1
|
Byun J, Han Y, Choi J, Sun R, Shaw VR, Zhu C, Xiao X, Lusk C, Badr H, Lee HS, Jang HJ, Li Y, Lim H, Long E, Liu Y, Kachuri L, Walsh KM, Wiencke JK, Albanes D, Lam S, Tardon A, Neuhouser ML, Barnett MJ, Chen C, Bojesen S, Brenner H, Landi MT, Johansson M, Risch A, Wichmann HE, Bickeböller H, Christiani DC, Rennert G, Arnold S, Field JK, Shete S, Le Marchand L, Liu G, Andrew AS, Zienolddiny S, Grankvist K, Johansson M, Caporaso N, Taylor F, Lazarus P, Schabath MB, Aldrich MC, Patel A, Lin X, Zanetti KA, Harris CC, Chanock S, McKay J, Schwartz AG, Hung RJ, Amos CI. Genome-wide association study for lung cancer in 6531 African Americans reveals new susceptibility loci. Hum Mol Genet 2025:ddaf059. [PMID: 40341939 DOI: 10.1093/hmg/ddaf059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 03/31/2025] [Accepted: 04/09/2025] [Indexed: 05/11/2025] Open
Abstract
Despite lung cancer affecting all races and ethnicities, disparities are observed in incidence and mortality rates among different ethnic groups in the United States. Non-Hispanic African Americans had a high incidence rate of lung cancer at 55.8 per 100 000 people, as well as the highest death rate at 37.2 per 100 000 people from 2016 to 2020. While previous genome-wide association studies (GWAS) have identified over 45 susceptibility risk loci that influence lung cancer development, few GWAS have investigated the etiology of lung cancer in African Americans. To address this gap in knowledge, we conducted GWAS of lung cancer focused on studying African Americans, comprising 2267 lung cancer cases and 4264 controls. We identified three loci associated with lung cancer, one with lung adenocarcinoma, and four with lung squamous cell carcinoma in this population at the genomic-wide significance level. Among them, three novel loci were identified near VWF at 12p13.31 for overall lung cancer and GACAT3 at 2p24.3 and LMAN1L at 15q24.1 for lung squamous cell carcinoma. In addition, we confirmed previously reported risk loci with known or new lead variants near CHRNA5 at 15q25.1 and CYP2A6 at 19q13.2 associated with lung cancer and TRIP13 at 5p15.33 and ERC1 at 12p13.33 associated with lung squamous cell carcinoma. Further multi-step functional analyses shed light on biological mechanisms underlying these associations of lung cancer in this population. Our study highlights the importance of ancestry-specific studies for the potential alleviation of lung cancer burden in African Americans.
Collapse
Affiliation(s)
- Jinyoung Byun
- Institute for Clinical and Translational Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- University of New Mexico Comprehensive Cancer Center, 1201 Camino de Salud NE, Albuquerque, NM, 87102, United States
| | - Younghun Han
- Institute for Clinical and Translational Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- University of New Mexico Comprehensive Cancer Center, 1201 Camino de Salud NE, Albuquerque, NM, 87102, United States
| | - Jiyeon Choi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9615 Medical Center Drive, Rockville, MD, 20850, United States
| | - Ryan Sun
- Department of Biostatistics, University of Texas, M.D. Anderson Cancer Center, 7007 Bertner Ave, Houston, TX, 77030, United States
| | - Vikram R Shaw
- Institute for Clinical and Translational Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Catherine Zhu
- Institute for Clinical and Translational Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Xiangjun Xiao
- Institute for Clinical and Translational Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Christine Lusk
- Department of Oncology, Wayne State University School of Medicine, 4100 John R, Detroit, MI, 48201, United States
- Karmanos Cancer Institute, 4100 John R Street, Detroit, MI, 48201, United States
| | - Hoda Badr
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Hyun-Sung Lee
- Systems Onco-Immunology Lab, David Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Hee-Jin Jang
- Systems Onco-Immunology Lab, David Sugarbaker Division of Thoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Yafang Li
- Institute for Clinical and Translational Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- University of New Mexico Comprehensive Cancer Center, 1201 Camino de Salud NE, Albuquerque, NM, 87102, United States
| | - Hyeyeun Lim
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - 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 100005, China
| | - Yanhong Liu
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
| | - Linda Kachuri
- Department of Epidemiology and Population Health, Stanford University, 300 Pasteur Drive, Stanford, CA, 94305, United States
| | - Kyle M Walsh
- Duke Cancer Institute, Duke University Medical Center, 20 Duke Medicine Cir, Durham, NC, 27701, United States
| | - John K Wiencke
- Department of Neurological Surgery, The University of California, San Francisco, 400 Parnassus Ave, San Francisco, CA, 94143, United States
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9615 Medical Center Drive, Rockville, MD, 20850, United States
| | - Stephen Lam
- Department of Integrative Oncology, University of British Columbia, 675 West 10th Ave, Vancouver, BC V5Z 1L3, Canada
| | - Adonina Tardon
- Public Health Department, University of Oviedo, and Health Research Institute of Asturias, ISPA, Av. del Hospital Universitario, s/n, 33011 Oviedo, Asturias, Spain
| | - Marian L Neuhouser
- Program in Cancer Prevention, Public Health Sciences Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, United States
| | - Matt J Barnett
- Program in Cancer Prevention, Public Health Sciences Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, United States
| | - Chu Chen
- Program in Cancer Prevention, Public Health Sciences Division, Fred Hutchinson Cancer Center, 1100 Fairview Ave N, Seattle, WA, 98109, United States
| | - Stig Bojesen
- Department of Clinical Biochemistry, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Deutsches Krebsforschungszentrum (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 581, 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9615 Medical Center Drive, Rockville, MD, 20850, United States
| | - Mattias Johansson
- Section of Genetics, International Agency for Research on Cancer, World Health Organization, 25 avenue Tony Garnier, CS 90627, 69366 LYON CEDEX 07, France
| | - Angela Risch
- Translational Lung Research Center Heidelberg (TLRC-H), German Center for Lung Research (DZL), Im Neuenheimer Feld 156, 69120, Heidelberg, Germany
- Division of Cancer Epigenomics, DKFZ-German Cancer Research Center, Im Neuenheimer Feld 280, D-69120, Heidelberg, Germany
- Department of Biosciences and Medical Biology, Center for Tumor Biology and Immunology, University of Salzburg and Cancer Cluster Hellbrunner Strasse 34, Salzburg, 5020, Austria
| | - H-Erich Wichmann
- Helmholtz-Munich Institute of Epidemiology, Ingolstädter Landstraße 1, Neuherberg, 85764, Germany
| | - Heike Bickeböller
- University Medical Center Göttingen, Institute of Genetic Epidemiology, Humboldtallee 32, 37073 Göttingen, Germany
| | - David C Christiani
- Department of Environmental Health and Epidemiology, Harvard T.H.Chan School of Public Health, 665 Huntington Avenue, Building 1, Boston, MA, 02115, United States
| | - Gad Rennert
- Clalit National Cancer Control Center at Carmel Medical Center and Technion Faculty of Medicine, Mikhal St 7, Haifa, 3436212, Israel
| | - Susanne Arnold
- University of Kentucky, Markey Cancer Center, 800 Rose Street, Lexington, KY, 40536, United States
| | - John K Field
- Institute of Translational Medicine, University of Liverpool, the Sherrington Building, Ashton St, Liverpool, L69 3GE, United Kingdom
| | - Sanjay Shete
- Department of Biostatistics, University of Texas, M.D. Anderson Cancer Center, 7007 Bertner Ave, Houston, TX, 77030, United States
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, United States
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 96813, United States
| | - Geoffrey Liu
- University Health Network- The Princess Margaret Cancer Centre, 610 University Ave, Toronto, ON M5G 2M9, Canada
| | - Angeline S Andrew
- Departments of Epidemiology and Community and Family Medicine, Dartmouth College, 1 Rope Ferry Road, Hanover, NH, 03755, United States
| | | | - Kjell Grankvist
- Department of Medical Biosciences, Umeå University, 901 87 Umeå, Sweden
| | - Mikael Johansson
- Department of Radiation Sciences, Oncology, Umeå University, 901 87 Umeå, Sweden
| | - Neil Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9615 Medical Center Drive, Rockville, MD, 20850, United States
| | - Fiona Taylor
- Sheffield Teaching Hospitals Foundation Trust, 8 Beech Hill Road, Sheffield, S10 2SB, United Kingdom
| | - Philip Lazarus
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, 412 East Spokane Falls Blvd, PBS 130, Spokane, WA, 99202, United States
| | - Matthew B Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL, 33612, United States
| | - Melinda C Aldrich
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, 1161 21st Ave S, Nashville, TN, 37232, United States
| | - Alpa Patel
- American Cancer Society, Inc., 270 Peachtree Street NW, Atlanta, GA, 30303, United States
| | - Xihong Lin
- Department of Biostatistics, Harvard TH Chan School of Public Health, 655 Huntington Avenue, Boston, MA, 02115, United States
| | - Krista A Zanetti
- Office of Nutrition Research, Division of Program Coordination, Planning, and Strategic Initiatives, Office of the Director, National Institutes of Health, 6705 Rockledge Drive, Bethesda, MD, 20817, United States
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, 37 Convent Dr, Bethesda, MD, 20892, United States
| | - Stephen Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9615 Medical Center Drive, Rockville, MD, 20850, United States
| | - James McKay
- Section of Genetics, International Agency for Research on Cancer, World Health Organization, 25 avenue Tony Garnier, CS 90627, 69366 LYON CEDEX 07, France
| | - Ann G Schwartz
- Department of Oncology, Wayne State University School of Medicine, 4100 John R, Detroit, MI, 48201, United States
- Karmanos Cancer Institute, 4100 John R Street, Detroit, MI, 48201, United States
| | - Rayjean J Hung
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 600 University Ave, Toronto, ON M5G 1X5, Canada
- Division of Epidemiology, Dalla Lana School of Public Health, University of Toronto, 155 College Street, Toronto, Ontario, M5T 3M7, Canada
| | - Christopher I Amos
- Institute for Clinical and Translational Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, United States
- University of New Mexico Comprehensive Cancer Center, 1201 Camino de Salud NE, Albuquerque, NM, 87102, United States
| |
Collapse
|
2
|
Gui A, Hollowell A, Wigdor EM, Morgan MJ, Hannigan LJ, Corfield EC, Odintsova V, Hottenga JJ, Wong A, Pool R, Cullen H, Wilson S, Warrier V, Eilertsen EM, Andreassen OA, Middeldorp CM, St Pourcain B, Bartels M, Boomsma DI, Hartman CA, Robinson EB, Arichi T, Edwards AD, Johnson MH, Dudbridge F, Sanders SJ, Havdahl A, Ronald A. Genome-wide association meta-analysis of age at onset of walking in over 70,000 infants of European ancestry. Nat Hum Behav 2025:10.1038/s41562-025-02145-1. [PMID: 40335706 DOI: 10.1038/s41562-025-02145-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 02/21/2025] [Indexed: 05/09/2025]
Abstract
Age at onset of walking is an important early childhood milestone which is used clinically and in public health screening. In this genome-wide association study meta-analysis of age at onset of walking (N = 70,560 European-ancestry infants), we identified 11 independent genome-wide significant loci. SNP-based heritability was 24.13% (95% confidence intervals = 21.86-26.40) with ~11,900 variants accounting for about 90% of it, suggesting high polygenicity. One of these loci, in gene RBL2, co-localized with an expression quantitative trait locus (eQTL) in the brain. Age at onset of walking (in months) was negatively genetically correlated with ADHD and body-mass index, and positively genetically correlated with brain gyrification in both infant and adult brains. The polygenic score showed out-of-sample prediction of 3-5.6%, confirmed as largely due to direct effects in sib-pair analyses, and was separately associated with volume of neonatal brain structures involved in motor control. This study offers biological insights into a key behavioural marker of neurodevelopment.
Collapse
Affiliation(s)
- Anna Gui
- Department of Psychology, University of Essex, Wivenhoe Park, Colchester, UK
- Centre for Brain and Cognitive Development, Department of Psychological Sciences, Birkbeck University of London, London, UK
| | - Anja Hollowell
- Centre for Brain and Cognitive Development, Department of Psychological Sciences, Birkbeck University of London, London, UK
| | - Emilie M Wigdor
- Institute of Developmental and Regenerative Medicine, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Morgan J Morgan
- School of Psychology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, UK
| | - Laurie J Hannigan
- Research Department, Lovisenberg Diaconal Hospital, Oslo, Norway
- PsychGen Centre for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Elizabeth C Corfield
- Research Department, Lovisenberg Diaconal Hospital, Oslo, Norway
- PsychGen Centre for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Veronika Odintsova
- Department of Biological Psychology, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Psychiatry, University Medical Center of Groningen, University of Groningen, Groningen, the Netherlands
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Andrew Wong
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, UK
| | - René Pool
- Department of Biological Psychology, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Harriet Cullen
- Research Department of Early Life Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- Department of Medical and Molecular Genetics, School of Basic and Medical Biosciences, King's College London, London, UK
| | - Siân Wilson
- Fetal-Neonatal Neuroimaging and Developmental Science Center, Boston Children's Hospital, Boston, MA, USA
- Division of Newborn Medicine, Harvard Medical School, Boston, MA, USA
| | - Varun Warrier
- Department of Psychiatry and Psychology, University of Cambridge, Cambridge, UK
| | | | - Ole A Andreassen
- Centre for Precision Psychiatry, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo, Oslo, Norway
| | - Christel M Middeldorp
- Department of Child and Youth Psychiatry and Psychology, Amsterdam Reproduction and Development Research Institute, Amsterdam Public Health Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
- Arkin Mental Health Care, Amsterdam, the Netherlands
- Levvel, Academic Center for Child and Adolescent Psychiatry, Amsterdam, the Netherlands
- Child Health Research Centre, University of Queensland, Brisbane, Australia
- Child and Youth Mental Health Service, Children's Health Queensland Hospital and Health Service, Brisbane, Australia
| | - Beate St Pourcain
- Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Meike Bartels
- Department of Biological Psychology, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Dorret I Boomsma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, the Netherlands
| | - Catharina A Hartman
- University Medical Center Psychopathology and Emotion Regulation (ICPE), Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Tomoki Arichi
- Research Department of Early Life Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Anthony D Edwards
- Research Department of Early Life Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Mark H Johnson
- Centre for Brain and Cognitive Development, Department of Psychological Sciences, Birkbeck University of London, London, UK
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - Frank Dudbridge
- Department of Population Health Sciences, University of Leicester, Leicester, UK
| | - Stephan J Sanders
- Institute of Developmental and Regenerative Medicine, Department of Paediatrics, University of Oxford, Oxford, UK
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Alexandra Havdahl
- Research Department, Lovisenberg Diaconal Hospital, Oslo, Norway
- PsychGen Centre for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
- PROMENTA Research Center, University of Oslo, Oslo, Norway
| | - Angelica Ronald
- Centre for Brain and Cognitive Development, Department of Psychological Sciences, Birkbeck University of London, London, UK.
- School of Psychology, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, UK.
| |
Collapse
|
3
|
Khan Y, Davis CN, Jinwala Z, Feuer KL, Toikumo S, Hartwell EE, Sanchez-Roige S, Peterson RE, Hatoum AS, Kranzler HR, Kember RL. Transdiagnostic and Disorder-Level GWAS Enhance Precision of Substance Use and Psychiatric Genetic Risk Profiles in African and European Ancestries. Biol Psychiatry 2025:S0006-3223(25)01180-1. [PMID: 40345609 DOI: 10.1016/j.biopsych.2025.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 02/20/2025] [Accepted: 04/21/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND Substance use disorders (SUDs) and psychiatric disorders frequently co-occur, and their etiology likely reflects both transdiagnostic (i.e., common/shared) and disorder-level (i.e., independent/nonshared) genetic influences. Understanding the genetic influences that are shared and those that operate independently of the shared risk could enhance precision in diagnosis, prevention, and treatment, but this remains underexplored, particularly in non-European ancestry groups. METHODS We applied genomic structural equation modeling to examine the common and independent genetic architecture among SUDs and psychotic, mood, and anxiety disorders using summary statistics from genome-wide association studies (GWAS) conducted in European- (EUR) and African-ancestry (AFR) individuals. To characterize the biological and phenotypic associations, we used FUMA, conducted genetic correlations, and performed phenome-wide association studies (PheWAS). RESULTS In EUR individuals, transdiagnostic genetic factors represented SUDs, psychotic, and mood/anxiety disorders, with GWAS identifying two novel lead single-nucleotide polymorphisms (SNPs) for the mood factor. In AFR individuals, genetic factors represented SUDs and psychiatric disorders, and GWAS identified one novel lead SNP for the SUD factor. In EUR individuals, second-order factor models showed phenotypic and genotypic associations with a broad range of physical and mental health traits. Finally, genetic correlations and PheWAS highlighted how common and independent genetic factors for SUD and psychotic disorders were differentially associated with psychiatric, sociodemographic, and medical phenotypes. CONCLUSIONS Combining transdiagnostic and disorder-level genetic approaches can improve our understanding of co-occurring conditions and increase the specificity of genetic discovery, which is critical for identifying more effective prevention and treatment strategies to reduce the burden of these disorders.
