyuan Lu
- Department of Statistical Sciences, Faculty of Arts and Science, University of Toronto, Toronto, Ontario, Canada
- Department of Population Health Sciences, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
- Center for Demography of Health and Aging, University of Wisconsin-Madison, Madison, WI
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, WI
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI
| | - Despoina Manousaki
- Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Research Center of the Sainte-Justine University Hospital, Université de Montréal, Montreal, Quebec, Canada
| | - Lei Sun
- Department of Statistical Sciences, Faculty of Arts and Science, University of Toronto, Toronto, Ontario, Canada
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Andrew D Paterson
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Division of Epidemiology, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
29
|
Song Z, Li H, Zhang J, Huang Y, Gao S. PLATELET TRAITS AND SEPSIS RISK AND PROGNOSIS: A BIDIRECTIONAL TWO-SAMPLE MENDELIAN RANDOMIZATION STUDY. Shock 2025; 63:520-526. [PMID: 39158958 DOI: 10.1097/shk.0000000000002447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
ABSTRACT Background: Sepsis is a critical medical condition characterized by a dysregulated host response to infection. Platelet abnormalities frequently manifest in sepsis patients, but the causal role of platelets in sepsis remains unclear. This study employed a bidirectional two-sample Mendelian randomization (MR) approach to investigate the causal direction between platelets and sepsis. Methods: MR analysis was used to investigate the causal effect of four platelet traits-platelet count (PLT), platelet crit (PCT), mean platelet volume (MPV), and platelet distribution width (PDW)-on sepsis risk and prognosis. Additionally, the study explored the reverse causality, assessing the impact of sepsis on these platelet traits. Genetic variants from large-scale genome-wide association studies served as instrumental variables to infer causality. Sensitivity analyses and heterogeneity tests were conducted to ensure the validity and robustness of the results. Results: Genetically predicted decreased PCT (OR = 0.938, P = 0.044) and MPV (OR = 0.410, P = 0.006) were associated with an increased risk of sepsis. In the reverse direction, 28-day sepsis mortality was significantly associated with decreased PLT (OR = 0.986, P = 0.034). No significant causal relationships were observed between sepsis and other platelet traits. Conclusions: This study suggests a causal association between low PCT and MPV levels and increased risk of sepsis. Additionally, sepsis with a poor prognosis was causally linked to decreased PLT. These findings provide novel evidence for the causal relationship between platelet traits and sepsis.
Collapse
Affiliation(s)
- Zhonghai Song
- Department of Pre-hospital Emergency, Xingtai People's Hospital, Hebei Medical University, Xingtai, Hebei, China
| | - Hua Li
- Department of Gastrointestinal Oncology Surgery, Xingtai People's Hospital, Hebei Medical University, Xingtai, Hebei, China
| | - Jing Zhang
- Department of Neurology, Xingtai People's Hospital, Hebei Medical University, Xingtai, Hebei, China
| | | | | |
Collapse
|
30
|
Henry A, Mo X, Finan C, Chaffin MD, Speed D, Issa H, Denaxas S, Ware JS, Zheng SL, Malarstig A, Gratton J, Bond I, Roselli C, Miller D, Chopade S, Schmidt AF, Abner E, Adams L, Andersson C, Aragam KG, Ärnlöv J, Asselin G, Raja AA, Backman JD, Bartz TM, Biddinger KJ, Biggs ML, Bloom HL, Boersma E, Brandimarto J, Brown MR, Brunak S, Bruun MT, Buckbinder L, Bundgaard H, Carey DJ, Chasman DI, Chen X, Cook JP, Czuba T, de Denus S, Dehghan A, Delgado GE, Doney AS, Dörr M, Dowsett J, Dudley SC, Engström G, Erikstrup C, Esko T, Farber-Eger EH, Felix SB, Finer S, Ford I, Ghanbari M, Ghasemi S, Ghouse J, Giedraitis V, Giulianini F, Gottdiener JS, Gross S, Guðbjartsson DF, Gui H, Gutmann R, Hägg S, Haggerty CM, Hedman ÅK, Helgadottir A, Hemingway H, Hillege H, Hyde CL, Aagaard Jensen B, Jukema JW, Kardys I, Karra R, Kavousi M, Kizer JR, Kleber ME, Køber L, Koekemoer A, Kuchenbaecker K, Lai YP, Lanfear D, Langenberg C, Lin H, Lind L, Lindgren CM, Liu PP, London B, Lowery BD, Luan J, Lubitz SA, Magnusson P, Margulies KB, Marston NA, Martin H, März W, Melander O, Mordi IR, Morley MP, et alHenry A, Mo X, Finan C, Chaffin MD, Speed D, Issa H, Denaxas S, Ware JS, Zheng SL, Malarstig A, Gratton J, Bond I, Roselli C, Miller D, Chopade S, Schmidt AF, Abner E, Adams L, Andersson C, Aragam KG, Ärnlöv J, Asselin G, Raja AA, Backman JD, Bartz TM, Biddinger KJ, Biggs ML, Bloom HL, Boersma E, Brandimarto J, Brown MR, Brunak S, Bruun MT, Buckbinder L, Bundgaard H, Carey DJ, Chasman DI, Chen X, Cook JP, Czuba T, de Denus S, Dehghan A, Delgado GE, Doney AS, Dörr M, Dowsett J, Dudley SC, Engström G, Erikstrup C, Esko T, Farber-Eger EH, Felix SB, Finer S, Ford I, Ghanbari M, Ghasemi S, Ghouse J, Giedraitis V, Giulianini F, Gottdiener JS, Gross S, Guðbjartsson DF, Gui H, Gutmann R, Hägg S, Haggerty CM, Hedman ÅK, Helgadottir A, Hemingway H, Hillege H, Hyde CL, Aagaard Jensen B, Jukema JW, Kardys I, Karra R, Kavousi M, Kizer JR, Kleber ME, Køber L, Koekemoer A, Kuchenbaecker K, Lai YP, Lanfear D, Langenberg C, Lin H, Lind L, Lindgren CM, Liu PP, London B, Lowery BD, Luan J, Lubitz SA, Magnusson P, Margulies KB, Marston NA, Martin H, März W, Melander O, Mordi IR, Morley MP, Morris AP, Morrison AC, Morton L, Nagle MW, Nelson CP, Niessner A, Niiranen T, Noordam R, Nowak C, O'Donoghue ML, Ostrowski SR, Owens AT, Palmer CNA, Paré G, Pedersen OB, Perola M, Pigeyre M, Psaty BM, Rice KM, Ridker PM, Romaine SPR, Rotter JI, Ruff CT, Sabatine MS, Sallah N, Salomaa V, Sattar N, Shalaby AA, Shekhar A, Smelser DT, Smith NL, Sørensen E, Srinivasan S, Stefansson K, Sveinbjörnsson G, Svensson P, Tammesoo ML, Tardif JC, Teder-Laving M, Teumer A, Thorgeirsson G, Thorsteinsdottir U, Torp-Pedersen C, Tragante V, Trompet S, Uitterlinden AG, Ullum H, van der Harst P, van Heel D, van Setten J, van Vugt M, Veluchamy A, Verschuuren M, Verweij N, Vissing CR, Völker U, Voors AA, Wallentin L, Wang Y, Weeke PE, Wiggins KL, Williams LK, Yang Y, Yu B, Zannad F, Zheng C, Asselbergs FW, Cappola TP, Dubé MP, Dunn ME, Lang CC, Samani NJ, Shah S, Vasan RS, Smith JG, Holm H, Shah S, Ellinor PT, Hingorani AD, Wells Q, Lumbers RT. Genome-wide association study meta-analysis provides insights into the etiology of heart failure and its subtypes. Nat Genet 2025; 57:815-828. [PMID: 40038546 PMCID: PMC11985341 DOI: 10.1038/s41588-024-02064-3] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/17/2024] [Indexed: 03/06/2025]
Abstract
Heart failure (HF) is a major contributor to global morbidity and mortality. While distinct clinical subtypes, defined by etiology and left ventricular ejection fraction, are well recognized, their genetic determinants remain inadequately understood. In this study, we report a genome-wide association study of HF and its subtypes in a sample of 1.9 million individuals. A total of 153,174 individuals had HF, of whom 44,012 had a nonischemic etiology (ni-HF). A subset of patients with ni-HF were stratified based on left ventricular systolic function, where data were available, identifying 5,406 individuals with reduced ejection fraction and 3,841 with preserved ejection fraction. We identify 66 genetic loci associated with HF and its subtypes, 37 of which have not previously been reported. Using functionally informed gene prioritization methods, we predict effector genes for each identified locus, and map these to etiologic disease clusters through phenome-wide association analysis, network analysis and colocalization. Through heritability enrichment analysis, we highlight the role of extracardiac tissues in disease etiology. We then examine the differential associations of upstream risk factors with HF subtypes using Mendelian randomization. These findings extend our understanding of the mechanisms underlying HF etiology and may inform future approaches to prevention and treatment.
Collapse
Affiliation(s)
- Albert Henry
- Institute of Cardiovascular Science, University College London, London, UK
- Institute of Health Informatics, University College London, London, UK
| | - Xiaodong Mo
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Chris Finan
- Institute of Cardiovascular Science, University College London, London, UK
| | - Mark D Chaffin
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Doug Speed
- Center for Quantitative Genetics and Genomics, Aarhus University, Aarhus, Denmark
| | - Hanane Issa
- Institute of Health Informatics, University College London, London, UK
| | - Spiros Denaxas
- Institute of Health Informatics, University College London, London, UK
- Health Data Research UK, London, UK
- British Heart Foundation Data Science Centre, London, UK
- The National Institute for Health Research University College London Hospitals Biomedical Research Centre, University College London, London, UK
| | - James S Ware
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- National Heart & Lung Institute, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
- Hammersmith Hospital, Imperial College Hospitals NHS Trust, London, UK
| | - Sean L Zheng
- National Heart & Lung Institute, Imperial College London, London, UK
- Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Anders Malarstig
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
- Pfizer Worldwide Research & Development, Cambridge, MA, USA
| | - Jasmine Gratton
- Institute of Cardiovascular Science, University College London, London, UK
| | - Isabelle Bond
- Institute of Cardiovascular Science, University College London, London, UK
| | - Carolina Roselli
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - David Miller
- Division of Biosciences, University College London, London, UK
| | - Sandesh Chopade
- Institute of Cardiovascular Science, University College London, London, UK
| | - A Floriaan Schmidt
- Institute of Cardiovascular Science, University College London, London, UK
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Erik Abner
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | | | - Charlotte Andersson
- Department of Cardiology, Herlev Gentofte Hospital, Herlev, Denmark
- National Heart, Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA
| | - Krishna G Aragam
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Johan Ärnlöv
- Department of Neurobiology, Care Sciences and Society/Section of Family Medicine and Primary Care, Karolinska Institutet, Stockholm, Sweden
- School of Health and Social Sciences, Dalarna University, Falun, Sweden
| | | | - Anna Axelsson Raja
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Joshua D Backman
- Analytical Genetics, Regeneron Genetics Center, Tarrytown, NY, USA
| | - Traci M Bartz
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Kiran J Biddinger
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Mary L Biggs
- Department of Biostatistics, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Heather L Bloom
- Department of Medicine, Division of Cardiology, Emory University Medical Center, Atlanta, GA, USA
| | - Eric Boersma
- Department of Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jeffrey Brandimarto
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael R Brown
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas School of Public Health, Houston, TX, USA
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mie Topholm Bruun
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
| | | | - Henning Bundgaard
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - David J Carey
- Department of Molecular and Functional Genomics, Geisinger, Danville, PA, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Xing Chen
- Pfizer Worldwide Research & Development, Cambridge, MA, USA
| | - James P Cook
- Department of Biostatistics, University of Liverpool, Liverpool, UK
| | - Tomasz Czuba
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Simon de Denus
- Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - Abbas Dehghan
- MRC-PHE Centre for Environment and Health, Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Graciela E Delgado
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty of Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Alexander S Doney
- Division of Molecular & Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Marcus Dörr
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Joseph Dowsett
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Samuel C Dudley
- Department of Medicine, Cardiovascular Division, University of Minnesota, Minneapolis, MN, USA
| | - Gunnar Engström
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
- Deparment of Clinical Medicine, Health, Aarhus University, Aarhus, Denmark
| | - Tõnu Esko
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Eric H Farber-Eger
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Stephan B Felix
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Sarah Finer
- Centre for Primary Care and Public Health, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Ian Ford
- Robertson Center for Biostatistics, Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Sahar Ghasemi
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Jonas Ghouse
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Franco Giulianini
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - John S Gottdiener
- Department of Medicine, Division of Cardiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Stefan Gross
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Daníel F Guðbjartsson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | - Hongsheng Gui
- Center for Individualized and Genomic Medicine Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - Rebecca Gutmann
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Sara Hägg
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | | | - Åsa K Hedman
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | | | - Harry Hemingway
- Institute of Health Informatics, University College London, London, UK
- Health Data Research UK, London, UK
| | - Hans Hillege
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Craig L Hyde
- Pfizer Worldwide Research & Development, Cambridge, MA, USA
| | | | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, LUMC, Leiden, the Netherlands
| | - Isabella Kardys
- Department of Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ravi Karra
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC, USA
- Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jorge R Kizer
- Cardiology Section, San Francisco Veterans Affairs Health System, and Departments of Medicine, Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Marcus E Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty of Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Lars Køber
- Department of Cardiology, Nordsjaellands Hospital, Copenhagen, Denmark
| | - Andrea Koekemoer
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Karoline Kuchenbaecker
- Division of Psychiatry, University College London, London, UK
- UCL Genetics Institute, University College London, London, UK
| | - Yi-Pin Lai
- Pfizer Worldwide Research & Development, Cambridge, MA, USA
| | - David Lanfear
- Center for Individualized and Genomic Medicine Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
- Heart and Vascular Institute, Henry Ford Hospital, Detroit, MI, USA
| | - Claudia Langenberg
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
- Computational Medicine, Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Honghuang Lin
- National Heart, Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Cecilia M Lindgren
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Peter P Liu
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
- Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Barry London
- Division of Cardiovascular Medicine and Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA, USA
| | - Brandon D Lowery
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jian'an Luan
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Steven A Lubitz
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiac Arrhythmia Service and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Patrik Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Kenneth B Margulies
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicholas A Marston
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Hilary Martin
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty of Mannheim, University of Heidelberg, Heidelberg, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
- Synlab Academy, Synlab Holding Deutschland GmbH, Mannheim, Germany
| | - Olle Melander
- Department of Internal Medicine, Clinical Sciences, Lund University and Skåne University Hospital, Malmö, Sweden
| | - Ify R Mordi
- Division of Molecular & Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Michael P Morley
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew P Morris
- Department of Biostatistics, University of Liverpool, Liverpool, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Alanna C Morrison
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas School of Public Health, Houston, TX, USA
| | - Lori Morton
- Cardiovascular Research, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | | | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Alexander Niessner
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Teemu Niiranen
- Department of Medicine, Turku University Hospital and University of Turku, Turku, Finland
- National Institute for Health and Welfare, Helsinki, Finland
| | - Raymond Noordam
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Christoph Nowak
- Department of Neurobiology, Care Sciences and Society/Section of Family Medicine and Primary Care, Karolinska Institutet, Stockholm, Sweden
| | - Michelle L O'Donoghue
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anjali T Owens
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Colin N A Palmer
- Division of Population Health and Genomics, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Guillaume Paré
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
- Thrombosis and Atherosclerosis Research Institute, Hamilton, Ontario, Canada
- Population Health Research Institute, Hamilton, Ontario, Canada
| | - Ole Birger Pedersen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Immunology, Zealand University Hospital, Køge, Denmark
| | - Markus Perola
- National Institute for Health and Welfare, Helsinki, Finland
| | - Marie Pigeyre
- Population Health Research Institute, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA, USA
| | - Kenneth M Rice
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Simon P R Romaine
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Harbor-UCLA Medical Center, Torrance, CA, USA
- Departments of Pediatrics and Medicine, Harbor-UCLA Medical Center, Torrance, CA, USA
- Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Christian T Ruff
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Marc S Sabatine
- TIMI Study Group, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Neneh Sallah
- Institute of Health Informatics, University College London, London, UK
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki, Finland
| | - Naveed Sattar
- BHF Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Alaa A Shalaby
- Department of Medicine, Division of Cardiology, University of Pittsburgh Medical Center and VA Pittsburgh HCS, Pittsburgh, PA, USA
| | - Akshay Shekhar
- Cardiovascular Research, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Diane T Smelser
- Department of Molecular and Functional Genomics, Geisinger, Danville, PA, USA
| | - Nicholas L Smith
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Veterans Affairs Office of Research & Development, Seattle Epidemiologic Research and Information Center, Seattle, WA, USA
| | - Erik Sørensen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Sundararajan Srinivasan
- Division of Population Health and Genomics, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Kari Stefansson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Department of Medicine, University of Iceland, Reykjavik, Iceland
| | | | - Per Svensson
- Department of Cardiology, Söderjukhuset, Stockholm, Sweden
- Department of Clinical Science and Education-Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Mari-Liis Tammesoo
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Jean-Claude Tardif
- Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Maris Teder-Laving
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Alexander Teumer
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Guðmundur Thorgeirsson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Department of Medicine, University of Iceland, Reykjavik, Iceland
- Department of Internal Medicine, Division of Cardiology, National University Hospital of Iceland, Reykjavik, Iceland
| | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Department of Medicine, University of Iceland, Reykjavik, Iceland
| | | | | | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Andre G Uitterlinden
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Pim van der Harst
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
| | - David van Heel
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK
| | - Jessica van Setten
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marion van Vugt
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Abirami Veluchamy
- Division of Molecular & Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
- Division of Population Health and Genomics, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Monique Verschuuren
- Department Life Course and Health, Centre for Nutrition, Prevention and Health Services, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Niek Verweij
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Christoffer Rasmus Vissing
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Uwe Völker
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Adriaan A Voors
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Lars Wallentin
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Yunzhang Wang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Peter E Weeke
- Department of Cardiology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kerri L Wiggins
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - L Keoki Williams
- Center for Individualized and Genomic Medicine Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - Yifan Yang
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas School of Public Health, Houston, TX, USA
| | - Faiez Zannad
- Université de Lorraine, CHU de Nancy, Inserm and INI-CRCT (F-CRIN), Institut Lorrain du Coeur et des Vaisseaux, Vandoeuvre Lès Nancy, France
| | - Chaoqun Zheng
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Folkert W Asselbergs
- Institute of Health Informatics, University College London, London, UK
- The National Institute for Health Research University College London Hospitals Biomedical Research Centre, University College London, London, UK
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Thomas P Cappola
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marie-Pierre Dubé
- Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Michael E Dunn
- Cardiovascular Research, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Chim C Lang
- Division of Molecular & Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Svati Shah
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
- Duke Molecular Physiology Institute, Durham, NC, USA
| | - Ramachandran S Vasan
- National Heart, Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA
- Sections of Cardiology, Preventive Medicine and Epidemiology, Department of Medicine, Boston University Schools of Medicine and Public Health, Boston, MA, USA
| | - J Gustav Smith
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University and Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Cardiology, Clinical Sciences, Lund University and Skåne University Hospital, Lund, Sweden
- Wallenberg Center for Molecular Medicine and Lund University Diabetes Center, Lund University, Lund, Sweden
| | - Hilma Holm
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
| | - Sonia Shah
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Patrick T Ellinor
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiac Arrhythmia Service and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiac Arrhythmia Service and Cardiovascular Research Center, Massachusetts General Hospital, Cambridge, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aroon D Hingorani
- Institute of Cardiovascular Science, University College London, London, UK
| | - Quinn Wells
- Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN, USA
| | - R Thomas Lumbers
- Institute of Health Informatics, University College London, London, UK.
- Health Data Research UK, London, UK.