Collapse
Affiliation(s)
- Yousef Khan
- Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Christal N Davis
- Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104; Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104
| | - Zeal Jinwala
- Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Kyra L Feuer
- Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Sylvanus Toikumo
- Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104; Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104
| | - Emily E Hartwell
- Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104; Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104
| | - Sandra Sanchez-Roige
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, United States; Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN 37235, United States; Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - Roseann E Peterson
- Institute for Department of Psychiatry and Behavioral Sciences, Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, NY 11203, United States
| | - Alexander S Hatoum
- Department of Psychological & Brain Sciences, Washington University in St. Louis, St. Louis, MO 63130, United States
| | - Henry R Kranzler
- Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104; Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104
| | - Rachel L Kember
- Department of Psychiatry, University of Pennsylvania School of Medicine, Philadelphia, PA 19104; Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, PA 19104.
| |
Collapse
|
4
|
An Y, Han P, Zhang C, Yue Y, Wen C, Meng Y, Li H, Li X. The role of NUDT3 in lipid accumulation and its functional variants related to backfat thickness in pigs. Int J Biol Macromol 2025; 307:141901. [PMID: 40096926 DOI: 10.1016/j.ijbiomac.2025.141901] [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: 12/28/2024] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/19/2025]
Abstract
NUDT3 is a leading candidate gene that strongly linked to pig fatness traits, however, its function in porcine adipocytes remains poorly understood. Here, the percentage of EdU+ cells was significantly reduced when NUDT3 was knocked down, as was the expression of cell cycle repressors. NUDT3 overexpression yielded the opposite outcome. Moreover, the knockdown of NUDT3 resulted in more lipid droplets in adipocytes, whereas its enforced expression had the reverse effect. In addition, exogenous expression of NUDT3 in adipose tissue significantly reduced fat expansion triggered by a high-fat diet in mice. At molecular level, integrative RIP-seq and RNA-seq analysis revealed that genes influenced by NUDT3 overexpression or knockdown were significantly enriched in the PI3K-AKT signaling pathway, and western blot confirmed that AKT phosphorylation was significantly increased by NUDT3 knockdown, while the phosphorylation levels of PI3K, AKT, and mTOR were significantly decreased by the enforced NUDT3 expression both ex vivo and in vivo. Notably, rs694899689 was identified as a potential genetic variant for modulates NUDT3 expression and impacting backfat thickness in pigs through analysis of multi-omics data, CRISPRi (CRISPR interference) and dual luciferase reporter assays. Overall, our work established NUDT3 as a novel negative regulator of adipogenesis and lipid deposition and revealed that rs694899689 might serve as a potential molecular marker for pig breeding.
Collapse
Affiliation(s)
- Yalong An
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, China
| | - Peiyuan Han
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, China
| | - Chen Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, China
| | - Yongqi Yue
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, China
| | - Chenglong Wen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, China
| | - Yingying Meng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, China
| | - Haoran Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, China
| | - Xiao Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, China; National Key Laboratory of Livestock Biology, Northwest A&F University, Shaanxi 712100, China.
| |
Collapse
|
5
|
Salih AM, Condurache DG, D’Angelo S, Curtis EM, Petersen SE, Altmann A, Harvey NC, Raisi-Estabragh Z. Bone mineral density and cardiovascular diseases: a two-sample Mendelian randomization study. JBMR Plus 2025; 9:ziaf037. [PMID: 40191156 PMCID: PMC11972088 DOI: 10.1093/jbmrpl/ziaf037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 04/09/2025] Open
Abstract
The link between BMD and cardiovascular disease (CVD) remains a topic of extensive debate in observational studies, with inconsistent reports regarding the causality of this relationship. This study implements robust methodologies to evaluate the causal relationship between BMD and various CVDs. Two sample Mendelian randomization (MR) method was used to estimate the relationship between genetically predicted BMD and seven key CVDs: atrial fibrillation and flutter, angina, ischemic heart disease, heart failure, hypertension, myocardial infarction, and non-ischemic cardiomyopathy. Data were obtained from independent publicly available genome-wide association studies (GWAS) for BMD and CVDs, using two separate datasets for the cardiovascular outcomes: the UK Biobank cohort (primary analysis) and the FinnGen cohort (validation analysis). The MR Pleiotropy RESidual Sum and Outlier test assessed the heterogeneity and pleiotropy of selected instrumental variables (IVs). We applied the inverse variance weighted model (IVW), weighted median, weighted mode method, and MR-Egger regression model to estimate causal effects. MR results indicate no relationship between BMD and atrial fibrillation and flutter (IVW, beta-estimate: 0.011, SE: 0.03, p = .73), angina (IVW, beta-estimate: 0.04, SE: 0.03, p = .17), chronic ischemic heart disease (IVW, beta-estimate: 0.009, SE: 0.03, p = .74), heart failure (IVW, beta-estimate: 0.004, SE: 0.04, p = .91), hypertension (IVW, beta-estimate: -0.01, SE: 0.01, p = .44), myocardial infarction (IVW, beta-estimate: 0.02, SE: 0.03, p = .36), or non-ischemic cardiomyopathy (IVW, beta-estimate: 0.1, SE: 0.08, p = .20). These findings remained consistent across all complementary analyses (MR-Egger, weighted median and weighted mode) and were validated using the FinnGen cohort GWAS dataset. This comprehensive analysis identified no evidence for a causal link between genetically predicted BMD and a range of key CVDs. Previously reported observational associations between bone and cardiovascular health likely represent shared risk factors rather than direct causal mechanisms.
Collapse
Affiliation(s)
- Ahmed M Salih
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London EC1M 6BQ, United Kingdom
- Department of Population Health Sciences, University of Leicester, Leicester LE1 7RH, United Kingdom
- PRIME Lab, Scientific Research Center, University of Zakho, Zakho, Kurdistan Region, Iraq
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, London EC1A 7BE, United Kingdom
| | - Dorina-Gabriela Condurache
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London EC1M 6BQ, United Kingdom
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, London EC1A 7BE, United Kingdom
| | - Stefania D’Angelo
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton SO16 6YD, United Kingdom
| | - Elizabeth M Curtis
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton SO16 6YD, United Kingdom
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, United Kingdom
| | - Steffen E Petersen
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London EC1M 6BQ, United Kingdom
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, London EC1A 7BE, United Kingdom
| | - Andre Altmann
- Department of Medical Physics and Biomedical Engineering, The UCL Hawkes Institute, University College London, London WC1E 6BT, United Kingdom
| | - Nicholas C Harvey
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton SO16 6YD, United Kingdom
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, United Kingdom
| | - Zahra Raisi-Estabragh
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, London EC1M 6BQ, United Kingdom
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, London EC1A 7BE, United Kingdom
| |
Collapse
|
6
|
Levin MG, Koyama S, Woerner J, Zhang DY, Rodriguez A, Nandi T, Truong B, Abramowitz SA, Gupta H, Kamineni H, Hornsby W, Li Z, Cohron T, Huffman JE, Ellinor P, Kim D, Liao KP, Madduri RK, Voight BF, Verma A, Damrauer SM, Natarajan P. Genome-Wide Assessment of Pleiotropy Across >1000 Traits from Global Biobanks. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.18.25326074. [PMID: 40313291 PMCID: PMC12045404 DOI: 10.1101/2025.04.18.25326074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Large-scale genetic association studies have identified thousands of trait-associated risk loci, establishing the polygenic basis for common complex traits and diseases. Although prior studies suggest that many trait-associated loci are pleiotropic, the extent to which this pleiotropy reflects shared causal variants or confounding by linkage disequilibrium remains poorly characterized. To define a set of candidate loci with potentially pleiotropic associations, we performed genome-wide association study (GWAS) meta-analyses of up to 1,167 clinically relevant traits and diseases across 1,789,365 diverse individuals genetically similar to Admixed American (AMR, NMax = 60,756), African (AFR, NMax = 128,361), East Asian (EAS, NMax = 307,465), European (EUR, NMax = 1,283,907), and South Asian (SAS, NMax = 8,876) reference populations from the VA Million Veteran Program (MVP), UK Biobank (UKB), FinnGen, Biobank Japan (BBJ), Tohoku Medical Megabank (ToMMO), and Korean Genome and Epidemiology Study (KoGES). We identified 27,193 genome-wide significant locus-trait pairs (1MB region with PGWAMA < 5 × 10-8) in within-population analysis and 29,139 in multi-population analysis (PMR-MEGA < 5 × 10-8). Among these, 11.5% (n = 3,149) of locus-trait pairs in population-wise and 6.4% (n = 1,875) in multi-population analyses did not reach genome-wide significance in previously published GWAS. In aggregate, the genome-wide significant loci fell within 2,624 non-overlapping autosomal genomic windows on average ~600kb in size. Each locus contained genome-wide significant signals for a median of 6 traits (IQR 2 to 18), including 2,110 (80%) pleiotropic loci associated with >1 trait. Multi-trait colocalization identified 1,902 (72%) loci with high-confidence (posterior probability > 0.9) evidence of a shared causal variant across two or more traits. Variants in pleiotropic loci were significantly enriched for a broad spectrum of functional annotations compared to non-pleiotropic counterparts. Polygenic scores (PGS) developed from these data generally improved prediction compared to existing PGS and were broadly associated with both on- and off-target phenotypes. These results provide a contemporary map of genetic pleiotropy across the spectrum of human traits/diseases and genetic backgrounds.
Collapse
Affiliation(s)
- Michael G Levin
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Medicine, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
| | - Satoshi Koyama
- Center for Genomic Medicine and Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Jakob Woerner
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - David Y Zhang
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Alexis Rodriguez
- Data Science and Learning, Argonne National Laboratory, Lemont, IL, 60439, USA
| | - Tarak Nandi
- Data Science and Learning, Argonne National Laboratory, Lemont, IL, 60439, USA
| | - Buu Truong
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Department of Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Sarah A Abramowitz
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Hritvik Gupta
- Division of Translational Medicine and Human Genetics, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Himani Kamineni
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Whitney Hornsby
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Zilinghan Li
- Data Science and Learning, Argonne National Laboratory, Lemont, IL, 60439, USA
| | | | - Jennifer E Huffman
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, 02130, USA
- Palo Alto Veterans Institute for Research (PAVIR), Palo Alto Health Care System, Palo Alto, CA, 94304, USA
- Department of Medicine, Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), Harvard Medical School, Boston, MA, 02115, USA
| | - Patrick Ellinor
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Dokyoon Kim
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Katherine P Liao
- Section of Rheumatology, Department of Medicine, VA Boston Healthcare System, Boston, MA, 02130, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, 02115, USA
| | - Ravi K Madduri
- Data Science and Learning, Argonne National Laboratory, Lemont, IL, 60439, USA
- Consortium for Advanced Science and Engineering, University of Chicago, Chicago, IL, 60616, USA
| | - Benjamin F Voight
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Corporal Michael J. Crescenz VA Medical Center, 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
| | - Anurag Verma
- Department of Medicine, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
- Division of Translational Medicine and Human Genetics, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Scott M Damrauer
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Surgery, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, 19104, USA
| | - Pradeep Natarajan
- Center for Genomic Medicine and Cardiovascular Research Center, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| |
Collapse
|
7
|
Nikolitsa EK, Kontou PI, Bagos PG. metacp: a versatile software package for combining dependent or independent p-values. BMC Bioinformatics 2025; 26:109. [PMID: 40253343 PMCID: PMC12008841 DOI: 10.1186/s12859-025-06126-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: 01/04/2025] [Accepted: 04/01/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND We present metacp an open-source software package which implements an abundance of statistical methods for the combination of both independent p-values, with methods such as Fisher's, Stouffer's and Edgington's, and dependent p-values, with methods such as Brown's method and the Cauchy Combination Test. RESULTS The tool is available in Python and STATA, it is very fast, and it is easy to use, requiring only minimal input. It offers a useful resource for combining both independent and dependent p-values, responding to diverse analytical needs for practitioners performing meta-analyses and bioinformaticians developing tools for a variety of applications. Depending on the input data it can be used for gene-based testing, for analysis of multiple traits in GWAS, or for combining diverse multi-omics data such as those of a TWAS, a colocalization or an RNA-seq study. CONCLUSIONS Compared to other similar packages (like poolr or metap), metacp implements the largest collection of statistical methods for this problem, offering users the flexibility to choose from a wide variety of approaches. Being available both as a standalone Python tool and as a STATA command, metacp is accessible to a broad and diverse audience, including practitioners conducting meta-analyses across various fields and bioinformaticians developing new tools where p-value combination is a crucial component.
Collapse
Affiliation(s)
- Evgenia K Nikolitsa
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 35100, Lamia, Greece
| | | | - Pantelis G Bagos
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 35100, Lamia, Greece.
| |
Collapse
|
8
|
Manios GA, Michailidi A, Kontou PI, Bagos PG. PRED-LD: efficient imputation of GWAS summary statistics. BMC Bioinformatics 2025; 26:107. [PMID: 40240925 PMCID: PMC12004831 DOI: 10.1186/s12859-025-06119-y] [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: 10/30/2024] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Genome-wide association studies have identified connections between genetic variations and diseases, but they only examine a small portion of single nucleotide polymorphisms. To enhance genetic findings, researchers suggest imputing genotypes for unmeasured SNPs to improve coverage and statistical power. When this is not possible, summary statistics imputation can be used as an alternative. The available summary statistics imputation tools rely on reference panels, such as the 1000 Genomes Project, to estimate linkage disequilibrium (LD) between variants for accurate imputation. Tools like FAPI and SSIMP use these reference panels in variant call format (VCF) for this purpose, though this process can be time-consuming. A more effective approach for processing reference panels in summary statistics imputation was proposed in RAISS. In this approach, the LD among the variants is precomputed from the reference panel, prior to imputation, thereby reducing computational time. RESULTS We present PRED-LD, an imputation method for GWAS summary statistics that aims to enhance the resolution of genetic association analyses. The proposed method uses precomputed linkage disequilibrium statistics from HapMap, Pheno Scanner and TOP-LD to impute summary statistics, given beta coefficients and standard errors. The single-point approach that we describe provides a fast and accurate way to estimate associations for untyped single nucleotide polymorphisms that exhibit high linkage disequilibrium (LD). The proposed method is faster, provides accurate imputation compared to existing tools, and has been implemented in both a web service ( https://compgen.dib.uth.gr/PRED-LD/ ) and a command-line tool ( https://github.com/pbagos/PRED-LD ), making it a useful resource for the research community. CONCLUSIONS PRED-LD offers an efficient and accurate method for GWAS summary statistics imputation, providing faster performance, direct result interpretation, and the ability to use multiple reference panels. Also, the online version of PRED-LD simplifies obtaining LD information and performing imputation tasks without downloading reference panels and will be continuously updated to support tools for meta-analysis and fine-mapping in GWAS.
Collapse
Affiliation(s)
- Georgios A Manios
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 35131, Lamia, Greece
| | - Aikaterini Michailidi
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 35131, Lamia, Greece
| | | | - Pantelis G Bagos
- Department of Computer Science and Biomedical Informatics, University of Thessaly, 35131, Lamia, Greece.
| |
Collapse
|
9
|
Lai WY, Hsu SK, Futschik A, Schlötterer C. Pleiotropy increases parallel selection signatures during adaptation from standing genetic variation. eLife 2025; 13:RP102321. [PMID: 40227842 PMCID: PMC11996171 DOI: 10.7554/elife.102321] [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] [Indexed: 04/15/2025] Open
Abstract
The phenomenon of parallel evolution, whereby similar genomic and phenotypic changes occur across replicated pairs of populations or species, is widely studied. Nevertheless, the determining factors of parallel evolution remain poorly understood. Theoretical studies have proposed that pleiotropy, the influence of a single gene on multiple traits, is an important factor. In order to gain a deeper insight into the role of pleiotropy for parallel evolution from standing genetic variation, we characterized the interplay between parallelism, polymorphism, and pleiotropy. The present study examined the parallel gene expression evolution in 10 replicated populations of Drosophila simulans, which were adapted from standing variation to the same new temperature regime. The data demonstrate that the parallel evolution of gene expression from standing genetic variation is positively correlated with the strength of pleiotropic effects. The ancestral variation in gene expression is, however, negatively correlated with parallelism. Given that pleiotropy is also negatively correlated with gene expression variation, we conducted a causal analysis to distinguish cause and correlation and evaluate the role of pleiotropy. The causal analysis indicated that both direct (causative) and indirect (correlational) effects of pleiotropy contribute to parallel evolution. The indirect effect is mediated by historic selective constraint in response to pleiotropy. This results in parallel selection responses due to the reduced standing variation of pleiotropic genes. The direct effect of pleiotropy is likely to reflect a genetic correlation among adaptive traits, which in turn gives rise to synergistic effects and higher parallelism.
Collapse
Affiliation(s)
- Wei-Yun Lai
- Institut für Populationsgenetik, Vetmeduni ViennaViennaAustria
- Vienna Graduate School of Population Genetics, Vetmeduni ViennaViennaAustria
| | - Sheng-Kai Hsu
- Institut für Populationsgenetik, Vetmeduni ViennaViennaAustria
- Vienna Graduate School of Population Genetics, Vetmeduni ViennaViennaAustria
| | - Andreas Futschik
- Department of Applied Statistics, Johannes Kepler University LinzLinzAustria
| | | |
Collapse
|
10
|
Fanelli G, Franke B, Fabbri C, Werme J, Erdogan I, De Witte W, Poelmans G, Ruisch IH, Reus LM, van Gils V, Jansen WJ, Vos SJB, Alam KA, Martinez A, Haavik J, Wimberley T, Dalsgaard S, Fóthi Á, Barta C, Fernandez-Aranda F, Jimenez-Murcia S, Berkel S, Matura S, Salas-Salvadó J, Arenella M, Serretti A, Mota NR, Bralten J. Local patterns of genetic sharing between neuropsychiatric and insulin resistance-related conditions. Transl Psychiatry 2025; 15:145. [PMID: 40221434 PMCID: PMC11993748 DOI: 10.1038/s41398-025-03349-9] [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: 07/25/2024] [Revised: 03/14/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
The co-occurrence of insulin resistance (IR)-related metabolic conditions with neuropsychiatric disorders is a major public health challenge. Evidence of the genetic links between these phenotypes is emerging, but little is currently known about the genomic regions and biological functions that are involved. To address this, we performed Local Analysis of [co]Variant Association (LAVA) using large-scale (N = 9,725-933,970) genome-wide association studies (GWASs) results for three IR-related conditions (type 2 diabetes mellitus, obesity, and metabolic syndrome) and nine neuropsychiatric disorders. Subsequently, positional and expression quantitative trait locus (eQTL)-based gene mapping and downstream functional genomic analyses were performed on the significant loci. Patterns of negative and positive local genetic correlations (|rg| = 0.21-1, pFDR < 0.05) were identified at 109 unique genomic regions across all phenotype pairs. Local correlations emerged even in the absence of global genetic correlations between IR-related conditions and Alzheimer's disease, bipolar disorder, and Tourette's syndrome. Genes mapped to the correlated regions showed enrichment in biological pathways integral to immune-inflammatory function, vesicle trafficking, insulin signalling, oxygen transport, and lipid metabolism. Colocalisation analyses further prioritised 10 genetically correlated regions for likely harbouring shared causal variants, displaying high deleterious or regulatory potential. These variants were found within or in close proximity to genes, such as SLC39A8 and HLA-DRB1, that can be targeted by supplements and already known drugs, including omega-3/6 fatty acids, immunomodulatory, antihypertensive, and cholesterol-lowering drugs. Overall, our findings highlight the complex genetic architecture of IR-neuropsychiatric multimorbidity, advocating for an integrated disease model and offering novel insights for research and treatment strategies in this domain.