- The National Institute for Health Research University College London Hospitals Biomedical Research Centre, University College London, London, UK.
| |
Collapse
|
31
|
Wenz BM, He Y, Chen NC, Pickrell JK, Li JH, Dudek MF, Li T, Keener R, Voight BF, Brown CD, Battle A. Genotype inference from aggregated chromatin accessibility data reveals genetic regulatory mechanisms. Genome Biol 2025; 26:81. [PMID: 40159496 PMCID: PMC11956263 DOI: 10.1186/s13059-025-03538-1] [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: 07/11/2024] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Understanding the genetic causes underlying variability in chromatin accessibility can shed light on the molecular mechanisms through which genetic variants may affect complex traits. Thousands of ATAC-seq samples have been collected that hold information about chromatin accessibility across diverse cell types and contexts, but most of these are not paired with genetic information and come from distinct projects and laboratories. RESULTS We report here joint genotyping, chromatin accessibility peak calling, and discovery of quantitative trait loci which influence chromatin accessibility (caQTLs), demonstrating the capability of performing caQTL analysis on a large scale in a diverse sample set without pre-existing genotype information. Using 10,293 profiling samples representing 1454 unique donor individuals across 653 studies from public databases, we catalog 24,159 caQTLs in total. After joint discovery analysis, we cluster samples based on accessible chromatin profiles to identify context-specific caQTLs. We find that caQTLs are strongly enriched for annotations of gene regulatory elements across diverse cell types and tissues and are often linked with genetic variation associated with changes in expression (eQTLs), indicating that caQTLs can mediate genetic effects on gene expression. We demonstrate sharing of causal variants for chromatin accessibility across human traits, enabling a more complete picture of the genetic mechanisms underlying complex human phenotypes. CONCLUSIONS Our work provides a proof of principle for caQTL calling from previously ungenotyped samples and represents one of the largest, most diverse caQTL resources currently available, informing mechanisms of genetic regulation of gene expression and contribution to disease.
Collapse
Affiliation(s)
- Brandon M Wenz
- Genetics and Epigenetics Program, Cell and Molecular Biology Graduate Group, Biomedical Graduate Studies, University of Pennsylvania-Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Yuan He
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Nae-Chyun Chen
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, 21218, USA
| | | | | | - Max F Dudek
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Taibo Li
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Rebecca Keener
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Benjamin F Voight
- Department of Genetics, University of Pennsylvania-Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania-Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania-Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Christopher D Brown
- Department of Genetics, University of Pennsylvania-Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Alexis Battle
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA.
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, 21218, USA.
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD, 21218, USA.
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, 21218, USA.
- Data Science and AI Institute, Johns Hopkins University, Baltimore, MD, 21218, USA.
| |
Collapse
|
32
|
Hazelett DJ. Rethinking GWAS: how lessons from genetic screens and artificial intelligence could reveal biological mechanisms. Bioinformatics 2025; 41:btaf153. [PMID: 40198231 PMCID: PMC12014097 DOI: 10.1093/bioinformatics/btaf153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 04/10/2025] Open
Abstract
MOTIVATION Modern single-cell omics data are key to unraveling the complex mechanisms underlying risk for complex diseases revealed by genome-wide association studies (GWAS). Phenotypic screens in model organisms have several important parallels to GWAS which the author explores in this essay. RESULTS The author provides the historical context of such screens, comparing and contrasting similarities to association studies, and how these screens in model organisms can teach us what to look for. Then the author considers how the results of GWAS might be exhaustively interrogated to interpret the biological mechanisms underpinning disease processes. Finally, the author proposes a general framework for tackling this problem computationally, and explore the data, mechanisms, and technology (both existing and yet to be invented) that are necessary to complete the task. AVAILABILITY AND IMPLEMENTATION There are no data or code associated with this article.
Collapse
Affiliation(s)
- Dennis J Hazelett
- Department of Computational Biomedicine at Cedars-Sinai Medical Center, West Hollywood, CA 90069, United States
- Cancer Prevention and Control—Samuel Oschin Cancer Center, Los Angeles, CA 90048, United States
| |
Collapse
|
33
|
Neylan CJ, Levin MG, Hartmann K, Beigel K, Khodursky S, DePaolo JS, Abramowitz S, Furth EE, Heuckeroth RO, Damrauer SM, Maguire LH. Genome-wide association meta-analysis identifies 126 novel loci for diverticular disease and implicates connective tissue and colonic motility. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.27.25324777. [PMID: 40196262 PMCID: PMC11974943 DOI: 10.1101/2025.03.27.25324777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Diverticular disease is a common and morbid complex phenotype influenced by both innate and environmental risk factors. We performed the largest genome-wide association study meta-analysis for diverticular disease, identifying 126 novel loci. Employing multiple downstream analytic strategies, including tissue and pathway enrichment, statistical fine-mapping, allele-specific expression, protein quantitative trait loci and drug-target investigations, and linkage disequilibrium score regression, we prioritized causal genes and produced several lines of evidence linking diverticular disease to connective tissue biology and colonic motility. We substantiated these findings by integrating single-cell RNA sequencing data, showing that prioritized diverticular disease-associated genes are enriched for expression in colonic smooth muscle, fibroblasts, and interstitial cells of Cajal. In quantitative analysis of surgical specimens, we found a substantial reduction in the density of elastin present in the sigmoid colon in severe diverticulitis.
Collapse
Affiliation(s)
- Christopher J. Neylan
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Michael G. Levin
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA 19104
| | - Katherine Hartmann
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Katherine Beigel
- Department of Biomedical and Health Informatics (DBHi), Children’s Hospital of Philadelphia, Philadelphia, PA 19104
| | - Sam Khodursky
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - John S. DePaolo
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Sarah Abramowitz
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
- The Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549
| | - Emma E. Furth
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104
| | - Robert O. Heuckeroth
- Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104
- The Children’s Hospital of Philadelphia Research Institute and Abramson Research Center, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
- Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Boulevard, Philadelphia, PA 19104
| | - Scott M. Damrauer
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA 19104
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Lillias H. Maguire
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA 19104
- Division of Colon and Rectal Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
34
|
Ye Z, Hong D, Yuan J, Xu P, Liu W. Assessing the influence of plasma metabolites on chronic skin ulcer risk: a two-sample Mendelian randomization study. Sci Rep 2025; 15:10001. [PMID: 40121277 PMCID: PMC11929753 DOI: 10.1038/s41598-025-94311-8] [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: 11/05/2024] [Accepted: 03/12/2025] [Indexed: 03/25/2025] Open
Abstract
Chronic skin ulcers, although rare, pose severe and debilitating challenges. The identification of causal metabolite biomarkers presents an opportunity to refine effective risk assessment strategies for this condition. In this study, we conducted a comprehensive Two-Sample Mendelian Randomization (TSMR) investigation to delineate the potential causal effects of plasma metabolites on chronic skin ulcer risk. Exposure data comprised 14,296 participants with 913 metabolites from INTERVAL/EPIC-Norfolk, and 8,299 participants with 1,091 metabolites and 309 ratios from the Canadian Longitudinal Study on Aging (CLSA). Outcome data came from the finngen_R9_L12_CHRONICULCEROFSKIN (1,840 cases, 353,088 controls) and UK Biobank Chronic ulcer of skin (495 cases, 455,853 controls) cohorts. Leveraging the inverse-variance weighted (IVW) method, alongside MR-Egger and MR-PRESSO sensitivity analyses, we evaluated metabolite associations with chronic skin ulcer risk. Further assessment involved a phenome-wide MR (Phe-MR) analysis to explore potential repercussions of targeting identified metabolites for intervention. Our study identified 12 distinct metabolites significantly associated with chronic skin ulcers, demonstrating consistent and replicable results. Notably, X-19,141 exhibited the highest reproducibility. These findings highlight novel plasma metabolites relevant to chronic skin ulcers, offering theoretical underpinnings for mechanistic research and clinical strategies in prevention and treatment.
Collapse
Affiliation(s)
- Zheng Ye
- School of Mathematics and Information Science, Guangzhou University, Guangzhou, 510006, China
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, 558000, Guizhou, China
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Deqing Hong
- School of Mathematics and Information Science, Guangzhou University, Guangzhou, 510006, China
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Jiaqi Yuan
- School of Mathematics and Information Science, Guangzhou University, Guangzhou, 510006, China
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Peng Xu
- School of Mathematics and Information Science, Guangzhou University, Guangzhou, 510006, China.
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, 558000, Guizhou, China.
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China.
| | - Wenbin Liu
- School of Mathematics and Information Science, Guangzhou University, Guangzhou, 510006, China.
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China.
| |
Collapse
|
35
|
Lim J, Vujkovic M, Levin MG, Lorenz K, Voight BF, Zhang DY, Dudek MF, Pahl MC, Pippin JA, Su C, Manduchi E, Wells AD, Grant SF, Abramowitz S, Damrauer SM, Mukherjee S, Yang G, Kaplan DE, Penn Medicine BioBank, Rader DJ. Trans-ancestry genome-wide association meta-analysis of gallstone disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.16.25324077. [PMID: 40166541 PMCID: PMC11957090 DOI: 10.1101/2025.03.16.25324077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Gallstone disease is a highly prevalent and costly gastrointestinal disease. Yet, genetic variation in susceptibility to gallstone disease and its implication in metabolic regulatory pathways remain to be explored. We report a trans-ancestry genome-wide association meta-analysis of gallstone disease including 88,063 cases and 1,490,087 controls in the UK Biobank, FinnGen, Biobank Japan, and Million Veteran Program. We identified 91 (37 novel) risk loci across the meta-analysis and found replication in statistically compelling signals in the All of Us Research Program. A polygenic risk score constructed from trans-ancestry lead variants was positively associated with liver chemistry and alpha-1-antitrypsin deficiency and negatively associated with total cholesterol and low-density lipoprotein levels among trans-ancestry and European ancestry groups in the Penn Medicine BioBank. Cross-trait colocalization analysis between risk loci and 44 liver, metabolic, renal, and inflammatory traits yielded 350 significant colocalizations as well as 97 significant colocalizations and 65 prioritized genes from expression quantitative trait loci from eight tissues. These findings broaden our understanding of the genetic architecture of gallstone disease.
Collapse
Affiliation(s)
- Junghyun Lim
- Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Division of Translational Medicine and Human Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Marijana Vujkovic
- Division of Translational Medicine and Human Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Michael G. Levin
- Division of Cardiovascular Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kim Lorenz
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Benjamin F. Voight
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - David Y. Zhang
- Division of Translational Medicine and Human Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Max F. Dudek
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Graduate Group in Genomics and Computational Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew C. Pahl
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - James A. Pippin
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Chun Su
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Elisabetta Manduchi
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Andrew D. Wells
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Struan F.A. Grant
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Sarah Abramowitz
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Scott M. Damrauer
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Samiran Mukherjee
- Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Guoyi Yang
- Division of Translational Medicine and Human Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - David E. Kaplan
- Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | | | - Daniel J. Rader
- Division of Translational Medicine and Human Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
36
|
Wang J, Wang C, Li S, Huang M, Zhang R, Chen Y, Kang Z, Li W. Causal effect between circulating metabolic markers and glioma: a bidirectional, two-sample, Bayesian weighted Mendelian randomization. Discov Oncol 2025; 16:315. [PMID: 40085281 PMCID: PMC11909308 DOI: 10.1007/s12672-025-02050-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 03/04/2025] [Indexed: 03/16/2025] Open
Abstract
Glioma is the most common malignant tumor in the central nervous system with significant challenges for its treatment and prognosis. Based on publicly available genome-wide association study data, this study employed a bidirectional, two-sample Mendelian randomization (MR) analysis, combined with Bayesian weighted MR, to investigate the causal effect of 233 circulating metabolic traits on glioma and its subtypes, with the expectation of discovering new diagnostic and therapeutic targets. The MR study revealed that the Total cholesterol to total lipids ratio in large VLDL and the Ratio of polyunsaturated fatty acids to total fatty acids (PUFAbyFA) are risk factors for glioma, while free cholesterol or phospholipids in small HDL are protective against glioma. The free cholesterol to total lipids ratio in IDL, the ratios of total cholesterol or cholesteryl esters to total lipids in small or medium VLDL, as well as linoleic acid (LA 18:2), phosphatidylcholine, and sphingomyelins levels are risk factors for non-GBM glioma. Free cholesterol in small HDL is identified as a protective factor for non-GBM glioma. The results were robust to sensitivity analyses and Bayesian weighted mendelian randomization. The causal effects of glioma on circulating metabolites were explored through reverse mendelian randomization. The results provide potential biomarkers for the early diagnosis and treatment of glioma.
Collapse
Affiliation(s)
- Jiachen Wang
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, No.119, West Nan Si Huan Road, Fengtai District, Beijing, 100071, China
| | - Chengzhuo Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100071, China
| | - Shenglan Li
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, No.119, West Nan Si Huan Road, Fengtai District, Beijing, 100071, China
| | - Mengqian Huang
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, No.119, West Nan Si Huan Road, Fengtai District, Beijing, 100071, China
| | - Rong Zhang
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, No.119, West Nan Si Huan Road, Fengtai District, Beijing, 100071, China
| | - Yuxiao Chen
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, No.119, West Nan Si Huan Road, Fengtai District, Beijing, 100071, China
- The First Clinical College of Capital Medical University, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Zhuang Kang
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, No.119, West Nan Si Huan Road, Fengtai District, Beijing, 100071, China
| | - Wenbin Li
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, No.119, West Nan Si Huan Road, Fengtai District, Beijing, 100071, China.
| |
Collapse
|
37
|
Pujol Gualdo N, Džigurski J, Rukins V, Pajuste FD, Wolford BN, Võsa M, Golob M, Haug L, Alver M, Läll K, Peters M, Brumpton BM, Palta P, Mägi R, Laisk T. Atlas of genetic and phenotypic associations across 42 female reproductive health diagnoses. Nat Med 2025:10.1038/s41591-025-03543-8. [PMID: 40069456 DOI: 10.1038/s41591-025-03543-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 01/28/2025] [Indexed: 04/02/2025]
Abstract
The genetic background of many female reproductive health diagnoses remains uncharacterized, compromising our understanding of the underlying biology. Here, we map the genetic architecture across 42 female-specific health conditions using data from up to 293,618 women from two large population-based cohorts, the Estonian Biobank and the FinnGen study. Our study illustrates the utility of genetic analyses in understanding women's health better. As specific examples, we describe genetic risk factors for ovarian cysts that elucidate the genetic determinants of folliculogenesis and, by leveraging population-specific variants, uncover new candidate genes for uterine fibroids. We find that most female reproductive health diagnoses have a heritable component, with varying degrees of polygenicity and discoverability. Finally, we identify pleiotropic loci and genes that function in genital tract development (WNT4, PAX8, WT1, SALL1), hormonal regulation (FSHB, GREB1, BMPR1B, SYNE1/ESR1) and folliculogenesis (CHEK2), underlining their integral roles in female reproductive health.
Collapse
Affiliation(s)
- Natàlia Pujol Gualdo
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Jelisaveta Džigurski
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Valentina Rukins
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Fanny-Dhelia Pajuste
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Brooke N Wolford
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - Mariann Võsa
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Mia Golob
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Lisette Haug
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Maris Alver
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kristi Läll
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Maire Peters
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Celvia CC AS, Tartu, Estonia
| | - Ben M Brumpton
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - Priit Palta
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Reedik Mägi
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Triin Laisk
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia.
| |
Collapse
|
38
|
Kim J, Williams A, Noh H, Jasper EA, Jones SH, Jaworski JA, Shuey MM, Ruiz-Narváez EA, Wise LA, Palmer JR, Connolly J, Keaton JM, Denny JC, Khan A, Abbass MA, Rasmussen-Torvik LJ, Kottyan LC, Madhivanan P, Krupp K, Wei WQ, Edwards TL, Velez Edwards DR, Hellwege JN. Genome-wide meta-analysis identifies novel risk loci for uterine fibroids within and across multiple ancestry groups. Nat Commun 2025; 16:2273. [PMID: 40050615 PMCID: PMC11885530 DOI: 10.1038/s41467-025-57483-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 02/12/2025] [Indexed: 03/09/2025] Open
Abstract
Uterine leiomyomata or fibroids are highly heritable, common, and benign tumors of the uterus with poorly understood etiology. Previous GWAS have reported 72 associated genes but included limited numbers of non-European individuals. Here, we identify 11 novel genes associated with fibroids across multi-ancestry and ancestry-stratified GWAS analyses. We replicate a known fibroid GWAS gene in African ancestry individuals and estimate the SNP-based heritability of fibroids in African ancestry populations as 15.9%. Using genetically predicted gene expression and colocalization analyses, we identify 46 novel genes associated with fibroids. These genes are significantly enriched in cancer, cell death and survival, reproductive system disease, and cellular growth and proliferation networks. We also find that increased predicted expression of HEATR3 in uterine tissue is associated with fibroids across ancestry strata. Overall, we report genetic variants associated with fibroids coupled with functional and gene pathway enrichment analyses.
Collapse
Affiliation(s)
- Jeewoo Kim
- Division of Quantitative and Clinical Sciences, Department of Obstetrics & Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
| | - Ariel Williams
- Center for Precision Health Research, National Human Genome Research Institute, National Institute of Health, Bethesda, MD, USA
| | - Hannah Noh
- Tufts University Medical School Graduate Programs, Boston, MA, USA
- Medicine, Health and Society, Vanderbilt University, Nashville, TN, USA
| | - Elizabeth A Jasper
- Division of Quantitative and Clinical Sciences, Department of Obstetrics & Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sarah H Jones
- Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James A Jaworski
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
- Division of Epidemiology, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Megan M Shuey
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Edward A Ruiz-Narváez
- Department of Nutritional Sciences University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Lauren A Wise
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Julie R Palmer
- Slone Epidemiology Center at Boston University, Boston, MA, USA
| | - John Connolly
- Center for Applied Genomics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jacob M Keaton
- Center for Precision Health Research, National Human Genome Research Institute, National Institute of Health, Bethesda, MD, USA
- Division of Epidemiology, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joshua C Denny
- Center for Precision Health Research, National Human Genome Research Institute, National Institute of Health, Bethesda, MD, USA
- All of Us Research Program, Office of the Director, National Institutes of Health, Bethesda, MD, USA
| | - Atlas Khan
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Mohammad A Abbass
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Leah C Kottyan
- Center for Autoimmune Genomics and Etiology, Department of Pediatrics, Cincinnati Children's Hospital, University of Cincinnati, Cincinnati, OH, USA
| | - Purnima Madhivanan
- University of Arizona Comprehensive Cancer Center, Tucson, AZ, USA
- Department of Health Promotion Sciences, Mel & Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
- Public Health Research Institute of India, Mysuru, India
- Department of Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Karl Krupp
- University of Arizona Comprehensive Cancer Center, Tucson, AZ, USA
- Public Health Research Institute of India, Mysuru, India
- Public Health Practice, Policy, & Translational Research Department, Mel & Enid Zuckerman College of Public Health, Phoenix, AZ, USA
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Digna R Velez Edwards
- Division of Quantitative and Clinical Sciences, Department of Obstetrics & Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA.
- Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Jacklyn N Hellwege
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
39
|
Dumesic PA, Wilensky SE, Bose S, Van Vranken JG, Gygi SP, Spiegelman BM. RBM43 controls PGC1α translation and a PGC1α-STING signaling axis. Cell Metab 2025; 37:742-757.e8. [PMID: 39965564 PMCID: PMC11885043 DOI: 10.1016/j.cmet.2025.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 10/17/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025]
Abstract
Obesity is associated with systemic inflammation that impairs mitochondrial function. This disruption curtails oxidative metabolism, limiting adipocyte lipid metabolism and thermogenesis, a metabolically beneficial program that dissipates chemical energy as heat. Here, we show that PGC1α, a key governor of mitochondrial biogenesis, is negatively regulated at the level of its mRNA translation by the RNA-binding protein RBM43. RBM43 is induced by inflammatory cytokines and suppresses mitochondrial biogenesis in a PGC1α-dependent manner. In mice, adipocyte-selective Rbm43 disruption elevates PGC1α translation and oxidative metabolism. In obesity, Rbm43 loss improves glucose tolerance, reduces adipose inflammation, and suppresses activation of the innate immune sensor cGAS-STING in adipocytes. We further identify a role for PGC1α in safeguarding against cytoplasmic accumulation of mitochondrial DNA, a cGAS ligand. The action of RBM43 defines a translational regulatory axis by which inflammatory signals dictate cellular energy metabolism and contribute to metabolic disease pathogenesis.