Collapse
Affiliation(s)
- Giuseppe Fanelli
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Barbara Franke
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Chiara Fabbri
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Josefin Werme
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Izel Erdogan
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ward De Witte
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Medical Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Geert Poelmans
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Medical Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - I Hyun Ruisch
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Medical Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lianne Maria Reus
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
- Center for Neurobehavioral Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Veerle van Gils
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
| | - Willemijn J Jansen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
| | - Stephanie J B Vos
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
| | | | - Aurora Martinez
- Department of Biomedicine, University of Bergen, Bergen, Norway
- K.G. Jebsen Center for Translational Research in Parkinson's Disease, University of Bergen, Bergen, Norway
| | - Jan Haavik
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Theresa Wimberley
- National Centre for Register-based Research, School of Business and Social Sciences, Aarhus University, Aarhus, Denmark
- iPSYCH - The Lundbeck Foundation Initiative for Integrated Psychiatric Research, Aarhus, Denmark
| | - Søren Dalsgaard
- National Centre for Register-based Research, School of Business and Social Sciences, Aarhus University, Aarhus, Denmark
- Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Child and Adolescent Psychiatry Glostrup, Mental Health Services of the Capital Region, Hellerup, Denmark
| | - Ábel Fóthi
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Csaba Barta
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Fernando Fernandez-Aranda
- Clinical Psychology Department, University Hospital of Bellvitge, Barcelona, Spain
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Barcelona, Spain
- Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Susana Jimenez-Murcia
- Clinical Psychology Department, University Hospital of Bellvitge, Barcelona, Spain
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Barcelona, Spain
- Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Simone Berkel
- Institute of Human Genetics, Heidelberg University, Heidelberg, Germany
| | - Silke Matura
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Jordi Salas-Salvadó
- Universitat Rovira i Virgili, Biochemistry and biotechnology Department, Grup Alimentació, Nutrició, Desenvolupament i Salut Mental, Unitat de Nutrició Humana, Reus, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Martina Arenella
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Forensic and Neurodevelopmental Science, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Alessandro Serretti
- Department of Medicine and Surgery, Kore University of Enna, Enna, Italy
- Oasi Research Institute-IRCCS, Troina, Italy
| | - Nina Roth Mota
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
- Department of Medical Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Janita Bralten
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands.
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
11
|
Moen GH, Hwang LD, Brito Nunes C, Warrington NM, Evans DM. The genetics of low and high birthweight and their relationship with cardiometabolic disease. Diabetologia 2025:10.1007/s00125-025-06420-8. [PMID: 40210729 DOI: 10.1007/s00125-025-06420-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/11/2025] [Indexed: 04/12/2025]
Abstract
AIMS/HYPOTHESIS Low birthweight infants are at increased risk not only of mortality, but also of type 2 diabetes mellitus and CVD in later life. At the opposite end of the spectrum, high birthweight infants have increased risk of birth complications, such as shoulder dystocia, neonatal hypoglycaemia and obesity, and similarly increased risk of type 2 diabetes mellitus and CVD. However, previous genome-wide association studies (GWAS) of birthweight in the UK Biobank have primarily focused on individuals within the 'normal' range and have excluded individuals with high and low birthweight (<2.5 kg or >4.5 kg). The aim of this study was to investigate genetic variation associated within the tail ends of the birthweight distribution, to: (1) see whether the genetic factors operating in these regions were different from those that explained variation in birthweight within the normal range; (2) explore the genetic correlation between extremes of birthweight and cardiometabolic disease; and (3) investigate whether analysing the full distribution of birthweight values, including the extremes, improved the ability to detect genuine loci in GWAS. METHODS We performed case-control GWAS analysis of low (<2.5 kg) and high (>4.5 kg) birthweight in the UK Biobank using REGENIE software (Nlow=20,947; Nhigh=12,715; Ncontrols=207,506) and conducted three continuous GWAS of birthweight, one including the full range of birthweights, one involving a truncated GWAS including only individuals with birthweights between 2.5 and 4.5 kg and a third GWAS that winsorised birthweight values <2.5 kg and >4.5 kg. Additionally, we performed bivariate linkage disequilibrium (LD) score regression to estimate the genetic correlation between low/normal/high birthweight and cardiometabolic traits. RESULTS Bivariate LD score regression analyses suggested that high birthweight had a mostly similar genetic aetiology to birthweight within the normal range (genetic correlation coefficient [rG]=0.91, 95% CI 0.83, 0.99), whereas there was more evidence for a separate set of genes underlying low birthweight (rG=-0.74, 95% CI 0.66, 0.82). Low birthweight was also significantly positively genetically correlated with most cardiometabolic traits and diseases we examined, whereas high birthweight was mostly positively genetically correlated with adiposity and anthropometric-related traits. The winsorisation strategy performed best in terms of locus detection, with the number of independent genome-wide significant associations (p<5×10-8) increasing from 120 genetic variants at 94 loci in the truncated GWAS to 270 genetic variants at 178 loci, including 27 variants at 25 loci that had not been identified in previous birthweight GWAS. This included a novel low-frequency missense variant in the ABCC8 gene, a gene known to be involved in congenital hyperinsulinism, neonatal diabetes mellitus and MODY, that was estimated to be responsible for a 170 g increase in birthweight amongst carriers. CONCLUSIONS/INTERPRETATION Our results underscore the importance of genetic factors in the genesis of the phenotypic correlation between birthweight and cardiometabolic traits and diseases.
Collapse
Affiliation(s)
- Gunn-Helen Moen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
- The Frazer Institute, The University of Queensland, Woolloongabba, QLD, Australia.
| | - Liang-Dar Hwang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Caroline Brito Nunes
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Nicole M Warrington
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
- The Frazer Institute, The University of Queensland, Woolloongabba, QLD, Australia
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - David M Evans
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.
- The Frazer Institute, The University of Queensland, Woolloongabba, QLD, Australia.
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.
| |
Collapse
|
12
|
Dalapati T, Wang L, Jones AG, Cardwell J, Konigsberg IR, Bossé Y, Sin DD, Timens W, Hao K, Yang I, Ko DC. Context-specific eQTLs provide deeper insight into causal genes underlying shared genetic architecture of COVID-19 and idiopathic pulmonary fibrosis. HGG ADVANCES 2025; 6:100410. [PMID: 39876559 PMCID: PMC11872446 DOI: 10.1016/j.xhgg.2025.100410] [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: 07/15/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 01/30/2025] Open
Abstract
Most genetic variants identified through genome-wide association studies (GWASs) are suspected to be regulatory in nature, but only a small fraction colocalize with expression quantitative trait loci (eQTLs, variants associated with expression of a gene). Therefore, it is hypothesized but largely untested that integration of disease GWAS with context-specific eQTLs will reveal the underlying genes driving disease associations. We used colocalization and transcriptomic analyses to identify shared genetic variants and likely causal genes associated with critically ill COVID-19 and idiopathic pulmonary fibrosis. We first identified five genome-wide significant variants associated with both diseases. Four of the variants did not demonstrate clear colocalization between GWAS and healthy lung eQTL signals. Instead, two of the four variants colocalized only in cell type- and disease-specific eQTL datasets. These analyses pointed to higher ATP11A expression from the C allele of rs12585036, in monocytes and in lung tissue from primarily smokers, which increased risk of idiopathic pulmonary fibrosis (IPF) and decreased risk of critically ill COVID-19. We also found lower DPP9 expression (and higher methylation at a specific CpG) from the G allele of rs12610495, acting in fibroblasts and in IPF lungs, and increased risk of IPF and critically ill COVID-19. We further found differential expression of the identified causal genes in diseased lungs when compared to non-diseased lungs, specifically in epithelial and immune cell types. These findings highlight the power of integrating GWASs, context-specific eQTLs, and transcriptomics of diseased tissue to harness human genetic variation to identify causal genes and where they function during multiple diseases.
Collapse
Affiliation(s)
- Trisha Dalapati
- Medical Scientist Training Program, Duke University School of Medicine, Durham, NC, USA; Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Angela G Jones
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA; University Program in Genetics and Genomics, Duke University, Durham, NC, USA
| | - Jonathan Cardwell
- Department of Biomedical Informatics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Iain R Konigsberg
- Department of Biomedical Informatics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Yohan Bossé
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Department of Molecular Medicine, Québec City, QC, Canada
| | - Don D Sin
- Center for Heart Lung Innovation, University of British Columbia and St. Paul's Hospital, Vancouver, BC, Canada
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ivana Yang
- Department of Biomedical Informatics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Dennis C Ko
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA; University Program in Genetics and Genomics, Duke University, Durham, NC, USA; Division of Infectious Diseases, Department of Medicine, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
13
|
Perez Palomeque G, Khacha-ananda S, Monum T, Wunnapuk K. Prediction of Skin Color Using Forensic DNA Phenotyping in Asian Populations: A Focus on Thailand. Biomolecules 2025; 15:548. [PMID: 40305359 PMCID: PMC12024907 DOI: 10.3390/biom15040548] [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: 03/03/2025] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
Forensic DNA phenotyping (FDP) has emerged as an essential tool in criminal investigations, enabling the prediction of physical traits based on genetic information. This review explores the genetic factors influencing skin pigmentation, particularly within Asian populations, with a focus on Thailand. Key genes such as Oculocutaneous Albinism II (OCA2), Dopachrome Tautomerase (DCT), KIT Ligand (KITLG), and Solute Carrier Family 24 Member 2 (SLC24A2) are examined for their roles in melanin production and variations that lead to different skin tones. The OCA2 gene is highlighted for its role in transporting ions that help stabilize melanosomes, while specific variants in the DCT gene, including single nucleotide polymorphisms (SNPs) rs2031526 and rs3782974, are discussed for their potential effects on pigmentation in Asian groups. The KITLG gene, crucial for developing melanocytes, includes the SNP rs642742, which is linked to lighter skin in East Asians. Additionally, recent findings on the SLC24A2 gene are presented, emphasizing its connection to pigmentation through calcium regulation in melanin production. Finally, the review addresses the ethical considerations of using FDP in Thailand, where advances in genetic profiling raise concerns about privacy, consent, and discrimination. Establishing clear guidelines is vital to balancing the benefits of forensic DNA applications with the protection of individual rights.
Collapse
Affiliation(s)
- Gabriel Perez Palomeque
- PhD Program in Medical Sciences, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Supakit Khacha-ananda
- Department of Forensic Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (T.M.); (K.W.)
| | - Tawachai Monum
- Department of Forensic Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (T.M.); (K.W.)
| | - Klintean Wunnapuk
- Department of Forensic Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (T.M.); (K.W.)
| |
Collapse
|
14
|
Zhou S, Zi J, Hu Y, Wang X, Cheng G, Xiong J. Genetic correlation, pleiotropic loci and shared risk genes between major depressive disorder and gastrointestinal tract disorders. J Affect Disord 2025; 374:84-90. [PMID: 39800072 DOI: 10.1016/j.jad.2025.01.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 01/07/2025] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Major depressive disorder (MDD) is associated with gastrointestinal tract (GIT) disorders, while genetic correlation, pleiotropic loci and shared risk genes remain to be explored. METHODS Leveraging genome-wide association study statistics for MDD (n = 170,756), peptic ulcer disease (PUD; n = 16,666), gastroesophageal reflux disease (GORD; n = 54,854), PUD and/or GORD and/or medications (PGM; n = 90,175), irritable bowel syndrome (IBS; n = 28,518), and inflammatory bowel disease (IBD; n = 7045), we determined global and local genetic correlations, identified pleiotropic loci, performed gene-level evaluations, and inferred causal associations using bidirectional Mendelian randomization. RESULTS We found global correlation of MDD with PUD (rg = 0.444, P = 3.135 × 10-24), GORD (rg = 0.459, P = 2.568 × 10-65), PGM (rg = 0.498, P = 6.094 × 10-114), IBS (rg = 0.621, P = 2.483 × 10-63), and IBD (rg = 0.171, P = 1.824 × 10-5). We identified 12 locally correlated regions between MDD and GIT disorders except for IBD, and one shared region (chr11:111985737-113,103,996) for PGM, GORD, and IBS. We found one pleiotropic locus for PUD, 12 for GORD, 30 for PGM, eight for IBS, and seven for IBD, and five shared loci (rs138786869, rs2284189, rs3130063, rs35789010, rs7568369) for GORD and PGM. We respectively observed 14 and 20 overlapping genes for MDD-GORD and MDD-PGM. We showed genetic liabilities to GORD, PGM, and IBS causally increase MDD risk, while all reverse causalities are significant. CONCLUSIONS Our work identifies genetic architectures shared between MDD and GIT disorders, contributes genetic insights to understand depression in the context of gut-brain interactions, and provides potential targets to treat gastrointestinal symptoms in depressive patients.
Collapse
Affiliation(s)
- Siquan Zhou
- Healthy Food Evaluation Research Center, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Jing Zi
- Healthy Food Evaluation Research Center, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yifan Hu
- Healthy Food Evaluation Research Center, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xiaoyu Wang
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Maternal & Child Nutrition Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Guo Cheng
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Maternal & Child Nutrition Center, West China Second University Hospital, Sichuan University, Chengdu, China.
| | - Jingyuan Xiong
- Healthy Food Evaluation Research Center, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu 610041, China.
| |
Collapse
|
15
|
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
|
16
|
Lee PH, Jung JY, Sanzo BT, Duan R, Ge T, Waldman I, Smoller JW, Schwaba T, Tucker-Drob EM, Grotzinger AD. Transdiagnostic Polygenic Risk Models for Psychopathology and Comorbidity: Cross-Ancestry Analysis in the All of Us Research Program. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.26.25324720. [PMID: 40196240 PMCID: PMC11974969 DOI: 10.1101/2025.03.26.25324720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Psychiatric disorders exhibit substantial genetic overlap, raising questions about the utility of transdiagnostic genetic risk models. Using data from the All of Us Research Program (N=102,091), we evaluated common psychiatric genetic (CPG) factor-based polygenic risk scores (PRSs) compared to standard disorder-specific PRSs. The CPG PRS consistently outperformed disorder-specific scores in predicting individual disorder risk, explaining 1.07 to 24.6 times more phenotypic variance across 11 psychiatric conditions. Meanwhile, many disorder-specific PRSs retained independent but smaller contributions, highlighting the complementary nature of shared and disorder-specific genetic risk. While alternative multi-factor models improved model fit, the CPG PRS provided comparable or superior predictive performance across most disorders, including overall comorbidity burden. Cross-ancestry analyses however revealed notable limitations of European-centric GWAS datasets for other populations due to ancestral differences in genetic architecture. These findings underscore the potential value of transdiagnostic PRSs for psychiatric genetics while highlighting the need for more equitable genetic risk models.
Collapse
Affiliation(s)
- Phil H. Lee
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Mass General Brigham, Boston, MA, USA
- Department of Psychiatry, Mass General Brigham and Harvard Medical School, Boston, MA, USA
- Stanly Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jae-Yoon Jung
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Brandon T. Sanzo
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Mass General Brigham, Boston, MA, USA
| | - Rui Duan
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Tian Ge
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Mass General Brigham, Boston, MA, USA
- Department of Psychiatry, Mass General Brigham and Harvard Medical School, Boston, MA, USA
- Stanly Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Irwin Waldman
- Department of Psychology, Emory University, Atlanta, GA, USA
| | - Jordan W. Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Mass General Brigham, Boston, MA, USA
- Department of Psychiatry, Mass General Brigham and Harvard Medical School, Boston, MA, USA
- Stanly Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ted Schwaba
- Department of Psychology, Michigan State University, MI, USA
| | | | - Andrew D. Grotzinger
- Institute for Behavioral Genetics, University of Colorado at Boulder, CO, USA
- Department of Psychology and Neuroscience, University of Colorado at Boulder, CO, USA
| |
Collapse
|
17
|
Wallis NJ, McClellan A, Mörseburg A, Kentistou KA, Jamaluddin A, Dowsett GKC, Schofield E, Morros-Nuevo A, Saeed S, Lam BYH, Sumanasekera NT, Chan J, Kumar SS, Zhang RM, Wainwright JF, Dittmann M, Lakatos G, Rainbow K, Withers D, Bounds R, Ma M, German AJ, Ladlow J, Sargan D, Froguel P, Farooqi IS, Ong KK, Yeo GSH, Tadross JA, Perry JRB, Gorvin CM, Raffan E. Canine genome-wide association study identifies DENND1B as an obesity gene in dogs and humans. Science 2025; 387:eads2145. [PMID: 40048553 DOI: 10.1126/science.ads2145] [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: 08/23/2024] [Accepted: 01/10/2025] [Indexed: 03/29/2025]
Abstract
Obesity is a heritable disease, but its genetic basis is incompletely understood. Canine population history facilitates trait mapping. We performed a canine genome-wide association study for body condition score-a measure of obesity-in 241 Labrador retrievers. Using a cross-species approach, we showed that canine obesity genes are also associated with rare and common forms of obesity in humans. The lead canine association was within the gene DENN domain containing 1B (DENND1B). Each copy of the alternate allele was associated with ~7.5% greater body fat. We demonstrate a role for this gene in regulating signaling and trafficking of melanocortin 4 receptor, a critical controller of energy homeostasis. Thus, canine genetics identified obesity genes and mechanisms relevant to both dogs and humans.