Collapse
Affiliation(s)
- Phillip A Dumesic
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Sarah E Wilensky
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Symanthika Bose
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
40
|
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
|
41
|
Roselli C, Surakka I, Olesen MS, Sveinbjornsson G, Marston NA, Choi SH, Holm H, Chaffin M, Gudbjartsson D, Hill MC, Aegisdottir H, Albert CM, Alonso A, Anderson CD, Arking DE, Arnar DO, Barnard J, Benjamin EJ, Braunwald E, Brumpton B, Campbell A, Chami N, Chasman DI, Cho K, Choi EK, Christophersen IE, Chung MK, Conen D, Crijns HJ, Cutler MJ, Czuba T, Damrauer SM, Dichgans M, Dörr M, Dudink E, Duong T, Erikstrup C, Esko T, Fatkin D, Faul JD, Ferreira M, Freitag DF, Ganesh SK, Gaziano JM, Geelhoed B, Ghouse J, Gieger C, Giulianini F, Graham SE, Gudnason V, Guo X, Haggerty C, Hayward C, Heckbert SR, Hveem K, Ito K, Johnson R, Jukema JW, Jurgens SJ, Kääb S, Kane JP, Kany S, Kardia SLR, Kavousi M, Khurshid S, Kamanu FK, Kirchhof P, Kleber ME, Knight S, Komuro I, Krieger JE, Launer LJ, Li D, Lin H, Lin HJ, Loos RJF, Lotta L, Lubitz SA, Lunetta KL, Macfarlane PW, Magnusson PKE, Malik R, Mantineo H, Marcus GM, März W, McManus DD, Melander O, Melloni GEM, Meyre PB, Miyazawa K, Mohanty S, Monfort LM, Müller-Nurasyid M, Nafissi NA, Natale A, Nazarian S, Ostrowski SR, Pak HN, Pang S, Pedersen OB, et alRoselli C, Surakka I, Olesen MS, Sveinbjornsson G, Marston NA, Choi SH, Holm H, Chaffin M, Gudbjartsson D, Hill MC, Aegisdottir H, Albert CM, Alonso A, Anderson CD, Arking DE, Arnar DO, Barnard J, Benjamin EJ, Braunwald E, Brumpton B, Campbell A, Chami N, Chasman DI, Cho K, Choi EK, Christophersen IE, Chung MK, Conen D, Crijns HJ, Cutler MJ, Czuba T, Damrauer SM, Dichgans M, Dörr M, Dudink E, Duong T, Erikstrup C, Esko T, Fatkin D, Faul JD, Ferreira M, Freitag DF, Ganesh SK, Gaziano JM, Geelhoed B, Ghouse J, Gieger C, Giulianini F, Graham SE, Gudnason V, Guo X, Haggerty C, Hayward C, Heckbert SR, Hveem K, Ito K, Johnson R, Jukema JW, Jurgens SJ, Kääb S, Kane JP, Kany S, Kardia SLR, Kavousi M, Khurshid S, Kamanu FK, Kirchhof P, Kleber ME, Knight S, Komuro I, Krieger JE, Launer LJ, Li D, Lin H, Lin HJ, Loos RJF, Lotta L, Lubitz SA, Lunetta KL, Macfarlane PW, Magnusson PKE, Malik R, Mantineo H, Marcus GM, März W, McManus DD, Melander O, Melloni GEM, Meyre PB, Miyazawa K, Mohanty S, Monfort LM, Müller-Nurasyid M, Nafissi NA, Natale A, Nazarian S, Ostrowski SR, Pak HN, Pang S, Pedersen OB, Pedersen NL, Pereira AC, Pirruccello JP, Preuss M, Psaty BM, Pullinger CR, Rader DJ, Rämö JT, Ridker PM, Rienstra M, Risch L, Roden DM, Rotter JI, Sabatine MS, Schunkert H, Shah SH, Shim J, Shoemaker MB, Simonson B, Sinner MF, Smit RAJ, Smith JA, Smith NL, Smith JG, Soliman EZ, Sørensen E, Sotoodehnia N, Strbian D, Stricker BH, Teder-Laving M, Sun YV, Thériault S, Thorolfsdottir RB, Thorsteinsdottir U, Tveit A, van der Harst P, van Meurs J, Wang B, Weiss S, Wells QS, Weng LC, Wilson PW, Xiao L, Yang PS, Yao J, Yoneda ZT, Zeller T, Zeng L, Zhao W, Zhou X, Zöllner S, BioBank Japan Project, Regeneron Genetics Center, DBDS Genomic Consortium, Ruff CT, Bundgaard H, Willer C, Stefansson K, Ellinor PT. Meta-analysis of genome-wide associations and polygenic risk prediction for atrial fibrillation in more than 180,000 cases. Nat Genet 2025; 57:539-547. [PMID: 40050429 PMCID: PMC12094172 DOI: 10.1038/s41588-024-02072-3] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 12/30/2024] [Indexed: 03/15/2025]
Abstract
Atrial fibrillation (AF) is the most common heart rhythm abnormality and is a leading cause of heart failure and stroke. This large-scale meta-analysis of genome-wide association studies increased the power to detect single-nucleotide variant associations and found more than 350 AF-associated genetic loci. We identified candidate genes related to muscle contractility, cardiac muscle development and cell-cell communication at 139 loci. Furthermore, we assayed chromatin accessibility using assay for transposase-accessible chromatin with sequencing and histone H3 lysine 4 trimethylation in stem cell-derived atrial cardiomyocytes. We observed a marked increase in chromatin accessibility for our sentinel variants and prioritized genes in atrial cardiomyocytes. Finally, a polygenic risk score (PRS) based on our updated effect estimates improved AF risk prediction compared to the CHARGE-AF clinical risk score and a previously reported PRS for AF. The doubling of known risk loci will facilitate a greater understanding of the pathways underlying AF.
Collapse
Affiliation(s)
- Carolina Roselli
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ida Surakka
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Morten S Olesen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Nicholas A Marston
- TIMI Study Group, Boston, MA, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Seung Hoan Choi
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hilma Holm
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
| | - Mark Chaffin
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Daniel Gudbjartsson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | - Matthew C Hill
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Christine M Albert
- Department of Cardiology, Cedars-Sinai, Los Angeles, CA, USA
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Christopher D Anderson
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Dan E Arking
- McKusick Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David O Arnar
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Cardiovascular Centre, Landspitali-The National University Hospital of Iceland, Reykjavik, Iceland
| | - John Barnard
- Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Emelia J Benjamin
- Department of Medicine, Boston Medical Center, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Eugene Braunwald
- TIMI Study Group, Boston, MA, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ben Brumpton
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Medicine, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Nathalie Chami
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Kelly Cho
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
- Division of Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Eue-Keun Choi
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ingrid E Christophersen
- Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Gjettum, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Mina K Chung
- Cardiovascular Medicine, Heart Vascular and Thoracic Institute and Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland Clinic Lerner College of Medicine, Cleveland, OH, USA
| | - David Conen
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Harry J Crijns
- Department of Cardiology and CARIM, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Michael J Cutler
- Intermountain Heart Institute, Intermountain Medical Center, Murray, UT, USA
| | - Tomasz Czuba
- The Wallenberg Laboratory/Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University and the Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Cardiology, Lund University Diabetes Center, Wallenberg Center for Molecular Medicine, Clinical Sciences, Lund University and Skåne University Hospital, Lund, Sweden
| | - Scott M Damrauer
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Marcus Dörr
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
| | - Elton Dudink
- Department of Cardiology and CARIM, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - ThuyVy Duong
- McKusick Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christian Erikstrup
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Tõnu Esko
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Diane Fatkin
- Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
- UNSW Sydney, Sydney, New South Wales, Australia
- St Vincent's Hospital, Sydney, New South Wales, Australia
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Santhi K Ganesh
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - J Michael Gaziano
- Million Veteran Program, VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Bastiaan Geelhoed
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jonas Ghouse
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christian Gieger
- Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Franco Giulianini
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Sarah E Graham
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Vilmundur Gudnason
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Icelandic Heart Association, Kópavogur, Iceland
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Christopher Haggerty
- Department of Translational Data Science and Informatics, Geisinger, Danville, PA, USA
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Susan R Heckbert
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kaoru Ito
- Laboratory for Cardiovascular Genomics and Informatics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Renee Johnson
- Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
- UNSW Sydney, Sydney, New South Wales, Australia
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Centre, Leiden, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| | - Sean J Jurgens
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Stefan Kääb
- Department of Cardiology, University Hospital, LMU Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner site: Munich Heart Alliance, Munich, Germany
| | - John P Kane
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Shinwan Kany
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Cardiology, University Heart and Vascular Center UKE Hamburg, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Hamburg, Germany
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Shaan Khurshid
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Demoulas Center for Cardiac Arrhythmias and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Frederick K Kamanu
- TIMI Study Group, Boston, MA, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Paulus Kirchhof
- Department of Cardiology, University Heart and Vascular Center UKE Hamburg, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Hamburg, Germany
- Institute of Cardiovascular Sciences, Birmingham, UK
- AFNET, Münster, Germany
| | - Marcus E Kleber
- SYNLAB MVZ Humangenetik Mannheim, Mannheim, Germany
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Stacey Knight
- Intermountain Heart Institute, Intermountain Medical Center, Murray, UT, USA
- Department of Medicine, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Issei Komuro
- International University of Health and Welfare, Tokyo, Japan
- Department of Frontier Cardiovascular Science, The University of Tokyo, Tokyo, Japan
| | - Jose E Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of Sao Paulo, Sao Paulo, Brazil
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Dadong Li
- Regeneron Genetics Center, Tarrytown, NY, USA
| | - Honghuang Lin
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Henry J Lin
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Ruth J F Loos
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Luca Lotta
- Regeneron Genetics Center, Tarrytown, NY, USA
| | - Steven A Lubitz
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Demoulas Center for Cardiac Arrhythmias and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Kathryn L Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Peter W Macfarlane
- School of Health and Wellbeing, University of Glasgow, Glasgow, Scotland, UK
- Electrocardiology Group, New Lister Building, Royal Infirmary, Glasgow, Scotland, UK
| | - Patrik K E Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Rainer Malik
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Helene Mantineo
- Precision Cardiology Laboratory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Gregory M Marcus
- Division of Cardiology, University of California, San Francisco, CA, USA
| | - Winfried März
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Synlab Academy, SYNLAB Holding Deutschland GmbH, Mannheim, Germany
| | - David D McManus
- University of Massachusetts Medical School, Worcester, MA, USA
| | - Olle Melander
- Department of Internal Medicine, Lund University and Skåne University Hospital, Malmö, Sweden
| | | | - Pascal B Meyre
- Cardiovascular Research Institute Basel (CRIB), University Hospital Basel, Basel, Switzerland
| | - Kazuo Miyazawa
- Laboratory for Cardiovascular Genomics and Informatics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | | | - Laia M Monfort
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Martina Müller-Nurasyid
- IBE, Faculty of Medicine, LMU Munich, Munich, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Navid A Nafissi
- Division of Cardiology, Duke University School of Medicine, Durham, NC, USA
| | - Andrea Natale
- Texas Cardiac Arrhythmia Institute, Austin, TX, USA
- Department of Biomedicine and Prevention, Division of Cardiology, University of Tor Vergata, Rome, Italy
| | - Saman Nazarian
- Section of Cardiac Electrophysiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sisse R Ostrowski
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Hui-Nam Pak
- Yonsei University College of Medicine, Yonsei University Health System, Seoul, Republic of Korea
| | - Shichao Pang
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
| | - Ole B Pedersen
- Department of Clinical Immunology, Zealand University Hospital, Køge, Denmark
| | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Alexandre C Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of Sao Paulo, Sao Paulo, Brazil
| | - James P Pirruccello
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael Preuss
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology and Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Clive R Pullinger
- Cardiovascular Research Institute and Department of Physiological Nursing, University of California, San Francisco, CA, USA
| | - Daniel J Rader
- Division of Cardiovascular Medicine, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Joel T Rämö