Collapse
Affiliation(s)
- Natalie J Wallis
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Alyce McClellan
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Alexander Mörseburg
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Katherine A Kentistou
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Aqfan Jamaluddin
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
| | - Georgina K C Dowsett
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Ellen Schofield
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Anna Morros-Nuevo
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Sadia Saeed
- INSERM UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille, France
- University of Lille, Lille University Hospital, Lille, France
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Brian Y H Lam
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Natasha T Sumanasekera
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Justine Chan
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Sambhavi S Kumar
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Rey M Zhang
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Jodie F Wainwright
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Marie Dittmann
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Gabriella Lakatos
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Kara Rainbow
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - David Withers
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Rebecca Bounds
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre, Cambridge, UK
| | - Marcella Ma
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Alexander J German
- Institute of Life Course and Medical Sciences and School of Veterinary Science, University of Liverpool, Neston, UK
| | - Jane Ladlow
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - David Sargan
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Philippe Froguel
- INSERM UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille, France
- University of Lille, Lille University Hospital, Lille, France
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - I Sadaf Farooqi
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre, Cambridge, UK
| | - Ken K Ong
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Giles S H Yeo
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - John A Tadross
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Histopathology and Cambridge Genomics Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - John R B Perry
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Caroline M Gorvin
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
| | - Eleanor Raffan
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| |
Collapse
|
18
|
Abbasova L, Urbanaviciute P, Hu D, Ismail JN, Schilder BM, Nott A, Skene NG, Marzi SJ. CUT&Tag recovers up to half of ENCODE ChIP-seq histone acetylation peaks. Nat Commun 2025; 16:2993. [PMID: 40148272 PMCID: PMC11950320 DOI: 10.1038/s41467-025-58137-2] [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: 07/28/2022] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
DNA-protein interactions have traditionally been profiled via chromatin immunoprecipitation followed by next-generation sequencing (ChIP-seq). Cleavage Under Targets & Tagmentation (CUT&Tag) is a rapidly expanding technique that enables the profiling of such interactions in situ at high sensitivity. However, thorough evaluation and benchmarking against established ChIP-seq datasets are lacking. Here, we comprehensively benchmarked CUT&Tag for H3K27ac and H3K27me3 against published ChIP-seq profiles from ENCODE in K562 cells. Combining multiple new and published CUT&Tag datasets, there was an average recall of 54% known ENCODE peaks for both histone modifications. We tested peak callers MACS2 and SEACR and identified optimal peak calling parameters. Overall, peaks identified by CUT&Tag represent the strongest ENCODE peaks and show the same functional and biological enrichments as ChIP-seq peaks identified by ENCODE. Our workflow systematically evaluates the merits of methodological adjustments, providing a benchmarking framework for the experimental design and analysis of CUT&Tag studies.
Collapse
Affiliation(s)
- Leyla Abbasova
- UK Dementia Research Institute at Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Paulina Urbanaviciute
- UK Dementia Research Institute at King's College London, London, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Di Hu
- UK Dementia Research Institute at Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Joy N Ismail
- UK Dementia Research Institute at King's College London, London, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Brian M Schilder
- UK Dementia Research Institute at Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Alexi Nott
- UK Dementia Research Institute at Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Nathan G Skene
- UK Dementia Research Institute at Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Sarah J Marzi
- Department of Brain Sciences, Imperial College London, London, UK.
- UK Dementia Research Institute at King's College London, London, UK.
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| |
Collapse
|
19
|
Zhi D, Sanzo BT, Jung DH, Cabana-Domínguez J, Fernàndez-Castillo N, Cormand B, Sui J, Jiang R, Evins EA, Hadland SE, Roffman JL, Liu RT, Gilman J, Lee PH. Unravelling Polygenic Risk and Environmental Interactions in Adolescent Polysubstance Use: a U.S. Population-Based Observational Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.21.25324407. [PMID: 40196248 PMCID: PMC11974769 DOI: 10.1101/2025.03.21.25324407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Background Polysubstance use (PSU), defined as the use of multiple psychoactive substances, is associated with a heightened risk of subsequent health issues, including substance use disorders. However, the interplay between genetic susceptibility and environmental exposures in PSU initiation during adolescence remains understudied. Methods We examined associations of polygenic scores (PGSs) for general addiction risk, environmental factors, and their joint interactions with PSU initiation among 11,868 adolescents (aged 11-15 years) from the Adolescent Brain and Cognitive Development study. PSU status was assessed through interviews and toxicology screenings. Results Our sample included 7,898 adolescents (mean age 12.9 [0.6] years; 4,150 [53%] male). Of these, 541 (6.8%) had initiated single substance use (SSU), and 162 (2.1%) reported PSU). PGSs for general addiction risk were significantly associated with PSU (Odds Ratios [OR]=1.62, 95% CI=1.30-2.01) but not with SSU. Key environmental risk factors for PSU included prenatal substance use and peer victimization, whereas protective factors included planned pregnancy and positive family dynamics. Notably, gene-environment interaction analyses revealed that peer victimization (OR=2.4, 95% CI=1.4-4.2), prenatal substance use (OR=2.1, 95% CI=1.2-3.6), and substance availability (OR=2.3, 95% CI=1.3-3.9) substantially increased PSU risk among adolescents with high genetic susceptibility, while having minimal influence at low genetic risk levels (all p < 0.05 after multiple testing correction). Conclusions This study provides novel evidence linking polygenic risk to PSU in early adolescence and highlights PSU as a more severe manifestation of substance use liability driven by heightened genetic vulnerability and adverse environmental exposures.
Collapse
Affiliation(s)
- Dongmei Zhi
- Center for Addiction Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Psychiatry, Mass General Brigham and Harvard Medical School, Boston, MA 02115, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Brandon T. Sanzo
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Daniel H. Jung
- Department of Psychology and Behavioral Neuroscience, Cornell University, Ithaca, NY 14853, USA
| | - Judit Cabana-Domínguez
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Catalunya, 08028, Spain
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona 08035, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona 08035, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Noelia Fernàndez-Castillo
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Catalunya, 08028, Spain
- Centro de Investigación Biomédica en Red de Enfermedades raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona, Barcelona, Catalonia, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Catalonia, Spain
| | - Bru Cormand
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Catalunya, 08028, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Institut de Biomedicina de la Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Jing Sui
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
| | - Rongtao Jiang
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Eden A. Evins
- Center for Addiction Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Psychiatry, Mass General Brigham and Harvard Medical School, Boston, MA 02115, USA
| | - Scott E. Hadland
- Division of Adolescent and Young Adult Medicine, Mass General for Children, Boston, MA 02114, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Joshua L. Roffman
- Department of Psychiatry, Mass General Brigham and Harvard Medical School, Boston, MA 02115, USA
| | - Richard T. Liu
- Department of Psychiatry, Mass General Brigham and Harvard Medical School, Boston, MA 02115, USA
- Stanly Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jodi Gilman
- Center for Addiction Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Psychiatry, Mass General Brigham and Harvard Medical School, Boston, MA 02115, USA
| | - Phil H. Lee
- Department of Psychiatry, Mass General Brigham and Harvard Medical School, Boston, MA 02115, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Stanly Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
20
|
Janivara R, Hazra U, Pfennig A, Harlemon M, Kim MS, Eaaswarkhanth M, Chen WC, Ogunbiyi A, Kachambwa P, Petersen LN, Jalloh M, Mensah JE, Adjei AA, Adusei B, Joffe M, Gueye SM, Aisuodionoe-Shadrach OI, Fernandez PW, Rohan TE, Andrews C, Rebbeck TR, Adebiyi AO, Agalliu I, Lachance J. Uncovering the genetic architecture and evolutionary roots of androgenetic alopecia in African men. HGG ADVANCES 2025; 6:100428. [PMID: 40134218 PMCID: PMC12000746 DOI: 10.1016/j.xhgg.2025.100428] [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: 01/22/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 03/27/2025] Open
Abstract
Androgenetic alopecia is a highly heritable trait. However, much of our understanding about the genetics of male-pattern baldness comes from individuals of European descent. Here, we examined a dataset comprising 2,136 men from Ghana, Nigeria, Senegal, and South Africa that were genotyped using the Men of African Descent and Carcinoma of the Prostate Array. We first tested how genetic predictions of baldness generalize from Europe to Africa and found that polygenic scores from European genome-wide association studies (GWASs) yielded area under the curve statistics that ranged from 0.513 to 0.546, indicating that genetic predictions of baldness generalized poorly from European to African populations. Subsequently, we conducted an African GWAS of androgenetic alopecia, focusing on self-reported baldness patterns at age 45. After correcting for age at recruitment, population structure, and study site, we identified 266 moderately significant associations, 51 of which were independent (p < 10-5, r2 < 0.2). Most baldness associations were autosomal, and the X chromosome does not seem to have a large impact on baldness in African men. Although Neanderthal alleles have previously been associated with skin and hair phenotypes, within the limits of statistical power, we did not find evidence that continental differences in the genetic architecture of baldness are due to Neanderthal introgression. While most loci that are associated with androgenetic alopecia do not have large integrative haplotype scores or fixation index statistics, multiple baldness-associated SNPs near the EDA2R and AR genes have large allele frequency differences between continents. Collectively, our findings illustrate how population genetic differences contribute to the limited portability of polygenic predictions across ancestries.
Collapse
Affiliation(s)
- Rohini Janivara
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Ujani Hazra
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Aaron Pfennig
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | - Maxine Harlemon
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA; Department of Biology, Morgan State University, Baltimore, MD, USA
| | - Michelle S Kim
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA; Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | | | - Wenlong C Chen
- Strengthening Oncology Services Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; National Cancer Registry, National Institute for Communicable Diseases a Division of the National Health Laboratory Service, Johannesburg, South Africa
| | | | - Paidamoyo Kachambwa
- Centre for Proteomic and Genomic Research, Cape Town, South Africa; Mediclinic Precise Southern Africa, Cape Town, South Africa
| | - Lindsay N Petersen
- Centre for Proteomic and Genomic Research, Cape Town, South Africa; Mediclinic Precise Southern Africa, Cape Town, South Africa
| | - Mohamed Jalloh
- Université Cheikh Anta Diop de Dakar, Dakar, Senegal; Université Iba Der Thiam de Thiès, Thiès, Senegal
| | - James E Mensah
- Korle-Bu Teaching Hospital and University of Ghana Medical School, Accra, Ghana
| | - Andrew A Adjei
- Department of Pathology, University of Ghana Medical School, Accra, Ghana
| | | | - Maureen Joffe
- Strengthening Oncology Services Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Oseremen I Aisuodionoe-Shadrach
- College of Health Sciences, University of Abuja, University of Abuja Teaching Hospital and Cancer Science Centre, Abuja, Nigeria
| | - Pedro W Fernandez
- Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Timothy R Rebbeck
- Dana-Farber Cancer Institute, Boston, MA, USA; Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Ilir Agalliu
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Joseph Lachance
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
21
|
Lawson LP, Parameswaran S, Panganiban RA, Constantine GM, Weirauch MT, Kottyan LC. Update on the genetics of allergic diseases. J Allergy Clin Immunol 2025:S0091-6749(25)00327-6. [PMID: 40139464 DOI: 10.1016/j.jaci.2025.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/24/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025]
Abstract
The field of genetic etiology of allergic diseases has advanced significantly in recent years. Shared risk loci reflect the contribution of genetic factors to the sequential development of allergic conditions across the atopic march, while unique risk loci provide opportunities to understand tissue specific manifestations of allergic disease. Most identified risk variants are noncoding, indicating that they likely influence gene expression through gene regulatory mechanisms. Despite recent advances, challenges persist, particularly regarding the need for increased ancestral diversity in research populations. Further, while polygenic risk scores show promise for identifying individuals at higher genetic risk for allergic diseases, their predictive accuracy varies across different ancestries and can be difficult to translate to an individual's absolute risk of developing a disease. Methodologies, including "nearest gene," 3D chromatin interaction analysis, expression quantitative trait locus analysis, experimental screens, and integrative bioinformatic models, have established connections between genetic variants and their regulatory targets, enhancing our understanding of disease risk and phenotypic variability. In this review, we focus on the state of knowledge of allergic sensitization and 5 allergic diseases: asthma, atopic dermatitis, allergic rhinitis, food allergy, and eosinophilic esophagitis. We summarize recent progress and highlight opportunities for advancing our understanding of their genetic etiology.
Collapse
Affiliation(s)
- Lucinda P Lawson
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Sreeja Parameswaran
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Ronald A Panganiban
- Asthma Research, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Gregory M Constantine
- Human Eosinophil Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, Md
| | - Matthew T Weirauch
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Leah C Kottyan
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| |
Collapse
|
22
|
Citu C, Chang L, Manuel AM, Enduru N, Zhao Z. Identification and catalog of viral transcriptional regulators in human diseases. iScience 2025; 28:112081. [PMID: 40124487 PMCID: PMC11928865 DOI: 10.1016/j.isci.2025.112081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/02/2025] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
Viral genomes encode viral transcriptional regulators (vTRs) that manipulate host gene expression to facilitate replication and evade immune detection. Nevertheless, their role in non-cancerous diseases remains largely underexplored. Here, we unveiled 268 new candidate vTRs from 14 of the 20 viral families we investigated. We mapped vTRs' genome-wide binding profiles and identified their potential human targets, which were enriched in immune-mediated pathways, neurodegenerative disorders, and cancers. Through vTR DNA-binding preference analysis, 283 virus-specific and human-like motifs were identified. Prioritized Epstein-Barr virus (EBV) vTR target genes were associated with multiple sclerosis (MS), rheumatoid arthritis, and systemic lupus erythematosus. The partitioned heritability study among 19 diseases indicated significant enrichment of these diseases in EBV vTR-binding sites, implicating EBV vTRs' roles in immune-mediated disorders. Finally, drug repurposing analysis pinpointed candidate drugs for MS, asthma, and Alzheimer disease. This study enhances our understanding of vTRs in diverse human diseases and identifies potential therapeutic targets for future investigation.
Collapse
Affiliation(s)
- Citu Citu
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Le Chang
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Astrid M. Manuel
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Nitesh Enduru
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Zhongming Zhao
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| |
Collapse
|
23
|
Wang Y, Luo J, Huang R, Xiao Y. Nonlinear association of TSH with pulmonary ventilation: insights from bidirectional Mendelian randomization and cross-sectional study. BMC Pulm Med 2025; 25:126. [PMID: 40108567 PMCID: PMC11921497 DOI: 10.1186/s12890-025-03584-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 03/06/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Thyroid hormones play a crucial role in numerous physiological processes, including pulmonary function. However, the relationship between thyroid function and different patterns of pulmonary ventilation remains unclear. METHODS This study employed a bidirectional two-sample Mendelian randomization (MR) approach combined with a cross-sectional study from the National Health and Nutrition Examination Survey (NHANES) to explore the relationship between thyroid function and pulmonary ventilation indicators. We used genomic data from the ThyroidOmics Consortium and the UK Biobank to derive instrumental variables for thyroid and pulmonary functions. Adults from the NHANES 2007-2012 were included to validate the MR findings through weighted generalized linear model (GLM) regression and restricted cubic spline (RCS) analysis. RESULTS Genetically predicted thyroid-stimulating hormone (TSH) was associated with pulmonary ventilatory function (forced expiratory volume in 1 s (FEV1): β = 0.0223, 95% confidence interval (CI) 0.0040-0.0406, p-value = 0.0170), particularly with a restrictive ventilatory pattern (forced vital capacity (FVC): β = 0.0237, 95% CI 0.0047-0.0427, p-value = 0.0143). This association was more robust in the low TSH subgroup. Additionally, the NHANES data revealed a nonlinear relationship between both FEV1% predicted and FVC% predicted and TSH, characterized by a positive relationship at lower TSH ranges and a negative relationship at higher TSH ranges. CONCLUSIONS Our findings highlight a significant association between TSH levels and a restrictive ventilatory pattern, underscoring the importance of thyroid health in the clinical evaluation of certain pulmonary diseases. These insights may guide more personalized interventions in respiratory medicine.
Collapse
Affiliation(s)
- Yuxin Wang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jinmei Luo
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Rong Huang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yi Xiao
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
24
|
Allegrini AG, Hannigan LJ, Frach L, Barkhuizen W, Baldwin JR, Andreassen OA, Bragantini D, Hegemann L, Havdahl A, Pingault JB. Intergenerational transmission of polygenic predisposition for neuropsychiatric traits on emotional and behavioural difficulties in childhood. Nat Commun 2025; 16:2674. [PMID: 40102402 PMCID: PMC11920414 DOI: 10.1038/s41467-025-57694-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: 03/22/2024] [Accepted: 02/28/2025] [Indexed: 03/20/2025] Open
Abstract
Childhood emotional and behavioural difficulties tend to co-occur and often precede diagnosed neuropsychiatric conditions. Identifying shared and specific risk factors for early-life mental health difficulties is therefore essential for prevention strategies. Here, we examine how parental risk factors shape their offspring's emotional and behavioural symptoms (e.g. feelings of anxiety, and restlessness) using data from 14,959 genotyped family trios from the Norwegian Mother, Father and Child Cohort Study (MoBa). We model maternal reports of emotional and behavioural symptoms, organizing them into general and specific domains. We then investigate the direct (genetically transmitted) and indirect (environmentally mediated) contributions of parental polygenic risk for neuropsychiatric-related traits and whether these are shared across symptoms. We observe evidence consistent with an environmental route to general symptomatology beyond genetic transmission, while also demonstrating domain-specific direct and indirect genetic contributions. These findings improve our understanding of early risk pathways that can be targeted in preventive interventions aiming to interrupt the intergenerational cycle of risk transmission.
Collapse
Affiliation(s)
- A G Allegrini
- Division of Psychology and Language Sciences, Department of Clinical, Educational and Health Psychology, University College London, London, UK.