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Michiel Rienstra
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Lorenz Risch
- Institute of Laboratory Medicine, Faculty of Medical Sciences, Private University of the Principality of Liechtenstein, Triesen, Liechtenstein
- Center of Laboratory Medicine, University Institute of Clinical Chemistry, University of Bern, Inselspital, Bern, Switzerland
| | - Dan M Roden
- Departments of Medicine Pharmacology and Biomedical Informatics, Divisions of Cardiovascular Medicine and Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
- Departments of Pediatrics and Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Marc S Sabatine
- TIMI Study Group, Boston, MA, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Heribert Schunkert
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- Deutsches Zentrum für Herz- und Kreislauferkrankungen (DZHK), Partner site: Munich Heart Alliance, Munich, Germany
| | - Svati H Shah
- Division of Cardiology, Duke University School of Medicine, Durham, NC, USA
| | - Jaemin Shim
- Korea University Cardiovascular Center, Seoul, Republic of Korea
| | - M Benjamin Shoemaker
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bridget Simonson
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Moritz F Sinner
- Department of Cardiology, University Hospital, LMU Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner site: Munich Heart Alliance, Munich, Germany
| | - Roelof A J Smit
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jennifer A Smith
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Nicholas L Smith
- Cardiovascular Health Research Unit and Department of Epidemiology, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
- Seattle Epidemiologic Research and Information Center, VA Puget Sound Health Care System, Seattle, WA, USA
| | - J Gustav Smith
- The Wallenberg Laboratory/Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University and the Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Cardiology, Lund University Diabetes Center, Wallenberg Center for Molecular Medicine, Clinical Sciences, Lund University and Skåne University Hospital, Lund, Sweden
| | - Elsayed Z Soliman
- Epidemiological Cardiology Research Center, Section on Cardiovascular Medicine, Department of Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Erik Sørensen
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Nona Sotoodehnia
- Division of Cardiology, Departments of Medicine and Epidemiology, University of Washington, Seattle, WA, USA
| | - Daniel Strbian
- Department of Neurology and Neurosciences, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Bruno H Stricker
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Yan V Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Atlanta VA Health Care System, Decatur, GA, USA
| | - Sébastien Thériault
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec City, Québec, Canada
| | | | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Arnljot Tveit
- Department of Medical Research, Bærum Hospital, Vestre Viken Hospital Trust, Gjettum, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Pim van der Harst
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Joyce van Meurs
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Biqi Wang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Stefan Weiss
- German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Quinn S Wells
- Departments of Medicine Pharmacology and Biomedical Informatics, Divisions of Cardiovascular Medicine and Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lu-Chen Weng
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - Peter W Wilson
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Atlanta VA Medical Center, Atlanta, GA, USA
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Ling Xiao
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Pil-Sung Yang
- Cha University College of Medicine, Seoul, Republic of Korea
| | - Jie Yao
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Zachary T Yoneda
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tanja Zeller
- German Center for Cardiovascular Research (DZHK), Hamburg, Germany
- University Center of Cardiovascular Sciences, Department of Cardiology, University Heart & Vascular Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Wei Zhao
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Xiang Zhou
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Sebastian Zöllner
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | - Christian T Ruff
- TIMI Study Group, Boston, MA, USA
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Henning Bundgaard
- Department of Cardiology, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Cristen Willer
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Kari Stefansson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Demoulas Center for Cardiac Arrhythmias and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
- Heart and Vascular Institute, Mass General Brigham, Boston, MA, USA.
| |
Collapse
|
42
|
Tadros R, Zheng SL, Grace C, Jordà P, Francis C, West DM, Jurgens SJ, Thomson KL, Harper AR, Ormondroyd E, Xu X, Theotokis PI, Buchan RJ, McGurk KA, Mazzarotto F, Boschi B, Pelo E, Lee M, Noseda M, Varnava A, Vermeer AMC, Walsh R, Amin AS, van Slegtenhorst MA, Roslin NM, Strug LJ, Salvi E, Lanzani C, de Marvao A, Roberts JD, Tremblay-Gravel M, Giraldeau G, Cadrin-Tourigny J, L'Allier PL, Garceau P, Talajic M, Gagliano Taliun SA, Pinto YM, Rakowski H, Pantazis A, Bai W, Baksi J, Halliday BP, Prasad SK, Barton PJR, O'Regan DP, Cook SA, de Boer RA, Christiaans I, Michels M, Kramer CM, Ho CY, Neubauer S, Matthews PM, Wilde AAM, Tardif JC, Olivotto I, Adler A, Goel A, Ware JS, Bezzina CR, Watkins H. Large-scale genome-wide association analyses identify novel genetic loci and mechanisms in hypertrophic cardiomyopathy. Nat Genet 2025; 57:530-538. [PMID: 39966646 PMCID: PMC11906354 DOI: 10.1038/s41588-025-02087-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 01/10/2025] [Indexed: 02/20/2025]
Abstract
Hypertrophic cardiomyopathy (HCM) is an important cause of morbidity and mortality with both monogenic and polygenic components. Here, we report results from a large genome-wide association study and multitrait analysis including 5,900 HCM cases, 68,359 controls and 36,083 UK Biobank participants with cardiac magnetic resonance imaging. We identified 70 loci (50 novel) associated with HCM and 62 loci (20 novel) associated with relevant left ventricular traits. Among the prioritized genes in the HCM loci, we identify a novel HCM disease gene, SVIL, which encodes the actin-binding protein supervillin, showing that rare truncating SVIL variants confer a roughly tenfold increased risk of HCM. Mendelian randomization analyses support a causal role of increased left ventricular contractility in both obstructive and nonobstructive forms of HCM, suggesting common disease mechanisms and anticipating shared response to therapy. Taken together, these findings increase our understanding of the genetic basis of HCM, with potential implications for disease management.
Collapse
Affiliation(s)
- Rafik Tadros
- Cardiovascular Genetics Centre and Research Centre, Montreal Heart Institute, Montreal, Quebec, Canada.
- Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada.
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands.
| | - 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 and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Christopher Grace
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Paloma Jordà
- Cardiovascular Genetics Centre and Research Centre, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Catherine Francis
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Dominique M West
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Sean J Jurgens
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kate L Thomson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
- Oxford Genetics Laboratories, Churchill Hospital, Oxford, UK
| | - Andrew R Harper
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Elizabeth Ormondroyd
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Xiao Xu
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
| | - Pantazis I Theotokis
- National Heart and Lung Institute, Imperial College London, London, UK
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Rachel J Buchan
- National Heart and Lung Institute, Imperial College London, London, UK
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Kathryn A McGurk
- National Heart and Lung Institute, Imperial College London, London, UK
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
| | - Francesco Mazzarotto
- National Heart and Lung Institute, Imperial College London, London, UK
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | | | - Michael Lee
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Michela Noseda
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Amanda Varnava
- National Heart and Lung Institute, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, Imperial College London, London, UK
| | - Alexa M C Vermeer
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Clinical Genetics, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART), Amsterdam, the Netherlands
| | - Roddy Walsh
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Ahmad S Amin
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART), Amsterdam, the Netherlands
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Marjon A van Slegtenhorst
- Department of Clinical Genetics, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Nicole M Roslin
- Program in Genetics and Genome Biology and The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lisa J Strug
- Program in Genetics and Genome Biology and The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Departments of Statistical Sciences and Computer Science, University of Toronto, Toronto, Ontario, Canada
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Erika Salvi
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico 'Carlo Besta', Milan, Italy
| | - Chiara Lanzani
- Genomics of Renal Diseases and Hypertension Unit and Nephrology Operative Unit, IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Antonio de Marvao
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
- King's College London, London, UK
| | - Jason D Roberts
- Department of Medicine, Section of Cardiac Electrophysiology, Division of Cardiology, Western University, London, Ontario, Canada
| | - Maxime Tremblay-Gravel
- Cardiovascular Genetics Centre and Research Centre, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Genevieve Giraldeau
- Cardiovascular Genetics Centre and Research Centre, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Julia Cadrin-Tourigny
- Cardiovascular Genetics Centre and Research Centre, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Philippe L L'Allier
- Cardiovascular Genetics Centre and Research Centre, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Patrick Garceau
- Cardiovascular Genetics Centre and Research Centre, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Mario Talajic
- Cardiovascular Genetics Centre and Research Centre, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Sarah A Gagliano Taliun
- Cardiovascular Genetics Centre and Research Centre, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Yigal M Pinto
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART), Amsterdam, the Netherlands
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Harry Rakowski
- Division of Cardiology, Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada
| | - Antonis Pantazis
- Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Wenjia Bai
- Department of Computing, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
- Data Science Institute, Imperial College London, London, UK
| | - John Baksi
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Brian P Halliday
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Sanjay K Prasad
- National Heart and Lung Institute, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Paul J R Barton
- National Heart and Lung Institute, Imperial College London, London, UK
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
- Royal Brompton and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Declan P O'Regan
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
| | - Stuart A Cook
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
- National Heart Centre, Singapore, Singapore
- Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Rudolf A de Boer
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Imke Christiaans
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Michelle Michels
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART), Amsterdam, the Netherlands
- Department of Cardiology, Thorax Center, Cardiovascular Institute, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Christopher M Kramer
- Department of Medicine, Cardiovascular Division, University of Virginia Health, Charlottesville, VA, USA
| | - Carolyn Y Ho
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Stefan Neubauer
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
| | - Arthur A M Wilde
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART), Amsterdam, the Netherlands
- Department of Clinical Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
- ECGen, Cardiogenetics Focus Group of EHRA, Biot, France
| | - Jean-Claude Tardif
- Cardiovascular Genetics Centre and Research Centre, Montreal Heart Institute, Montreal, Quebec, Canada
- Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | | | - Arnon Adler
- Division of Cardiology, Peter Munk Cardiac Centre, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Anuj Goel
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, 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 and Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Connie R Bezzina
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, University of Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands.
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART), Amsterdam, the Netherlands.
| | - Hugh Watkins
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK.
| |
Collapse
|
43
|
Minikel EV, Nelson MR. Human genetic evidence enriched for side effects of approved drugs. PLoS Genet 2025; 21:e1011638. [PMID: 40163513 PMCID: PMC11977994 DOI: 10.1371/journal.pgen.1011638] [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: 06/17/2024] [Revised: 04/08/2025] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
Safety failures are an important factor in low drug development success rates. Human genetic evidence can select drug targets causal in disease and enrich for successful programs. Here, we sought to determine whether human genetic evidence can also enrich for labeled side effects (SEs) of approved drugs. We combined the SIDER database of SEs with human genetic evidence from genome-wide association studies, Mendelian disease, and somatic mutations. SEs were 2.0 times more likely to occur for drugs whose target possessed human genetic evidence for a trait similar to the SE. Enrichment was highest when the trait and SE were most similar to each other, and was robust to removing drugs where the approved indication was also similar to the SE. The enrichment of genetic evidence was greatest for SEs that were more drug specific, affected more people, and were more severe. There was significant heterogeneity among disease areas the SEs mapped to, with the highest positive predictive value for cardiovascular SEs. This supports the integration of human genetic evidence early in the drug discovery process to identify potential SE risks to be monitored or mitigated in the course of drug development.
Collapse
Affiliation(s)
- Eric Vallabh Minikel
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, Massachusetts, United States of America
| | - Matthew R. Nelson
- Deerfield Management Company, L.P, New York City, New York, United States of America
- Genscience LLC, New York City, New York, United States of America
| |
Collapse
|
44
|
Loh NY, Vasan SK, Rosoff DB, Roberts E, van Dam AD, Verma M, Phillips D, Wesolowska-Andersen A, Neville MJ, Noordam R, Ray DW, Tobias JH, Gregson CL, Karpe F, Christodoulides C. LRP5 promotes adipose progenitor cell fitness and adipocyte insulin sensitivity. COMMUNICATIONS MEDICINE 2025; 5:51. [PMID: 40000740 PMCID: PMC11862225 DOI: 10.1038/s43856-025-00774-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND WNT signaling plays a key role in postnatal bone formation. Individuals with gain-of-function mutations in the WNT co-receptor LRP5 exhibit increased lower-body fat mass and potentially enhanced glucose metabolism, alongside high bone mass. However, the mechanisms by which LRP5 regulates fat distribution and its effects on systemic metabolism remain unclear. This study aims to explore the role of LRP5 in adipose tissue biology and its impact on metabolism. METHODS Metabolic assessments and imaging were conducted on individuals with gain- and loss-of-function LRP5 mutations, along with age- and BMI-matched controls. Mendelian randomization analyses were used to investigate the relationship between bone, fat distribution, and systemic metabolism. Functional studies and RNA sequencing were performed on abdominal and gluteal adipose cells with LRP5 knockdown. RESULTS Here we show that LRP5 promotes lower-body fat distribution and enhances systemic and adipocyte insulin sensitivity through cell-autonomous mechanisms, independent of its bone-related functions. LRP5 supports adipose progenitor cell function by activating WNT/β-catenin signaling and preserving valosin-containing protein (VCP)-mediated proteostasis. LRP5 expression in adipose progenitors declines with age, but gain-of-function LRP5 variants protect against age-related fat loss in the lower body. CONCLUSIONS Our findings underscore the critical role of LRP5 in regulating lower-body fat distribution and insulin sensitivity, independent of its effects on bone. Pharmacological activation of LRP5 in adipose tissue may offer a promising strategy to prevent age-related fat redistribution and metabolic disorders.
Collapse
Affiliation(s)
- Nellie Y Loh
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Senthil K Vasan
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Daniel B Rosoff
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Emile Roberts
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Andrea D van Dam
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Manu Verma
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Daniel Phillips
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Agata Wesolowska-Andersen
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Matt J Neville
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - David W Ray
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK
| | - Jonathan H Tobias
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, Southmead Hospital, University of Bristol, Bristol, UK
| | - Celia L Gregson
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, Southmead Hospital, University of Bristol, Bristol, UK
| | - Fredrik Karpe
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK
| | - Constantinos Christodoulides
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK.
- NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK.
| |
Collapse
|
45
|
Nurtdinov RN, Guigó R. EPIraction - an atlas of candidate enhancer-gene interactions in human tissues and cell lines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.638885. [PMID: 40027634 PMCID: PMC11870479 DOI: 10.1101/2025.02.18.638885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Enhancer-gene maps are essential to understand gene regulation. The identification of enhancers contributing to regulating a given gene, however, is challenging as they may lie far from the gene and act in a tissue-specific manner. Here we present EPIraction, an enhancer-gene atlas in the human genome. As H3K27ac is a typical mark of enhancer function, EPIraction relies on an exhaustive collection of 1,538 H3K27ac ChIP-Seq datasets. We used these data together with other epigenomic data to measure enhancer activity. To predict enhancer-gene interactions we first applied the conventional Activity By Contact algorithm to score enhancer-gene pairs in individual tissues. Then, we re-scored the interaction rewarding those present in biologically similar tissues. We used our predictions to annotate the candidate target genes for 1,664,956 SNPs from 3,693 UK Biobank and 760 FinnGen GWAS traits. The EPIraction predictions can be accessed through the portal at https://epiraction.crg.es . The portal can be employed to produce annotations for user-provided lists of SNPs or genomic intervals.
Collapse
|
46
|
Seagle HM, Akerele AT, DeCorte JA, Hellwege JN, Breeyear JH, Kim J, Levin M, Khodurksy S, Bress A, Lee K, Meiler J, Gill D, Lee JS, Heberer K, Miller DR, Reaven P, Chang KM, Lynch JA, Khankari NK, Shuey MM, Edwards TL, Vujkovic M. Genomics-Informed Drug Repurposing Strategy Identifies Novel Therapeutic Targets for Metabolic Dysfunction-Associated Steatotic Liver Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.18.25321035. [PMID: 40034783 PMCID: PMC11875238 DOI: 10.1101/2025.02.18.25321035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Identification of drug-repurposing targets with genetic and biological support is an economically and temporally efficient strategy for improving treatment of diseases. We employed a cross-disciplinary approach to identify potential treatments for metabolic dysfunction associated steatotic liver disease (MASLD) using humans as a model organism. We identified 212 putative causal genes associated with MASLD using data from a large multi-ancestry genetic association study, of which 158 (74.5%) are novel. From this set we identified 57 genes that encode for druggable protein targets, and where the effects of increasing genetically predicted gene expression on MASLD risk align with the function of that drug on the protein target. These potential targets were then evaluated for evidence of efficacy using Mendelian randomization, pathway analysis, and protein structural modeling. Using these approaches, we present compelling evidence to suggest activation of FADS1 by icosopent ethyl as well as S1PR2 by fingolimod could be promising therapeutic strategies for MASLD.
Collapse
Affiliation(s)
- Hannah M Seagle
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Joseph Maxwell Cleland Atlanta VA Medical Center, Atlanta, Georgia, United States of America
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Alexis T Akerele
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- School of Graduate Studies and Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
- Division of Quantitative Science, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Joseph A DeCorte
- Vanderbilt Medical Scientist Training Program, Vanderbilt University Medical Center, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Department of Chemical and Physical Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Jacklyn N Hellwege
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- VA Tennessee Valley Healthcare System (626), Nashville, Tennessee, United States of America
| | - Joseph H Breeyear
- Biostatistics and Computational Biology Branch, National Institute for Environmental Health Sciences, National Institutes of Health, Durham, North Carolina, United States of America
| | - Jeewoo Kim
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Division of Quantitative Science, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Medical Scientist Training Program, Vanderbilt University Medical Center, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Michael Levin
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Phoenix VA Health Care System; University of Arizona, Phoenix, Arizona, United States of
| | - Samuel Khodurksy
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Phoenix VA Health Care System; University of Arizona, Phoenix, Arizona, United States of
| | - Adam Bress
- Salt Lake City VA Medical Center, Salt Lake City, Utah, United States of America
- University of Utah, School of Medicine, Salt Lake City, Utah, United States of America
| | - Kyung Lee
- Salt Lake City VA Medical Center, Salt Lake City, Utah, United States of America
| | - Jens Meiler
- Department of Chemical and Physical Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Institute for Drug Discovery, Leipzig University Medical School, Leipzig, Germany
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Jennifer S Lee
- Stanford University, Stanford, California, United States of America
- Palo Alto VA Medical Center, Palo Alto, California, United States of America
| | - Kent Heberer
- Palo Alto VA Medical Center, Palo Alto, California, United States of America
| | - Donald R Miller
- VA Center for Medication Safety, Department of Veterans Affairs, Chicago, Illinois, United States of America
- Center for Population Health, Department of Biomedical and Nutritional Sciences, University of Massachusetts, Lowell, MA, United States of America
| | - Peter Reaven
- Phoenix VA Health Care System; University of Arizona, Phoenix, Arizona, United States of
| | - Kyong-Mi Chang
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Corporal Michael J. Crescenz Philadelphia VA Medical Center, Philadelphia, Pennsylvania, United States of America
| | - Julie A Lynch
- Salt Lake City VA Medical Center, Salt Lake City, Utah, United States of America
- University of Utah, School of Medicine, Salt Lake City, Utah, United States of America
| | - Nikhil K Khankari
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Megan M Shuey
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- VA Tennessee Valley Healthcare System (626), Nashville, Tennessee, United States of America
| | - Marijana Vujkovic
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Corporal Michael J. Crescenz Philadelphia VA Medical Center, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
47
|
Zou X, Li D, Zhang L, Shen J. Genetically predicted inflammatory proteins mediate the association between gut microbiota and renal cell carcinoma. Discov Oncol 2025; 16:216. [PMID: 39976778 PMCID: PMC11842667 DOI: 10.1007/s12672-025-01980-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 02/12/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Studies have indicated a potential relationship between gut microbiota and renal cell carcinoma. However, the causal relationship between various types of gut microbiota and renal cell carcinoma, as well as the role of inflammatory protein as mediators, remains unclear. METHODS This study aimed to identify the relationship between gut microbiota, inflammatory protein, and renal cell cancer through a large-scale genome-wide association study (GWAS) utilizing pooled data. We employed Mendelian randomization (MR) to investigate the causal relationship among these variables. Inverse variance weighting (IVW) was utilized as the primary statistical method. Furthermore, we examined the mediating role of inflammatory protein in the pathway through which gut microbiota influences the development of renal cell cancer. RESULTS The analysis revealed 12 positive causal relationships and 15 negative causal relationships between the genetic liability of gut microbiota and renal cell cancer. Furthermore, there were three positive causal relationships and one negative causal relationship between inflammatory proteins and renal cell cancer. There were two axes of relationships in which gut microbiota promote the development of renal cell cancer. through inflammatory proteins acting as mediators. CONCLUSIONS Gut microbiota and inflammatory protein were causally related to renal cell cancer, and inflammatory protein were intermediary factors in the pathway between gut microbiota and renal cell cancer.