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| | - L J Hannigan
- Research Department, Lovisenberg Diaconal Hospital, Oslo, Norway
- PsychGen Center for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
- MRC Integrative Epidemiology Unit (IEU), University of Bristol, Bristol, United Kingdom
| | - L Frach
- Division of Psychology and Language Sciences, Department of Clinical, Educational and Health Psychology, University College London, London, UK
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - W Barkhuizen
- Division of Psychology and Language Sciences, Department of Clinical, Educational and Health Psychology, University College London, London, UK
| | - J R Baldwin
- Division of Psychology and Language Sciences, Department of Clinical, Educational and Health Psychology, University College London, London, UK
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - O A Andreassen
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - D Bragantini
- Research Department, Lovisenberg Diaconal Hospital, Oslo, Norway
- PsychGen Center for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - L Hegemann
- Research Department, Lovisenberg Diaconal Hospital, Oslo, Norway
- PsychGen Center for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - A Havdahl
- Research Department, Lovisenberg Diaconal Hospital, Oslo, Norway
- PsychGen Center for Genetic Epidemiology and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
- Department of Psychology, PROMENTA Research Centre, University of Oslo, Oslo, Norway
| | - J-B Pingault
- Division of Psychology and Language Sciences, Department of Clinical, Educational and Health Psychology, University College London, London, UK
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
25
|
Fujii R, Nagayoshi M, Nakatochi M, Sato S, Tsuboi Y, Suzuki K, Ikezaki H, Nishida Y, Kubo Y, Tanoue S, Suzuki S, Koyama T, Kuriki K, Takashima N, Katsuura-Kamano S, Momozawa Y, Wakai K, Matsuo K. Multi-Trait Polygenic Risk Score, Nongenetic Determinants, and Cardiovascular Disease Death: A Cohort Study of 14 086 Japanese Individuals. J Am Heart Assoc 2025; 14:e038572. [PMID: 40079315 DOI: 10.1161/jaha.124.038572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 02/04/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Although utility of composite trait-specific polygenic risk score (multi-trait PRS) has been examined among European ancestries, few studies investigated among East Asians and incorporated modifiable risk factors. We examined the associations of multi-trait PRS for cardiometabolic factors with cardiovascular disease mortality by integrating nongenetic determinants. METHODS A total of 14 086 Japanese participants (mean age, 55±9; 55.8% women) of the J-MICC (Japan Multi-Institutional Collaborative Cohort) study were analyzed in this study. We calculated 6 PRSs for cardiometabolic traits (systolic blood pressure, body mass index, triglycerides, low-density lipoprotein cholesterol, estimated glomerular filtration rate, and hemoglobin A1c). Based on these PRSs, we developed multi-trait PRS and considered as a primary exposure. Three nongenetic factors (smoking, alcohol drinking, and educational attainment) from the self-reported questionnaire were also examined. RESULTS During a median 12.1-year follow-up period, a total of 472 all-cause and 79 cardiovascular disease mortality cases were documented. Compared with 0% to 90% of multi-trait PRSs, an adjusted hazard ratio (HR) among the top 10% of multi-trait PRSs was 1.32 (95% CI, 1.00-1.73) for all-cause death and 2.63 (95% CI, 1.48-4.67) for cardiovascular disease death. Incorporation of educational attainment with multi-trait PRSs showed null associations in those who went beyond high school (HR, 2.07 [95% CI, 0.44-9.66]) even in the top 10% of multi-trait PRS. CONCLUSIONS Our analysis combining both genetic and nongenetic determinants highlighted that lifestyle factors and educational attainment can slightly reduce an individual's composite genetic risk for cardiovascular disease death.
Collapse
Affiliation(s)
- Ryosuke Fujii
- Department of Preventive Medical Sciences Fujita Health University School of Medical Sciences Toyoake Japan
- Department of Preventive Medicine Nagoya University Graduate School of Medicine Nagoya Japan
| | - Mako Nagayoshi
- Department of Preventive Medicine Nagoya University Graduate School of Medicine Nagoya Japan
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Department of Integrated Health Sciences Nagoya University Graduate School of Medicine Nagoya Japan
| | - Shuntaro Sato
- Clinical Research Center Nagasaki University Hospital Nagasaki Japan
| | - Yoshiki Tsuboi
- Department of Preventive Medical Sciences Fujita Health University School of Medical Sciences Toyoake Japan
| | - Koji Suzuki
- Department of Preventive Medical Sciences Fujita Health University School of Medical Sciences Toyoake Japan
| | - Hiroaki Ikezaki
- Department of Environmental Medicine and Infectious Disease, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
- Department of General Internal Medicine Kyushu University Hospital Fukuoka Japan
| | - Yuichiro Nishida
- Department of Preventive Medicine, Faculty of Medicine Saga University Saga Japan
| | - Yoko Kubo
- Department of Preventive Medicine Nagoya University Graduate School of Medicine Nagoya Japan
| | - Shiroh Tanoue
- Department of Epidemiology and Preventive Medicine Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Sadao Suzuki
- Department of Public Health Nagoya City University Graduate School of Medical Sciences Nagoya Japan
| | - Teruhide Koyama
- Department of Epidemiology for Community Health and Medicine Kyoto Prefectural University of Medicine Kyoto Japan
| | - Kiyonori Kuriki
- Laboratory of Public Health, Division of Nutritional Sciences, School of Food and Nutritional Sciences University of Shizuoka Shizuoka Japan
| | - Naoyuki Takashima
- Department of Epidemiology for Community Health and Medicine Kyoto Prefectural University of Medicine Kyoto Japan
- NCD Epidemiology Research Center Shiga University of Medical Science Otsu Shiga Japan
| | - Sakurako Katsuura-Kamano
- Department of Preventive Medicine Tokushima University Graduate School of Biomedical Sciences Tokushima Japan
| | - Yukihide Momozawa
- Laboratory for Genotyping Development Center for Integrative Medical Sciences, RIKEN Yokohama Kanagawa Japan
| | - Kenji Wakai
- Department of Preventive Medicine Nagoya University Graduate School of Medicine Nagoya Japan
| | - Keitaro Matsuo
- Division of Cancer Information and Control Aichi Cancer Center Research Institute Nagoya Japan
- Division of Descriptive Cancer Epidemiology Nagoya University Graduate School of Medicine Nagoya Japan
| |
Collapse
|
26
|
Grimmett ZW, Zhang R, Zhou HL, Chen Q, Miller D, Qian Z, Lin J, Kalra R, Gross SS, Koch WJ, Premont RT, Stamler JS. The denitrosylase SCoR2 controls cardioprotective metabolic reprogramming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642752. [PMID: 40161620 PMCID: PMC11952481 DOI: 10.1101/2025.03.12.642752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Acute myocardial infarction (MI) is a leading cause of morbidity and mortality, and therapeutic options remain limited. Endogenously generated nitric oxide (NO) is highly cardioprotective, but protection is not replicated by nitroso-vasodilators (e.g., nitrates, nitroprusside) used in clinical practice, highlighting specificity in NO-based signaling and untapped therapeutic potential. Signaling by NO is mediated largely by S-nitrosylation, entailing specific enzymes that form and degrade S-nitrosothiols in proteins (SNO-proteins), termed nitrosylases and denitrosylases, respectively. SNO-CoA Reductase 2 (SCoR2; product of the Akr1a1 gene) is a recently discovered protein denitrosylase. Genetic variants in SCoR2 have been associated with cardiovascular disease, but its function is unknown. Here we show that mice lacking SCoR2 exhibit robust protection in an animal model of MI. SCoR2 regulates ketolytic energy availability, antioxidant levels and polyol homeostasis via S-nitrosylation of key metabolic effectors. Human cardiomyopathy shows reduced SCoR2 expression and an S-nitrosylation signature of metabolic reprogramming, mirroring SCoR2-/- mice. Deletion of SCoR2 thus coordinately reprograms multiple metabolic pathways-ketone body utilization, glycolysis, pentose phosphate shunt and polyol metabolism-to limit infarct size, establishing SCoR2 as a novel regulator in the injured myocardium and a potential drug target. Impact statement Mice lacking the denitrosylase enzyme SCoR2/AKR1A1 demonstrate robust cardioprotection resulting from reprogramming of multiple metabolic pathways, revealing widespread, coordinated metabolic regulation by SCoR2.
Collapse
Affiliation(s)
- Zachary W. Grimmett
- Medical Scientist Training Program, Case Western Reserve University School of Medicine, Cleveland OH, 44106
- Institute for Transformative Molecular Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland OH, 44106
| | - Rongli Zhang
- Institute for Transformative Molecular Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland OH, 44106
- Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland OH, 44106
| | - Hua-Lin Zhou
- Institute for Transformative Molecular Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland OH, 44106
| | - Qiuying Chen
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, 10065
| | - Dawson Miller
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, 10065
| | - Zhaoxia Qian
- Institute for Transformative Molecular Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland OH, 44106
| | - Justin Lin
- Institute for Transformative Molecular Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland OH, 44106
| | - Riti Kalra
- Institute for Transformative Molecular Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland OH, 44106
| | - Steven S. Gross
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, 10065
| | - Walter J. Koch
- Department of Surgery, Duke University School of Medicine, Durham NC, 27710
- Department of Medicine, Duke University School of Medicine, Durham NC, 27710
| | - Richard T. Premont
- Institute for Transformative Molecular Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland OH, 44106
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland OH, 44106
| | - Jonathan S. Stamler
- Institute for Transformative Molecular Medicine, Department of Medicine, Case Western Reserve University School of Medicine, Cleveland OH, 44106
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland OH, 44106
| |
Collapse
|
27
|
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
|
28
|
Asare Y, Yan G, Schlegl C, Prestel M, van der Vorst EPC, Teunissen AJP, Aronova A, Tosato F, Naser N, Caputo J, Prevot G, Azzun A, Wefers B, Wurst W, Schneider M, Forne I, Bidzhekov K, Naumann R, van der Laan SW, Brandhofer M, Cao J, Roth S, Malik R, Tiedt S, Mulder WJM, Imhof A, Liesz A, Weber C, Bernhagen J, Dichgans M. A cis-regulatory element controls expression of histone deacetylase 9 to fine-tune inflammasome-dependent chronic inflammation in atherosclerosis. Immunity 2025; 58:555-567.e9. [PMID: 39879983 DOI: 10.1016/j.immuni.2025.01.003] [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: 01/24/2024] [Revised: 09/03/2024] [Accepted: 01/03/2025] [Indexed: 01/31/2025]
Abstract
Common genetic variants in a conserved cis-regulatory element (CRE) at histone deacetylase (HDAC)9 are a major risk factor for cardiovascular disease, including stroke and coronary artery disease. Given the consistency of this association and its proinflammatory properties, we examined the mechanisms whereby HDAC9 regulates vascular inflammation. HDAC9 bound and mediated deacetylation of NLRP3 in the NACHT and LRR domains leading to inflammasome activation and lytic cell death. Targeted deletion of the critical CRE in mice increased Hdac9 expression in myeloid cells to exacerbate inflammasome-dependent chronic inflammation. In human carotid endarterectomy samples, increased HDAC9 expression was associated with atheroprogression and clinical plaque instability. Incorporation of TMP195, a class IIa HDAC inhibitor, into lipoprotein-based nanoparticles to target HDAC9 at the site of myeloid-driven vascular inflammation stabilized atherosclerotic plaques, implying a lower risk of plaque rupture and cardiovascular events. Our findings link HDAC9 to atherogenic inflammation and provide a paradigm for anti-inflammatory therapeutics for atherosclerosis.
Collapse
Affiliation(s)
- Yaw Asare
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany.
| | - Guangyao Yan
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Christina Schlegl
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Matthias Prestel
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Emiel P C van der Vorst
- Institute for Cardiovascular Prevention (IPEK), LMU, Munich, Germany; Institute for Molecular Cardiovascular Research (IMCAR), Aachen-Maastricht Institute for CardioRenal Disease (AMICARE) & Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
| | - Abraham J P Teunissen
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Arailym Aronova
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Federica Tosato
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Nawraa Naser
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Julio Caputo
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Geoffrey Prevot
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anthony Azzun
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benedikt Wefers
- Deutsches Zentrum für Neurodegenerative Erkrankungen e. V. (DZNE), Munich, Germany
| | - Wolfgang Wurst
- Deutsches Zentrum für Neurodegenerative Erkrankungen e. V. (DZNE), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Melanie Schneider
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Ignasi Forne
- Protein Analysis Unit, Faculty of Medicine, Biomedical Center, LMU, Martinsried, Germany
| | - Kiril Bidzhekov
- Institute for Cardiovascular Prevention (IPEK), LMU, Munich, Germany
| | - Ronald Naumann
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Sander W van der Laan
- Central Diagnostics Laboratory, Division of Laboratory, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, University of Utrecht, Utrecht, the Netherlands
| | - Markus Brandhofer
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Jiayu Cao
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Stefan Roth
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Rainer Malik
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Steffen Tiedt
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany
| | - Willem J M Mulder
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Center, Laboratory of Chemical Biology, Nijmegen, the Netherlands
| | - Axel Imhof
- Protein Analysis Unit, Faculty of Medicine, Biomedical Center, LMU, Martinsried, Germany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), LMU, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (MHA), Munich, Germany; Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Jürgen Bernhagen
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (MHA), Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), Munich, Germany; Deutsches Zentrum für Neurodegenerative Erkrankungen e. V. (DZNE), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (MHA), Munich, Germany.
| |
Collapse
|
29
|
Zhan H, Cammann D, Cummings JL, Dong X, Chen J. Biomarker identification for Alzheimer's disease through integration of comprehensive Mendelian randomization and proteomics data. J Transl Med 2025; 23:278. [PMID: 40050982 PMCID: PMC11884171 DOI: 10.1186/s12967-025-06317-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/23/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the main cause of dementia with few effective therapies. We aimed to identify potential plasma biomarkers or drug targets for AD by investigating the causal association between plasma proteins and AD by integrating comprehensive Mendelian randomization (MR) and multi-omics data. METHODS Using two-sample MR, cis protein quantitative trait loci (cis-pQTLs) for 1,916 plasma proteins were used as an exposure to infer their causal effect on AD liability in individuals of European ancestry, with two large-scale AD genome-wide association study (GWAS) datasets as the outcome for discovery and replication. Significant causal relationships were validated by sensitivity analyses, reverse MR analysis, and Bayesian colocalization analysis. Additionally, we investigated the causal associations at the transcriptional level with cis gene expression quantitative trait loci (cis-eQTLs) data across brain tissues and blood in European ancestry populations, as well as causal plasma proteins in African ancestry populations. RESULTS In those of European ancestry, the genetically predicted levels of five plasma proteins (BLNK, CD2AP, GRN, PILRA, and PILRB) were causally associated with AD. Among these five proteins, GRN was protective against AD, while the rest were risk factors. Consistent causal effects were found in the brain for cis-eQTLs of GRN, BLNK, and CD2AP, while the same was true for PILRA in the blood. None of the plasma proteins were significantly associated with AD in persons of African ancestry. CONCLUSIONS Comprehensive MR analyses with multi-omics data identified five plasma proteins that had causal effects on AD, highlighting potential biomarkers or drug targets for better diagnosis and treatment for AD.
Collapse
Affiliation(s)
- Hui Zhan
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA
| | - Davis Cammann
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA
- School of Life Science, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA
| | - Jeffrey L Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, Kirk Kerkorian School of Medicine, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA
| | - Xianjun Dong
- Stephen and Denise Adams Center for Parkinson's Disease Research, Yale School of Medicine, Yale University, New Haven, CT, USA
- Department of Neurology and Section of Biomedical Informatics and Data Science (BIDS), Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Jingchun Chen
- Interdisciplinary Neuroscience Program, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA.
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA.
- School of Life Science, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, USA.
| |
Collapse
|
30
|
Adewuyi EO, Laws SM. Genomic Characterisation of the Relationship and Causal Links Between Vascular Calcification, Alzheimer's Disease, and Cognitive Traits. Biomedicines 2025; 13:618. [PMID: 40149595 PMCID: PMC11940612 DOI: 10.3390/biomedicines13030618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Observational studies suggest a link between vascular calcification and dementia or cognitive decline, but the evidence is conflicting, and the underlying mechanisms are unclear. Here, we investigate the shared genetic and causal relationships of vascular calcification-coronary artery calcification (CAC) and abdominal aortic calcification (AAC)-with Alzheimer's disease (AD), and five cognitive traits. Methods: We analyse large-scale genome-wide association studies (GWAS) summary statistics, using well-regarded methods, including linkage disequilibrium score regression (LDSC), Mendelian randomisation (MR), pairwise GWAS (GWAS-PW), and gene-based association analysis. Results: Our findings reveal a nominally significant positive genome-wide genetic correlation between CAC and AD, which becomes non-significant after excluding the APOE region. CAC and AAC demonstrate significant negative correlations with cognitive performance and educational attainment. MR found no causal association between CAC or AAC and AD or cognitive traits, except for a bidirectional borderline-significant association between AAC and fluid intelligence scores. Pairwise-GWAS analysis identifies no shared causal SNPs (posterior probability of association [PPA]3 < 0.5). However, we find pleiotropic loci (PPA4 > 0.9), particularly on chromosome 19, with gene association analyses revealing significant genes in shared regions, including APOE, TOMM40, NECTIN2, and APOC1. Moreover, we identify suggestively significant loci (PPA4 > 0.5) on chromosomes 1, 6, 7, 9 and 19, implicating pleiotropic genes, including NAV1, IPO9, PHACTR1, UFL1, FHL5, and FOCAD. Conclusions: Current findings reveal limited genetic correlation and no significant causal associations of CAC and AAC with AD or cognitive traits. However, significant pleiotropic loci, particularly at the APOE region, highlight the complex interplay between vascular calcification and neurodegenerative processes. Given APOE's roles in lipid metabolism, neuroinflammation, and vascular integrity, its involvement may link vascular and neurodegenerative disorders, pointing to potential targets for further investigation.