Collapse
Affiliation(s)
- Xinyun Zou
- Department of Oncology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Dong Li
- Department of Oncology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Ling Zhang
- Department of Oncology, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Jinlan Shen
- Department of Laboratory Medicine, The General Hospital of Western Theater Command, Chengdu 610083, China.
| |
Collapse
|
48
|
Kelchtermans J, March ME, Mentch F, Qu H, Liu Y, Nguyen K, Hakonarson H. Genetic modifiers of asthma response to air pollution in children: An African ancestry GWAS and PM 2.5 polygenic risk score study. ENVIRONMENTAL RESEARCH 2025; 267:120666. [PMID: 39725137 PMCID: PMC11800831 DOI: 10.1016/j.envres.2024.120666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
RATIONALE Ambient air pollution (AAP) is linked to asthma outcomes, but predicting individual risk remains challenging. Understanding genetic contributors to AAP sensitivity may help overcome this gap. OBJECTIVES To determine if single nucleotide polymorphisms (SNPs) are associated with AAP sensitivity in children with asthma. METHODS We complete a GWAS in pediatric patients with asthma frequently exposed to AAP, comparing patients with exacerbations following spikes in AAP to patients without this temporal association and calculate a polygenic risk score (PRS) for PM2.5. This PRS was validated using internal data and data from the All of Us cohort. MEASUREMENTS AND RESULTS We included 6023 patients in the GWAS, restricted to the African ancestry cohort due to the association between AAP exposure and race. Three loci reached genome-wide significance, including rs111970601, associated with CO sensitivity (odds ratio [OR] 6.58; P = 1.63 × 10-8) and rs9836522 with PM2.5 sensitivity (OR 0.75; P = 3,87 × 10-9), both externally validated. PRS z-scores were associated with increased asthma exacerbations in patients frequently exposed to poor air quality (β = 0.15; P = 2.67 × 10⁻⁵). Spirometry data from 4138 patients showed that having a high PRS was associated with lower FVC z-scores in patients frequently exposed to AAP (β = -0.44; P = 0.035). External validation confirmed a significant interaction between high PRS and frequent AAP exposure (β = 0.30; P = 0.012) CONCLUSIONS: We associate specific SNPs with AAP-related asthma exacerbations and introduce a PM2.5 sensitivity PRS, paving the way for future research aimed at protecting genetically predisposed patients.
Collapse
Affiliation(s)
- Jelte Kelchtermans
- Perelman School of Medicine, University of Pennsylvania, 3451 Walnut St, Philadelphia, PA, 19104, USA; The Center of Applied Genomics, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, 19104, Philadelphia, PA, USA; Division of Pulmonary and Sleep Medicine, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, 19104, Philadelphia, PA, USA.
| | - Michael E March
- The Center of Applied Genomics, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, 19104, Philadelphia, PA, USA
| | - Frank Mentch
- The Center of Applied Genomics, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, 19104, Philadelphia, PA, USA
| | - Huiqi Qu
- The Center of Applied Genomics, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, 19104, Philadelphia, PA, USA
| | - Yichuan Liu
- The Center of Applied Genomics, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, 19104, Philadelphia, PA, USA
| | - Kenny Nguyen
- The Center of Applied Genomics, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, 19104, Philadelphia, PA, USA
| | - Hakon Hakonarson
- Perelman School of Medicine, University of Pennsylvania, 3451 Walnut St, Philadelphia, PA, 19104, USA; The Center of Applied Genomics, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, 19104, Philadelphia, PA, USA; Division of Pulmonary and Sleep Medicine, The Children's Hospital of Philadelphia, 3615 Civic Center Blvd, 19104, Philadelphia, PA, USA
| |
Collapse
|
49
|
Liu R, Fan W, Hu J, Xu K, Huang Z, Liu Y, Sun C. The mediating role of thyroid-related hormones between thyroid dysfunction diseases and osteoporosis: a mediation mendelian randomization study. Sci Rep 2025; 15:4121. [PMID: 39901040 PMCID: PMC11791035 DOI: 10.1038/s41598-025-88412-7] [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: 07/25/2024] [Accepted: 01/28/2025] [Indexed: 02/05/2025] Open
Abstract
The links between Thyroid dysfunction diseases (TDFDs) and osteoporosis (OP) has received widespread attention, but the causal relationships and mediating factors have not been systematically studied. We used two-sample Mendelian randomization (MR) analysis to elucidate the causal relationship between TDFDs and OP. Moreover, we performed mediation MR analyses to explore the role of thyroid-related hormones and OP risk factors in the association between TDFDs and OP. Two sample MR analyses showed that hyperthyroidism increased OP (OR = 1.080, 95% CI 1.026 to 1.137; P = 0.0032) risk. Hypothyroidism increases OP (OR = 1.183, 95% CI 1.125 to 1.244; P < 0.0001) risk. Furthermore, mediation analysis revealed that TSH mediated 5.314% of the relationship between hypothyroidism and OP. In contrast, FT4 mediated 9.670% of the relationship between hyperthyroidism and OP. In European populations, TDFDs may increase OP risk. TSH mediates in the causal association between hypothyroidism and OP, and similarly, FT4 mediates in the causal link between hyperthyroidism and OP. Our findings underscore the significance of improving integrative care for individuals with TDFDs to mitigate the risk of OP. It is essential to maintain stable levels of thyroid hormones and closely monitor bone health to effectively mitigate and prevent OP.
Collapse
Affiliation(s)
- Ruikang Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Weiming Fan
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Jiaming Hu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Ke Xu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Zhijian Huang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Liu
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China.
- Hubei Sizhen Laboratory, Hubei, China.
- Affiliated Hospital of Hubei University of Chinese Medicine, Hubei, China.
| | - Chiyun Sun
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
50
|
Adli M, Przybyla L, Burdett T, Burridge PW, Cacheiro P, Chang HY, Engreitz JM, Gilbert LA, Greenleaf WJ, Hsu L, Huangfu D, Hung LH, Kundaje A, Li S, Parkinson H, Qiu X, Robson P, Schürer SC, Shojaie A, Skarnes WC, Smedley D, Studer L, Sun W, Vidović D, Vierbuchen T, White BS, Yeung KY, Yue F, Zhou T. MorPhiC Consortium: towards functional characterization of all human genes. Nature 2025; 638:351-359. [PMID: 39939790 DOI: 10.1038/s41586-024-08243-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 10/17/2024] [Indexed: 02/14/2025]
Abstract
Recent advances in functional genomics and human cellular models have substantially enhanced our understanding of the structure and regulation of the human genome. However, our grasp of the molecular functions of human genes remains incomplete and biased towards specific gene classes. The Molecular Phenotypes of Null Alleles in Cells (MorPhiC) Consortium aims to address this gap by creating a comprehensive catalogue of the molecular and cellular phenotypes associated with null alleles of all human genes using in vitro multicellular systems. In this Perspective, we present the strategic vision of the MorPhiC Consortium and discuss various strategies for generating null alleles, as well as the challenges involved. We describe the cellular models and scalable phenotypic readouts that will be used in the consortium's initial phase, focusing on 1,000 protein-coding genes. The resulting molecular and cellular data will be compiled into a catalogue of null-allele phenotypes. The methodologies developed in this phase will establish best practices for extending these approaches to all human protein-coding genes. The resources generated-including engineered cell lines, plasmids, phenotypic data, genomic information and computational tools-will be made available to the broader research community to facilitate deeper insights into human gene functions.
Collapse
Affiliation(s)
- Mazhar Adli
- Robert H. Lurie Comprehensive Cancer Center, Department of Obstetrics and Gynecology, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| | - Laralynne Przybyla
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Tony Burdett
- Omics Section, European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Hinxton, UK
| | - Paul W Burridge
- Department of Pharmacology, Center for Pharmacogenomics, Northwestern University, Feinberg School of Medicine, Evanston, IL, USA
| | - Pilar Cacheiro
- William Harvey Research Institute, Clinical Pharmacology and Precision Medicine, Queen Mary University of London, London, UK
| | - Howard Y Chang
- Department of Dermatology, Stanford University, Stanford, CA, USA
| | - Jesse M Engreitz
- Department of Genetics, Stanford University, Stanford, CA, USA
- Basic Science and Engineering (BASE) Initiative, Stanford University, Stanford, CA, USA
| | - Luke A Gilbert
- Department of Urology, University of California, San Francisco, CA, USA
| | | | - Li Hsu
- Department of Biostatistics, Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Danwei Huangfu
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Ling-Hong Hung
- School of Engineering and Technology, University of Washington Tacoma, Tacoma, WA, USA
| | - Anshul Kundaje
- Departments of Genetics and Computer Science, Stanford University, Stanford, CA, USA
| | - Sheng Li
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Helen Parkinson
- Knowledge Management Section, European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Hinxton, UK
| | - Xiaojie Qiu
- Basic Science and Engineering (BASE) Initiative, Stanford University, Stanford, CA, USA
- Departments of Genetics and Computer Science, Stanford University, Stanford, CA, USA
| | - Paul Robson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Stephan C Schürer
- Molecular and Cellular Pharmacology; Sylvester Comprehensive Cancer Center, University of Miami, Coral Gables, FL, USA
| | - Ali Shojaie
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | | | - Damian Smedley
- William Harvey Research Institute, Clinical Pharmacology and Precision Medicine, Queen Mary University of London, London, UK
| | - Lorenz Studer
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Wei Sun
- Department of Biostatistics, Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Dušica Vidović
- Molecular and Cellular Pharmacology; Sylvester Comprehensive Cancer Center, University of Miami, Coral Gables, FL, USA
| | - Thomas Vierbuchen
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Brian S White
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Ka Yee Yeung
- School of Engineering and Technology, University of Washington Tacoma, Tacoma, WA, USA
| | - Feng Yue
- Department of Biochemistry and Molecular Genetics, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA
| | - Ting Zhou
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| |
Collapse
|