Collapse
Affiliation(s)
- Emmanuel O. Adewuyi
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
- Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia;
| | - Simon M. Laws
- Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia;
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia
| |
Collapse
|
31
|
Pilalis E, Zisis D, Andrinopoulou C, Karamanidou T, Antonara M, Stavropoulos TG, Chatziioannou A. Genome-wide functional annotation of variants: a systematic review of state-of-the-art tools, techniques and resources. Front Pharmacol 2025; 16:1474026. [PMID: 40098614 PMCID: PMC11911558 DOI: 10.3389/fphar.2025.1474026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
The recent advancement of sequencing technologies marks a significant shift in the character and complexity of the digital genomic data universe, encompassing diverse types of molecular data, screened through manifold technological platforms. As a result, a plethora of fully assembled genomes are generated that span vertically the evolutionary scale. Notwithstanding the tsunami of thriving innovations that accomplish unprecedented, nucleotide-level, structural and functional annotation, an exhaustive, systemic, massive genome-wide functional annotation remains elusive, particularly when the criterion is automation and efficiency in data-agnostic interpretation. The latter is of paramount importance for the elaboration of strategies for sophisticated, data-driven genome-wide annotation, which aim to impart a sustainable and comprehensive systemic approach to addressing whole genome variation. Therefore, it is essential to develop methods and tools that promote systematic functional genomic annotation, with emphasis on mechanistic information exceeding the limits of coding regions, and exploiting the chunks of pertinent information residing in non-coding regions, including promoter and enhancer sequences, non-coding RNAs, DNA methylation sites, transcription factor binding sites, transposable elements and more. This review provides an overview of the current state-of-the-art in genome-wide functional annotation of genetic variation, including existing bioinformatic tools, resources, databases and platforms currently available or reported in the literature. Particular emphasis is placed on the functional annotation of variants that lie outside protein-coding genomic regions (intronic or intergenic), their potential co-localization with regulatory element areas, such as putative non-coding RNA regions, and the assessment of their functional impact on the investigated phenotype. In addition, state-of-the-art tools that leverage data obtained from WGS and GWAS-based analyses are discussed, along with future bioinformatics directions and developments. These future directions emphasize efficient, comprehensive, and largely automated functional annotation of both coding and non-coding genomic variants, as well as their optimal evaluation.
Collapse
Affiliation(s)
| | | | | | | | - Maria Antonara
- Pfizer Center for Digital Innovation, Thessaloniki, Greece
| | | | - Aristotelis Chatziioannou
- e-NIOS Applications PC, Kallithea, Greece
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
32
|
Yamamoto Y, Shirai Y, Edahiro R, Kumanogoh A, Okada Y. Large-scale cross-trait genetic analysis highlights shared genetic backgrounds of autoimmune diseases. Immunol Med 2025; 48:1-10. [PMID: 39171621 DOI: 10.1080/25785826.2024.2394258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/15/2024] [Indexed: 08/23/2024] Open
Abstract
Disorders associated with the immune system burden multiple organs, although the shared biology exists across the diseases. Preceding family-based studies reveal that immune diseases are heritable to varying degrees, providing the basis for immunogenomics. The recent cost reduction in genetic analysis intensively promotes biobank-scale studies and the development of frameworks for statistical genetics. The accumulating multi-layer omics data, including genome-wide association studies (GWAS) and RNA-sequencing at single-cell resolution, enable us to dissect the genetic backgrounds of immune-related disorders. Although autoimmune and allergic diseases are generally categorized into different disease categories, epidemiological studies reveal the high incidence of autoimmune and allergic disease complications, suggesting the shared genetics and biology between the disease categories. Biobank resources and consortia cover multiple immune-related disorders to accumulate phenome-wide associations of genetic variants and enhance researchers to analyze the shared and heterogeneous genetic backgrounds. The emerging post-GWAS and integrative multi-omics analyses provide genetic and biological insights into the multicategorical disease associations.
Collapse
Affiliation(s)
- Yuji Yamamoto
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yuya Shirai
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- RIKEN Center for Integrative Medical Sciences, Laboratory for Systems Genetics, Yokohama, Japan
| | - Ryuya Edahiro
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
- RIKEN Center for Integrative Medical Sciences, Laboratory for Systems Genetics, Yokohama, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Japan
- Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Japan
- Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Suita, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- RIKEN Center for Integrative Medical Sciences, Laboratory for Systems Genetics, Yokohama, Japan
- Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Japan
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Japan
| |
Collapse
|
33
|
Dou C, Liu D, Kong L, Chen M, Ye C, Zhu Z, Zheng J, Xu M, Xu Y, Li M, Zhao Z, Lu J, Chen Y, Ning G, Wang W, Bi Y, Wang T. Shared genetic architecture of type 2 diabetes with muscle mass and function and frailty reveals comorbidity etiology and pleiotropic druggable targets. Metabolism 2025; 164:156112. [PMID: 39710002 DOI: 10.1016/j.metabol.2024.156112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/19/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Delineating the shared genetic architecture of type 2 diabetes with muscle mass and function and frailty is essential for unraveling the common etiology and developing holistic therapeutic strategies for these co-existing conditions. METHODS In this genome-wide pleiotropic association study, we performed multi-level pairwise trait pleiotropic analyses using genome-wide association study summary statistics from up to 461,026 European ancestry individuals to dissect the shared genetic factors and causal relationships of type 2 diabetes and seven glycemic traits with four muscle mass- and function-related phenotypes and the frailty index. RESULTS We first identified 27 pairs with significant genetic correlations through the linkage disequilibrium score regression and high-definition likelihood analysis. Then we determined 79 pleiotropic loci and 109 pleiotropic genes across linkage pairs via the pleiotropic analysis under the composite null hypothesis (PLACO), the colocalization, and the Multi-marker Analysis of GenoMic Annotation (MAGMA) analyses. We subsequently performed transcriptome-wide association study (TWAS) analyses using joint-tissue imputation, refined by gene-based integrative fine-mapping through a conditional TWAS approach, and identified 44 unique causal shared genes across 13 tissues in linkage pairs, including eight druggable genes (ABO, AOC1, FTO, GCKR, MTOR, POLK, PPARG, and APEH), with MTOR and PPARG categorized as clinically actionable. Two-sample Mendelian randomization analysis supported bidirectional causality between diabetes and frailty index and unidirectional causal effects of muscle phenotypes on glycemic profiles. CONCLUSIONS Our findings highlight the common genetic underpinnings between type 2 diabetes and muscle loss and frailty and inform drug targets with pleiotropic effects on both of these aging-related challenges.
Collapse
Affiliation(s)
- Chun Dou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dong Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lijie Kong
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingling Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chaojie Ye
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Zhu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mian Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyun Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhong Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiange Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
34
|
Li B, Xu H, Wu L. Genetic insights into cardiac conduction disorders from genome-wide association studies. Hum Genomics 2025; 19:20. [PMID: 40022259 PMCID: PMC11871809 DOI: 10.1186/s40246-025-00732-x] [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: 11/18/2024] [Accepted: 02/15/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Substantial data support a heritable basis for cardiac conduction disorders (CCDs), but the genetic determinants and molecular mechanisms of these arrhythmias are poorly understood, therefore, we sought to identify genetic loci associated with CCDs. METHODS We performed meta-analyses of genome-wide association studies to identify genetic loci for atrioventricular block (AVB), left bundle branch block (LBBB), and right bundle branch block (RBBB) from public data from the UK Biobank and FinnGen consortium. We assessed evidence supporting the potential causal effects of candidate genes by analyzing relations between associated variants and cardiac gene expression, performing transcriptome-wide analyses, and ECG-wide phenome-wide associations for each indexed SNP. RESULTS Analysis comprised over 700,000 individuals for each trait. We identified 10, 4 and 0 significant loci for AVB (PLEKHA3, TTN, FNDC3B, SENP2, SCN10A, RRH, PPARGC1A, PKD2L2, NKX2-5 and TBX20), LBBB (PPARGC1A, HAND1, TBX5, and ADAMTS5) and RBBB, respectively. Transcriptome-wide association analysis supported an association between reduced predicted cardiac expression of SCN10A and AVB. Phenome-wide associations identified traits with both cardiovascular and non- cardiovascular traits with indexed SNPs. CONCLUSIONS Our analysis highlight gene regions associated with channel function, cardiac development, sarcomere function and energy modulation as important potential effectors of CCDs susceptibility.
Collapse
Affiliation(s)
- Bingxun Li
- Department of Cardiology, Peking University First Hospital, NO.8 Xishiku Street, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Hongxuan Xu
- Department of Cardiology, Peking University First Hospital, NO.8 Xishiku Street, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Lin Wu
- Department of Cardiology, Peking University First Hospital, NO.8 Xishiku Street, Beijing, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.
| |
Collapse
|
35
|
Reus LM, Boltz T, Francia M, Bot M, Ramesh N, Koromina M, Pijnenburg YAL, den Braber A, van der Flier WM, Visser PJ, van der Lee SJ, Tijms BM, Teunissen CE, Loohuis LO, Ophoff RA. Quantitative trait loci mapping of circulating metabolites in cerebrospinal fluid to uncover biological mechanisms involved in brain-related phenotypes. Mol Psychiatry 2025:10.1038/s41380-025-02934-0. [PMID: 40021830 DOI: 10.1038/s41380-025-02934-0] [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: 11/07/2023] [Revised: 12/16/2024] [Accepted: 02/12/2025] [Indexed: 03/03/2025]
Abstract
Genomic studies of molecular traits have provided mechanistic insights into complex disease, though these lag behind for brain-related traits due to the inaccessibility of brain tissue. We leveraged cerebrospinal fluid (CSF) to study neurobiological mechanisms in vivo, measuring 5543 CSF metabolites, the largest panel in CSF to date, in 977 individuals of European ancestry. Individuals originated from two separate cohorts including cognitively healthy subjects (n = 490) and a well-characterized memory clinic sample, the Amsterdam Dementia Cohort (ADC, n = 487). We performed metabolite quantitative trait loci (mQTL) mapping on CSF metabolomics and found 126 significant mQTLs, representing 65 unique CSF metabolites across 51 independent loci. To better understand the role of CSF mQTLs in brain-related disorders we integrated our CSF mQTL results with pre-existing summary statistics on brain traits, identifying 34 genetic associations between CSF metabolites and brain traits. Over 90% of significant mQTLs demonstrated colocalized associations with brain-specific gene expression, unveiling potential neurobiological pathways.
Collapse
Affiliation(s)
- Lianne M Reus
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA.
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands.
| | - Toni Boltz
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA.
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| | - Marcelo Francia
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Merel Bot
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Naren Ramesh
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Maria Koromina
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, NY, USA
| | - Yolande A L Pijnenburg
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Anouk den Braber
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Department of Biological Psychology, Amsterdam Public Health Research Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Department of Psychiatry, Maastricht University, Maastricht, The Netherlands
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Sven J van der Lee
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Section Genomics of Neurodegenerative Diseases and Aging, Department of Human Genetics, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Betty M Tijms
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Neurodegeneration, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Charlotte E Teunissen
- Neurochemistry Lab, Department of Laboratory Medicine, Amsterdam Neuroscience, Neurodegeneration, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Loes Olde Loohuis
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Computational Medicine, University of California, Los Angeles, CA, USA
| | - Roel A Ophoff
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA.
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Psychiatry, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
36
|
Xu B, Forthman KL, Kuplicki R, Ahern J, Loughnan R, Naber F, Thompson WK, Nemeroff CB, Paulus MP, Fan CC. Genetic Correlates of Treatment-Resistant Depression. JAMA Psychiatry 2025:2830400. [PMID: 40009368 PMCID: PMC11866074 DOI: 10.1001/jamapsychiatry.2024.4825] [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: 07/08/2024] [Accepted: 12/03/2024] [Indexed: 02/27/2025]
Abstract
Importance Treatment-resistant depression (TRD) is a major challenge in mental health, affecting a significant number of patients and leading to considerable burdens. The etiological factors contributing to TRD are complex and not fully understood. Objective To investigate the genetic factors associated with TRD using polygenic scores (PGS) across various traits and explore their potential role in the etiology of TRD using large-scale genomic data from the All of Us (AoU) Research Program. Design, Setting, and Participants This study was a cohort design with observational data from participants in the AoU Research Program who have both electronic health records and genomic data. Data analysis was performed from March 27 to October 24, 2024. Exposures PGS for 61 unique traits from 7 domains. Main Outcomes and Measures Logistic regressions to test if PGS was associated with treatment-resistant depression (TRD) compared with treatment-responsive major depressive disorder (trMDD). Cox proportional hazard model was used to determine if the progressions from MDD to TRD were associated with PGS. Results A total of 292 663 participants (median [IQR] age, 57 (41-69) years; 175 981 female [60.1%]) from the AoU Research Program were included in this analysis. In the discovery set (124 945 participants), 11 of the selected PGS were found to have stronger associations with TRD than with trMDD, encompassing PGS from domains in education, cognition, personality, sleep, and temperament. Genetic predisposition for insomnia (odds ratio [OR], 1.11; 95% CI, 1.07-1.15) and specific neuroticism (OR, 1.11; 95% CI, 1.07-1.16) traits were associated with increased TRD risk, whereas higher education (OR, 0.88; 95% CI, 0.85-0.91) and intelligence (OR, 0.91; 95% CI, 0.88-0.94) scores were protective. The associations held across different TRD definitions (meta-analytic R2 >83%) and were consistent across 2 other independent sets within AoU (the whole-genome sequencing Diversity dataset, 104 388, and Microarray dataset, 63 330). Among 28 964 individuals followed up over time, 3854 developed TRD within a mean of 944 days (95% CI, 883-992 days). All 11 previously identified and replicated PGS were found to be modulating the conversion rate from MDD to TRD. Conclusions and Relevance Results of this cohort study suggest that genetic predisposition related to neuroticism, cognitive function, and sleep patterns had a significant association with the development of TRD. These findings underscore the importance of considering psychosocial factors in managing and treating TRD. Future research should focus on integrating genetic data with clinical outcomes to enhance understanding of pathways leading to treatment resistance.
Collapse
Affiliation(s)
- Bohan Xu
- Population Neuroscience and Genetics Center, Laureate Institute for Brain Research, Tulsa, Oklahoma
- Laureate Institute for Brain Research, Tulsa, Oklahoma
| | | | | | - Jonathan Ahern
- Population Neuroscience and Genetics Center, Laureate Institute for Brain Research, Tulsa, Oklahoma
- Center for Human Development, University of California, San Diego, La Jolla
| | - Robert Loughnan
- Population Neuroscience and Genetics Center, Laureate Institute for Brain Research, Tulsa, Oklahoma
- Center for Human Development, University of California, San Diego, La Jolla
| | - Firas Naber
- Population Neuroscience and Genetics Center, Laureate Institute for Brain Research, Tulsa, Oklahoma
- Laureate Institute for Brain Research, Tulsa, Oklahoma
| | - Wesley K. Thompson
- Population Neuroscience and Genetics Center, Laureate Institute for Brain Research, Tulsa, Oklahoma
- Laureate Institute for Brain Research, Tulsa, Oklahoma
- Division of Biostatistics and Bioinformatics, the Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla
| | - Charles B. Nemeroff
- Department of Psychiatry and Behavioral Sciences, Dell Medical School, The University of Texas at Austin, Austin
| | - Martin P. Paulus
- Laureate Institute for Brain Research, Tulsa, Oklahoma
- Department of Psychiatry, University of California, San Diego, La Jolla
| | - Chun Chieh Fan
- Population Neuroscience and Genetics Center, Laureate Institute for Brain Research, Tulsa, Oklahoma
- Laureate Institute for Brain Research, Tulsa, Oklahoma
- Department of Radiology, University of California, San Diego, La Jolla
| |
Collapse
|
37
|
Zhu L, Zhang Y, Chen X, Li Y, Pan H, Wang Y, Chen N, Wu Y, Li Y, Zhao M. Correlation Analysis of Pyroptosis-Related Genes CASP1, NLRP3, AIM2, and NLRP1 With Lung Adenocarcinoma. Int J Genomics 2025; 2025:8282590. [PMID: 40026444 PMCID: PMC11871981 DOI: 10.1155/ijog/8282590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 01/08/2025] [Indexed: 03/05/2025] Open
Abstract
Purpose: This study is aimed at exploring the role of pyroptosis-related genes in the development, immune infiltration, and clinical features of lung adenocarcinoma. Method: Pyroptosis-related genes were searched using online databases, including MSigDB, Gene, and GeneCards. We explored pyroptosis-related gene expression patterns in lung adenocarcinoma using the UALCAN database. Functional enrichment analysis of pyroptosis-related genes in lung adenocarcinoma was performed using the Metascape database. A protein-protein interaction network was constructed using the STRING database, and the outcomes were visualized using Cytoscape. The top five core genes were screened utilizing the MCC algorithm with its cytoHubba plugin. The correlation between immune cell infiltration, diagnosis, and prognosis of core genes in lung adenocarcinoma was explored using the TIMER 2.0, TCGA, and Kaplan-Meier plotter databases. A nomogram was constructed to predict the survival of patients with lung adenocarcinoma using Cox regression analysis, and its clinical value was validated. Samples of paraffin-embedded lung adenocarcinoma tissues were collected and subjected to immunohistochemical tests to verify the expression of core genes in lung adenocarcinoma and adjacent tissues. Results: Overall, 202 genes related to pyroptosis were identified, with 67 upregulated and 60 downregulated in lung adenocarcinomas. The top five core genes-namely, CASP1 (caspase1), PYCARD (PYD and CARD domain-containing protein), NLRP3 (NOD-like receptor protein 3), AIM2 (absent in melanoma 2), and NLRP1 (NOD-like receptor protein 1)-related to lung adenocarcinoma pyroptosis were selected. The correlation analysis of immune cell infiltration showed that CASP1, NLRP3, and AIM2, which showed that pyroptosis was involved in the infiltration of immune cells in the tumor microenvironment and NLRP1 exhibited high diagnostic efficacy, while PYCARD demonstrated poor diagnostic efficacy. High expression of CASP1, NLRP3, and NLRP1 correlated with a better prognosis (p < 0.05), while elevated AIM2 expression was associated with a poor prognosis (p < 0.05). However, PYCARD exhibited no significant correlation with prognosis (p > 0.05). The immunohistochemistry results showed that positive rates of CASP1, NLRP3, AIM2, and NLRP1 were 20%, 15%, 70%, and 10%, respectively, while in adjacent tissues, the positive rates were 60%, 60%, 20%, and70%, indicating high expression of AIM2 and low expression of CASP1, NLRP3, and NLRP1 in lung adenocarcinoma. Conclusion: CASP1, NLRP3, AIM2, and NLRP1 are core pyroptotic genes in lung adenocarcinoma and exhibit a strong correlation with immune cell infiltration, diagnosis, and prognosis of this condition. These genes may be useful in the clinical diagnosis and treatment of patients with lung adenocarcinoma.
Collapse
Affiliation(s)
- Lingling Zhu
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yongqian Zhang
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaojing Chen
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yuehang Li
- Department of Respiratory and Critical Care Medicine Ward 1, Handan Central Hospital, Handan, Hebei, China
| | - Haiqiao Pan
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Graduate School, Chengde Medical University, Chengde, Hebei, China
| | - Yuan Wang
- Department of Respiratory, Hebei Chest Hospital, Shijiazhuang, Hebei, China
| | - Ning Chen
- Department of Pathology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yajing Wu
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yishuai Li
- Department of Thoracic Surgery, Hebei Chest Hospital, Shijiazhuang, Hebei, China
| | - Min Zhao
- Department of Oncology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
38
|
van der Meer D, Hindley G, Shadrin AA, Smeland OB, Parker N, Dale AM, Frei O, Andreassen OA. Mapping the Genetic Landscape of Psychiatric Disorders With the MiXeR Toolset. Biol Psychiatry 2025:S0006-3223(25)00984-9. [PMID: 39983952 DOI: 10.1016/j.biopsych.2025.02.886] [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: 06/30/2024] [Revised: 01/29/2025] [Accepted: 02/11/2025] [Indexed: 02/23/2025]
Abstract
Psychiatric disorders have complex genetic architectures with substantial genetic overlap across conditions, which may partially explain their high levels of comorbidity. This presents significant challenges to research. Genome-wide association studies (GWASs) have uncovered hundreds of loci associated with single disorders, but the genetic landscape of psychiatric disorders has remained largely obscure. Moving beyond the conventional infinitesimal model, uni-, bi-, and trivariate MiXeR tools, applied to GWAS summary statistics, has enabled us to more comprehensively describe the genetic architecture of complex disorders and traits and their overlap. Furthermore, the GSA-MiXeR tool improves biological interpretation of GWAS findings to better elucidate causal mechanisms. Here, we outline the methodology that underlies the MiXeR tools together with instructions for their optimal use. We review results from studies that have investigated the genetic architecture of psychiatric disorders and their overlap using the MiXeR toolset. These studies have revealed generally high polygenicity and low discoverability among psychiatric disorders, particularly in contrast to somatic disorders. There is also pervasive genetic overlap across psychiatric disorders and behavioral traits, while their overlap with somatic traits is smaller, consistent with differences in polygenicity. Finally, GSA-MiXeR has quantified the contribution of gene sets to the heritability of psychiatric disorders, prioritizing small, biologically coherent gene sets. Together, these findings have implications for our understanding of the complex relationships between psychiatric disorders and related traits. MiXeR tools have provided new insights into the genetic architecture of psychiatric disorders, generating a better understanding of their underlying biological mechanisms and potential for clinical utility.
Collapse
Affiliation(s)
- Dennis van der Meer
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Guy Hindley
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Psychosis Studies, Institute of Psychiatry, Psychology and Neurosciences, King's College London, London, United Kingdom
| | - Alexey A Shadrin
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway; KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| | - Olav B Smeland
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nadine Parker
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anders M Dale
- Department of Radiology, School of Medicine, University of California, San Diego, La Jolla, California; Department of Psychiatry, University of California, San Diego, La Jolla, California; Department of Neurosciences, University of California, San Diego, La Jolla, California; Department of Cognitive Science, University of California, San Diego, La Jolla, California; Multimodal Imaging Laboratory, University of California, San Diego, La Jolla, California
| | - Oleksandr Frei
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Center for Bioinformatics, Department of Informatics, University of Oslo, Blindern, Oslo, Norway
| | - Ole A Andreassen
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway; KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway.
| |
Collapse
|
39
|
Lee S, Miller CL, Bentley AR, Brown MR, Nagarajan P, Noordam R, Morrison J, Schwander K, Westerman K, Kho M, Kraja AT, de Vries PS, Ammous F, Aschard H, Bartz TM, Do A, Dupont CT, Feitosa MF, Gudmundsdottir V, Guo X, Harris SE, Hikino K, Huang Z, Lefevre C, Lyytikäinen LP, Milaneschi Y, Nardone GG, Santin A, Schmidt H, Shen B, Sofer T, Sun Q, Tan YA, Tang J, Thériault S, van der Most PJ, Ware EB, Weiss S, Ya Xing W, Yu C, Zhao W, Ansari MAY, Anugu P, Attia JR, Bazzano LA, Bis JC, Breyer M, Cade B, Chen G, Collins S, Corley J, Davies G, Dörr M, Du J, Edwards TL, Faquih T, Faul JD, Fohner AE, Fretts AM, Gangireddy S, Gepner A, Graff M, Hofer E, Homuth G, Hood MM, Jie X, Kähönen M, Kardia SL, Karvonen-Gutierrez CA, Launer LJ, Levy D, Maheshwari M, Martin LW, Matsuda K, McNeil JJ, Nolte IM, Okochi T, Raffield LM, Raitakari OT, Risch L, Risch M, Roux AD, Ruiz-Narvaez EA, Russ TC, Saito T, Schreiner PJ, Scott RJ, Shikany J, Smith JA, Snieder H, Spedicati B, Tai ES, Taylor AM, Taylor KD, Tesolin P, van Dam RM, Wang R, Wenbin W, Xie T, Yao J, et alLee S, Miller CL, Bentley AR, Brown MR, Nagarajan P, Noordam R, Morrison J, Schwander K, Westerman K, Kho M, Kraja AT, de Vries PS, Ammous F, Aschard H, Bartz TM, Do A, Dupont CT, Feitosa MF, Gudmundsdottir V, Guo X, Harris SE, Hikino K, Huang Z, Lefevre C, Lyytikäinen LP, Milaneschi Y, Nardone GG, Santin A, Schmidt H, Shen B, Sofer T, Sun Q, Tan YA, Tang J, Thériault S, van der Most PJ, Ware EB, Weiss S, Ya Xing W, Yu C, Zhao W, Ansari MAY, Anugu P, Attia JR, Bazzano LA, Bis JC, Breyer M, Cade B, Chen G, Collins S, Corley J, Davies G, Dörr M, Du J, Edwards TL, Faquih T, Faul JD, Fohner AE, Fretts AM, Gangireddy S, Gepner A, Graff M, Hofer E, Homuth G, Hood MM, Jie X, Kähönen M, Kardia SL, Karvonen-Gutierrez CA, Launer LJ, Levy D, Maheshwari M, Martin LW, Matsuda K, McNeil JJ, Nolte IM, Okochi T, Raffield LM, Raitakari OT, Risch L, Risch M, Roux AD, Ruiz-Narvaez EA, Russ TC, Saito T, Schreiner PJ, Scott RJ, Shikany J, Smith JA, Snieder H, Spedicati B, Tai ES, Taylor AM, Taylor KD, Tesolin P, van Dam RM, Wang R, Wenbin W, Xie T, Yao J, Young KL, Zhang R, Zonderman AB, Concas MP, Conen D, Cox SR, Evans MK, Fox ER, de Las Fuentes L, Giri A, Girotto G, Grabe HJ, Gu C, Gudnason V, Harlow SD, Holliday E, Jost JB, Lacaze P, Lee S, Lehtimäki T, Li C, Liu CT, Morrison AC, North KE, Penninx BW, Peyser PA, Province MM, Psaty BM, Redline S, Rosendaal FR, Rotimi CN, Rotter JI, Schmidt R, Sim X, Terao C, Weir DR, Zhu X, Franceschini N, O'Connell JR, Jaquish CE, Wang H, Manning A, Munroe PB, Rao DC, Chen H, Gauderman WJ, Bierut L, Winkler TW, Fornage M. A Large-Scale Genome-wide Association Study of Blood Pressure Accounting for Gene-Depressive Symptomatology Interactions in 564,680 Individuals from Diverse Populations. RESEARCH SQUARE 2025:rs.3.rs-6025759. [PMID: 40034430 PMCID: PMC11875294 DOI: 10.21203/rs.3.rs-6025759/v1] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Background Gene-environment interactions may enhance our understanding of hypertension. Our previous study highlighted the importance of considering psychosocial factors in gene discovery for blood pressure (BP) but was limited in statistical power and population diversity. To address these challenges, we conducted a multi-population genome-wide association study (GWAS) of BP accounting for gene-depressive symptomatology (DEPR) interactions in a larger and more diverse sample. Results Our study included 564,680 adults aged 18 years or older from 67 cohorts and 4 population backgrounds (African (5%), Asian (7%), European (85%), and Hispanic (3%)). We discovered seven novel gene-DEPR interaction loci for BP traits. These loci mapped to genes implicated in neurogenesis (TGFA, CASP3), lipid metabolism (ACSL1), neuronal apoptosis (CASP3), and synaptic activity (CNTN6, DBI). We also identified evidence for gene-DEPR interaction at nine known BP loci, further suggesting links between mood disturbance and BP regulation. Of the 16 identified loci, 11 loci were derived from African, Asian, or Hispanic populations. Post-GWAS analyses prioritized 36 genes, including genes involved in synaptic functions (DOCK4, MAGI2) and neuronal signaling (CCK, UGDH, SLC01A2). Integrative druggability analyses identified 11 druggable candidate gene targets, including genes implicated in pathways linked to mood disorders as well as gene products targeted by known antihypertensive drugs. Conclusions Our findings emphasize the importance of considering gene-DEPR interactions on BP, particularly in non-European populations. Our prioritized genes and druggable targets highlight biological pathways connecting mood disorders and hypertension and suggest opportunities for BP drug repurposing and risk factor prevention, especially in individuals with DEPR.
Collapse
Affiliation(s)
- Songmi Lee
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX
| | - Clint L Miller
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA
| | - Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Michael R Brown
- Human Genetics Center, Department of Epidemiology, The University of Texas Health Science Center at Houston School of Public Health, Houston, TX
| | - Pavithra Nagarajan
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden
| | - John Morrison
- Division of Biostatistics, Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA
| | - Karen Schwander
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO
| | - Kenneth Westerman
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA
| | - Minjung Kho
- Graduate School of Data Science, Seoul National University, Seoul
| | - Aldi T Kraja
- University of Mississippi Medical Center, Jackson, MS
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, The University of Texas Health Science Center at Houston School of Public Health, Houston, TX
| | - Farah Ammous
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI
| | - Hughes Aschard
- Department of Computational Biology, F-75015 Paris, France Institut Pasteur, Université Paris Cité, Paris
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
| | - Anh Do
- Center for Biostatistics and Data Science, Institute for Informatics, Data Science, and Biostatistics, Washington University in St. Louis, School of Medicine, St. Louis, MO
| | - Charles T Dupont
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO
| | | | - 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
| | - Sarah E Harris
- Lothian Birth Cohorts, Department of Psychology, The University of Edinburgh, Edinburgh
| | - Keiko Hikino
- Laboratory for Pharmacogenomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa
| | - Zhijie Huang
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| | - Christophe Lefevre
- Department of Data Sciences, Hunter Medical Research Institute, New Lambton Heights, NSW
| | - Leo-Pekka Lyytikäinen
- Finnish Cardiovascular Research Center - Tampere, Department of Clinical Chemistry, Fimlab Laboratories and Faculty of Medicine and Health Technology, Tampere University, Tampere
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam UMC/Vrije universiteit, Amsterdam
| | | | - Aurora Santin
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste
| | - Helena Schmidt
- Department of Molecular Biology and Biochemistry, Medical University Graz, Graz, Styria
| | - Botong Shen
- Laboratory of Epidemiology and Population Sciences, Health Disparities Research Section, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Quan Sun
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Ye An Tan
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Jingxian Tang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Sébastien Thériault
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Quebec City, QC
| | - Peter J van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen
| | - Erin B Ware
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI
| | - Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald
| | - Wang Ya Xing
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Sciences Key Laboratory, Beijing, Beijing
| | - Chenglong Yu
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC
| | - Wei Zhao
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI
| | - Md Abu Yusuf Ansari
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS
| | - Pramod Anugu
- Jackson Heart Study, University of Mississippi Medical Center, Jackson, MS
| | - John R Attia
- School of Medicine and Public Health, College of Health Medicine and Wellbeing, University of Newcastle, New Lambton Heights, NSW
| | - Lydia A Bazzano
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
| | - Max Breyer
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Brian Cade
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Guanjie Chen
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Stacey Collins
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI
| | - Janie Corley
- Lothian Birth Cohorts, Department of Psychology, The University of Edinburgh, Edinburgh
| | - Gail Davies
- Lothian Birth Cohorts, Department of Psychology, The University of Edinburgh, Edinburgh
| | - Marcus Dörr
- German Center for Cardiovascular Research (DZHK), partner site Greifswald, Greifswald
| | - Jiawen Du
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Tariq Faquih
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI
| | - Alison E Fohner
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
| | - Amanda M Fretts
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA
| | - Srushti Gangireddy
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN
| | - Adam Gepner
- Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - MariaElisa Graff
- Cardiovascular Disease (CVD) Genetic Epidemiology Laboratory, Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Edith Hofer
- Department of Neurology, Medical University Graz, Graz, Styria
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald
| | - Michelle M Hood
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Xu Jie
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology and Visual Sciences Key Laboratory, Beijing, Beijing
| | - Mika Kähönen
- Finnish Cardiovascular Research Center - Tampere, Department of Clinical Physiology, Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere University, Tampere
| | - Sharon Lr Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
| | | | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Daniel Levy
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | | | - Lisa W Martin
- Department of Cardiology, George Washington University, Washington, DC
| | - Koichi Matsuda
- Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo
| | - John J McNeil
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC
| | - Ilja M Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen
| | - Tomo Okochi
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Aichi
| | - Laura M Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, NC
| | - Olli T Raitakari
- Centre for Population Health Research, Department of Clinical Physiology and Nuclear Medicine, InFLAMES Research Flagship, Turku University Hospital and University of Turku, Turku
| | - Lorenz Risch
- Faculty of Medical Sciences , Institute for Laboratory Medicine, Private University in the Principality of Liechtenstein, Vaduz
| | - Martin Risch
- Central Laboratory, Cantonal Hospital Graubünden, Chur
| | - Ana Diez Roux
- Urban Health Collaborative, Department of Epidemiology and Biostatistics, Drexel University, Philadelphia, PA
| | | | - Tom C Russ
- Lothian Birth Cohorts, Department of Psychology, The University of Edinburgh, Edinburgh
| | - Takeo Saito
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Aichi
| | - Pamela J Schreiner
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Rodney J Scott
- School of Medicine and Public Health, College of Health Medicine and Wellbeing, University of Newcastle, New Lambton Heights, NSW
| | - James Shikany
- Division of General Internal Medicine and Population Science, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Jennifer A Smith
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen
| | - Beatrice Spedicati
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste
| | - E Shyong Tai
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Adele M Taylor
- Lothian Birth Cohorts, Department of Psychology, The University of Edinburgh, Edinburgh
| | - 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
| | - Paola Tesolin
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste
| | - Rob M van Dam
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Rujia Wang
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen
| | - Wei Wenbin
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, Beijing
| | - Tian Xie
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Kristin L Young
- Cardiovascular Disease (CVD) Genetic Epidemiology Laboratory, Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Ruiyuan Zhang
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, Health Disparities Research Section, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Maria Pina Concas
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste
| | - David Conen
- Population Health Research Institute, Department of Medicine, McMaster University, Hamilton, ON
| | - Simon R Cox
- Lothian Birth Cohorts, Department of Psychology, The University of Edinburgh, Edinburgh
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, Health Disparities Research Section, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Ervin R Fox
- Jackson Heart Study, University of Mississippi Medical Center, Jackson, MS
| | - Lisa de Las Fuentes
- Center for Biostatistics and Data Science, Institute for Informatics, Data Science, and Biostatistics, Washington University in St. Louis, School of Medicine, St. Louis, MO
| | - Ayush Giri
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Giorgia Girotto
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Mecklenburg-Western Pomerania
| | - Charles Gu
- Center for Biostatistics and Data Science, Institute for Informatics, Data Science, and Biostatistics, Washington University in St. Louis, School of Medicine, St. Louis, MO
| | | | - Sioban D Harlow
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Elizabeth Holliday
- School of Medicine and Public Health, College of Health Medicine and Wellbeing, University of Newcastle, New Lambton Heights, NSW
| | - Jonas B Jost
- Rothschild Foundation Hospital, Institut Français de Myopie, Paris
| | - Paul Lacaze
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC
| | - Seunggeun Lee
- Graduate School of Data Science, Seoul National University, Seoul
| | - Terho Lehtimäki
- Finnish Cardiovascular Research Center - Tampere, Department of Clinical Chemistry, Fimlab Laboratories and Faculty of Medicine and Health Technology, Tampere University, Tampere
| | - Changwei Li
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, The University of Texas Health Science Center at Houston School of Public Health, Houston, TX
| | - Kari E North
- Cardiovascular Disease (CVD) Genetic Epidemiology Laboratory, Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | - Patricia A Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Michael M Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden
| | - Charles N Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - 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
| | | | - Xueling Sim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Chikashi Terao
- The Clinical Research Center at Shizuoka General Hospital, Shizuoka
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI
| | - Xiaofeng Zhu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Jeffrey R O'Connell
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD
| | - Cashell E Jaquish
- Division of Cardiovascular Science, Epidemiology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Heming Wang
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Alisa Manning
- Metabolism Program, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Patricia B Munroe
- Clinical Pharmacology and Precision Medicine, Queen Mary University of London, London
| | - Dabeeru C Rao
- Center for Biostatistics and Data Science, Institute for Informatics, Data Science, and Biostatistics, Washington University in St. Louis, School of Medicine, St. Louis, MO
| | - Han Chen
- Human Genetics Center, Department of Epidemiology, The University of Texas Health Science Center at Houston School of Public Health, Houston, TX
| | - W James Gauderman
- Division of Biostatistics, Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA
| | - Laura Bierut
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO
| | - Thomas W Winkler
- Department of Genetic Epidemiology, University of Regensburg, Regensburg
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX
| |
Collapse
|
40
|
R Muralitharan R, Zheng T, Dinakis E, Xie L, Barbaro-Wahl A, Jama HA, Nakai M, Paterson M, Leung KC, McArdle Z, Mirabito Colafella K, Johnson C, Qin W, Salimova E, Bitto NJ, Kaparakis-Liaskos M, Kaye DM, O'Donnell JA, Mackay CR, Marques FZ. Gut Microbiota Metabolites Sensed by Host GPR41/43 Protect Against Hypertension. Circ Res 2025; 136:e20-e33. [PMID: 39840468 DOI: 10.1161/circresaha.124.325770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND Fermentation of dietary fiber by the gut microbiota leads to the production of metabolites called short-chain fatty acids, which lower blood pressure and exert cardioprotective effects. Short-chain fatty acids activate host signaling responses via the functionally redundant receptors GPR41 (G-protein-coupled receptor 41) and GPR43 (G-protein-coupled receptor 43), which are highly expressed by immune cells. Whether and how these receptors protect against hypertension or mediate the cardioprotective effects of dietary fiber remains unknown. METHODS Cardiovascular phenotype was assessed in untreated and Ang II (angiotensin II) treated hypertensive wild-type and GPR41/43 knockout (KO) double knockout male mice fed diets with different levels of fiber content. Some mice received TLR4 (toll-like receptor 4)-antagonist treatment and bone marrow chimeras. SNPs (single-nucleotide polymorphisms) associated with GPR41 and GPR43 expression were assessed in UK Biobank participants. RESULTS Untreated GPR41/43KO mice had unaltered blood pressure but had greater cardiac and renal collagen deposition with higher macrophage numbers in the kidney compared with wild-type mice. Ang II-treated GPR41/43KO mice showed higher systolic blood pressure, cardiorenal weights and collagen deposition, and increased gut permeability, which allows the translocation of gastrointestinal bacterial components such as lipopolysaccharides into the circulation. The use of an antagonist to the lipopolysaccharide receptor, TLR4, a potent proinflammatory signaling molecule, restored the cardiovascular phenotype in GPR41/43KO mice. The lack of GPR41/43 expression in the immune compartment was sufficient to lead to a worsened hypertensive phenotype. We also demonstrate that GPR41/43 is, at least partially, responsible for the blood pressure-lowering and cardioprotective effects of a high-fiber diet. Finally, using the UK Biobank, we provide translational evidence that variants associated with lower expression of both GPR41 and GPR43 are more prevalent in participants with hypertension. CONCLUSIONS Our findings highlight that lack of short-chain fatty acid-receptor signaling via both GPR41 and GPR43 increases risk of high blood pressure, suggesting treatments that target these receptors could be a novel strategy to prevent or treat hypertension.
Collapse
Affiliation(s)
- Rikeish R Muralitharan
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Tenghao Zheng
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Evany Dinakis
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Liang Xie
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
- Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia (L.X., C.R.M.)
- Now with Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore (L.X.)
| | - Anastasia Barbaro-Wahl
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Hamdi A Jama
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Michael Nakai
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Madeleine Paterson
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Kwan Charmaine Leung
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Zoe McArdle
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Australia (Z.M., K.M.C.)
| | - Katrina Mirabito Colafella
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, Australia (Z.M., K.M.C.)
| | - Chad Johnson
- Bioimaging Platform, La Trobe University, Melbourne, Australia (C.J.)
| | - Wendy Qin
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Ekaterina Salimova
- Monash Bioimaging Facility, Monash University, Melbourne, Australia (E.S.)
| | - Natalie J Bitto
- Department of Microbiology, Anatomy, Physiology and Pharmacology (N.J.B., M. K-L.), La Trobe University, Melbourne, Australia
- La Trobe Research Centre for Extracellular Vesicles (N.J.B., M. K-L.), La Trobe University, Melbourne, Australia
| | - Maria Kaparakis-Liaskos
- Department of Microbiology, Anatomy, Physiology and Pharmacology (N.J.B., M. K-L.), La Trobe University, Melbourne, Australia
- La Trobe Research Centre for Extracellular Vesicles (N.J.B., M. K-L.), La Trobe University, Melbourne, Australia
- Now with Department of Microbiology & Immunology, University of Melbourne, Australia (M. K-L.)
| | - David M Kaye
- Central Clinical School, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, Australia (D.M.K.)
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia (D.M.K., F.Z.M.)
- Department of Cardiology, Alfred Hospital, Melbourne, Australia (D.M.K.)
| | - Joanne A O'Donnell
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
| | - Charles R Mackay
- Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Australia (L.X., C.R.M.)
- School of Pharmaceutical Sciences, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China (C.R.M.)
| | - Francine Z Marques
- Hypertension Research Laboratory, Victorian Heart Institute and Department of Pharmacology, Biomedical Discovery Institute, Faculty of Medicine, Nursing, and Health Sciences, Monash University, Melbourne, Australia (R.R.M., T.Z., E.D., L.X., A.B.-W., H.A.J., M.N., M.P., K.C.L., W.Q., J.A.O.D., F.Z.M.)
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia (D.M.K., F.Z.M.)
| |
Collapse
|
41
|
Wan J, van Ouwerkerk A, Mouren JC, Heredia C, Pradel L, Ballester B, Andrau JC, Spicuglia S. Comprehensive mapping of genetic variation at Epromoters reveals pleiotropic association with multiple disease traits. Nucleic Acids Res 2025; 53:gkae1270. [PMID: 39727170 PMCID: PMC11879118 DOI: 10.1093/nar/gkae1270] [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: 06/03/2024] [Revised: 10/28/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024] Open
Abstract
There is growing evidence that a wide range of human diseases and physiological traits are influenced by genetic variation of cis-regulatory elements. We and others have shown that a subset of promoter elements, termed Epromoters, also function as enhancer regulators of distal genes. This opens a paradigm in the study of regulatory variants, as single nucleotide polymorphisms (SNPs) within Epromoters might influence the expression of several (distal) genes at the same time, which could disentangle the identification of disease-associated genes. Here, we built a comprehensive resource of human Epromoters using newly generated and publicly available high-throughput reporter assays. We showed that Epromoters display intrinsic and epigenetic features that distinguish them from typical promoters. By integrating Genome-Wide Association Studies (GWAS), expression Quantitative Trait Loci (eQTLs) and 3D chromatin interactions, we found that regulatory variants at Epromoters are concurrently associated with more disease and physiological traits, as compared with typical promoters. To dissect the regulatory impact of Epromoter variants, we evaluated their impact on regulatory activity by analyzing allelic-specific high-throughput reporter assays and provided reliable examples of pleiotropic Epromoters. In summary, our study represents a comprehensive resource of regulatory variants supporting the pleiotropic role of Epromoters.
Collapse
Affiliation(s)
- Jing Wan
- Aix-Marseille University, INSERM, TAGC, UMR 1090 Marseille, France
- Equipe Labellisée LIGUE, 2023 Marseille, France
| | - Antoinette van Ouwerkerk
- Aix-Marseille University, INSERM, TAGC, UMR 1090 Marseille, France
- Equipe Labellisée LIGUE, 2023 Marseille, France
| | | | - Carla Heredia
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, UMR 5535, Montpellier, France
| | - Lydie Pradel
- Aix-Marseille University, INSERM, TAGC, UMR 1090 Marseille, France
- Equipe Labellisée LIGUE, 2023 Marseille, France
| | - Benoit Ballester
- Aix-Marseille University, INSERM, TAGC, UMR 1090 Marseille, France
| | - Jean-Christophe Andrau
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, UMR 5535, Montpellier, France
| | - Salvatore Spicuglia
- Aix-Marseille University, INSERM, TAGC, UMR 1090 Marseille, France
- Equipe Labellisée LIGUE, 2023 Marseille, France
| |
Collapse
|
42
|
Zhao Y, Fu Z, Barnett EJ, Wang N, Zhang K, Gao X, Zheng X, Tian J, Zhang H, Ding X, Li S, Li S, Cao Q, Chang S, Wang Y, Faraone SV, Yang L. Genome data based deep learning identified new genes predicting pharmacological treatment response of attention deficit hyperactivity disorder. Transl Psychiatry 2025; 15:46. [PMID: 39920114 PMCID: PMC11806042 DOI: 10.1038/s41398-025-03250-5] [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: 03/17/2024] [Revised: 12/06/2024] [Accepted: 01/17/2025] [Indexed: 02/09/2025] Open
Abstract
Although the efficacy of pharmacy in the treatment of attention deficit/hyperactivity disorder (ADHD) has been well established, the lack of predictors of treatment response poses great challenges for personalized treatment. The current study employed a comprehensive approach, combining genome-wide association analyses (GWAS) and deep learning (DL) methods, to elucidate the genetic underpinnings of pharmacological treatment response in ADHD. Based on genotype data of medication-naïve patients with ADHD who received pharmacological treatments for 12 weeks, the current study performed GWAS using the percentage changes in ADHD-RS score as phenotype. Then, DL models were constructed to predict percentage changes in symptom scores using genetic variants selected based on four different genome-wide P thresholds (E-02, E-03, E-04, E-05) as inputs. The current GWAS results identified two significant loci (rs10880574, P = 2.39E-09; rs2000900, P = 3.31E-09) which implicated two genes, TMEM117 and MYO5B, that were primarily associated with both brain- and gut-related disorders. The convolutional neural network (CNN) model, using variants with genome-wide P values less than E-02 (5516 SNPs), demonstrated the best performance with mean squared error (MSE) equals 0.012 (Accuracy = 0.83; Sensitivity = 0.90; Specificity = 0.75) in the validation dataset, 0.081 in an independent test dataset (Acc = 0.61, Sensitivity = 0.81; Specificity = 0.26). Notably, the variant that contributed most to the CNN model was NKAIN2, an ADHD-related gene, which is also associated with metabolic processes. To conclude, the integration of GWAS and DL methods revealed new genes contribute to ADHD pharmacological treatment responses, and underscored the interplay between neural systems and metabolic processes, potentially providing critical insights into precision treatment. Furthermore, our CNN model exhibited good performance in an independent dataset, encouraged future studies and implied potential clinical applications.
Collapse
Affiliation(s)
- Yilu Zhao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Clinical Research Center for Mental Disorders, (Peking University S+ixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Beijing, China
| | - Zhao Fu
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Clinical Research Center for Mental Disorders, (Peking University S+ixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Beijing, China
| | - Eric J Barnett
- Departments of Psychiatry and of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Ning Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Clinical Research Center for Mental Disorders, (Peking University S+ixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Beijing, China
| | - Kangfuxi Zhang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Clinical Research Center for Mental Disorders, (Peking University S+ixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Beijing, China
| | - Xuping Gao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Clinical Research Center for Mental Disorders, (Peking University S+ixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Beijing, China
| | - Xiangyu Zheng
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Clinical Research Center for Mental Disorders, (Peking University S+ixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Beijing, China
| | - Junbin Tian
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Clinical Research Center for Mental Disorders, (Peking University S+ixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Beijing, China
| | - Hui Zhang
- School of Engineering Medicine, Beihang University, Beijing, China
| | - XueTong Ding
- School of Engineering Medicine, Beihang University, Beijing, China
| | - Shaoxian Li
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Shuyu Li
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Qingjiu Cao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Clinical Research Center for Mental Disorders, (Peking University S+ixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Beijing, China
| | - Suhua Chang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Clinical Research Center for Mental Disorders, (Peking University S+ixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Beijing, China
| | - Yufeng Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Clinical Research Center for Mental Disorders, (Peking University S+ixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Beijing, China
| | - Stephen V Faraone
- Departments of Psychiatry and of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, USA.
| | - Li Yang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Clinical Research Center for Mental Disorders, (Peking University S+ixth Hospital), NHC Key Laboratory of Mental Health (Peking University), Beijing, China.
| |
Collapse
|
43
|
Chen Y, Bajpai AK, Li N, Xiang J, Wang A, Gu Q, Ruan J, Zhang R, Chen G, Lu L. Discovery of Novel Pain Regulators Through Integration of Cross-Species High-Throughput Data. CNS Neurosci Ther 2025; 31:e70255. [PMID: 39924344 PMCID: PMC11807727 DOI: 10.1111/cns.70255] [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: 11/19/2024] [Revised: 01/15/2025] [Accepted: 01/22/2025] [Indexed: 02/11/2025] Open
Abstract
AIMS Chronic pain is an impeding condition that affects day-to-day life and poses a substantial economic burden, surpassing many other health conditions. This study employs a cross-species integrated approach to uncover novel pain mediators/regulators. METHODS We used weighted gene coexpression network analysis to identify pain-enriched gene module. Functional analysis and protein-protein interaction (PPI) network analysis of the module genes were conducted. RNA sequencing compared pain model and control mice. PheWAS was performed to link genes to pain-related GWAS traits. Finally, candidates were prioritized based on node degree, differential expression, GWAS associations, and phenotype correlations. RESULTS A gene module significantly over-enriched with the pain reference set was identified (referred to as "pain module"). Analysis revealed 141 pain module genes interacting with 46 pain reference genes in the PPI network, which included 88 differentially expressed genes. PheWAS analysis linked 53 of these genes to pain-related GWAS traits. Expression correlation analysis identified Vdac1, Add2, Syt2, and Syt4 as significantly correlated with pain phenotypes across eight brain regions. NCAM1, VAMP2, SYT2, ADD2, and KCND3 were identified as top pain response/regulator genes. CONCLUSION The identified genes and molecular mechanisms may enhance understanding of pain pathways and contribute to better drug target identification.
Collapse
Affiliation(s)
- Ying Chen
- Department of Histology and Embryology, Medical CollegeNantong UniversityNantongJiangsuChina
| | - Akhilesh K. Bajpai
- Department of Genetics, Genomics and InformaticsUniversity of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Nan Li
- Department of Histology and Embryology, Medical CollegeNantong UniversityNantongJiangsuChina
| | - Jiahui Xiang
- Medical CollegeNantong UniversityNantongJiangsuChina
| | - Angelina Wang
- Department of Genetics, Genomics and InformaticsUniversity of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Qingqing Gu
- Department of Genetics, Genomics and InformaticsUniversity of Tennessee Health Science CenterMemphisTennesseeUSA
- Department of CardiologyAffiliated Hospital of Nantong UniversityJiangsuChina
| | - Junpu Ruan
- Medical CollegeNantong UniversityNantongJiangsuChina
| | - Ran Zhang
- Medical CollegeNantong UniversityNantongJiangsuChina
| | - Gang Chen
- Department of Histology and Embryology, Medical CollegeNantong UniversityNantongJiangsuChina
- Department of AnesthesiologyAffiliated Hospital of Nantong UniversityJiangsu ProvinceChina
| | - Lu Lu
- Department of Genetics, Genomics and InformaticsUniversity of Tennessee Health Science CenterMemphisTennesseeUSA
| |
Collapse
|
44
|
Li X, Chen H, Selvaraj MS, Van Buren E, Zhou H, Wang Y, Sun R, McCaw ZR, Yu Z, Jiang MZ, DiCorpo D, Gaynor SM, Dey R, Arnett DK, Benjamin EJ, Bis JC, Blangero J, Boerwinkle E, Bowden DW, Brody JA, Cade BE, Carson AP, Carlson JC, Chami N, Chen YDI, Curran JE, de Vries PS, Fornage M, Franceschini N, Freedman BI, Gu C, Heard-Costa NL, He J, Hou L, Hung YJ, Irvin MR, Kaplan RC, Kardia SLR, Kelly TN, Konigsberg I, Kooperberg C, Kral BG, Li C, Li Y, Lin H, Liu CT, Loos RJF, Mahaney MC, Martin LW, Mathias RA, Mitchell BD, Montasser ME, Morrison AC, Naseri T, North KE, Palmer ND, Peyser PA, Psaty BM, Redline S, Reiner AP, Rich SS, Sitlani CM, Smith JA, Taylor KD, Tiwari HK, Vasan RS, Viali S, Wang Z, Wessel J, Yanek LR, Yu B, Dupuis J, Meigs JB, Auer PL, Raffield LM, Manning AK, Rice KM, Rotter JI, Peloso GM, Natarajan P, Li Z, Liu Z, Lin X. A statistical framework for multi-trait rare variant analysis in large-scale whole-genome sequencing studies. NATURE COMPUTATIONAL SCIENCE 2025; 5:125-143. [PMID: 39920506 PMCID: PMC11981678 DOI: 10.1038/s43588-024-00764-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 12/20/2024] [Indexed: 02/09/2025]
Abstract
Large-scale whole-genome sequencing (WGS) studies have improved our understanding of the contributions of coding and noncoding rare variants to complex human traits. Leveraging association effect sizes across multiple traits in WGS rare variant association analysis can improve statistical power over single-trait analysis, and also detect pleiotropic genes and regions. Existing multi-trait methods have limited ability to perform rare variant analysis of large-scale WGS data. We propose MultiSTAAR, a statistical framework and computationally scalable analytical pipeline for functionally informed multi-trait rare variant analysis in large-scale WGS studies. MultiSTAAR accounts for relatedness, population structure and correlation among phenotypes by jointly analyzing multiple traits, and further empowers rare variant association analysis by incorporating multiple functional annotations. We applied MultiSTAAR to jointly analyze three lipid traits in 61,838 multi-ethnic samples from the Trans-Omics for Precision Medicine (TOPMed) Program. We discovered and replicated new associations with lipid traits missed by single-trait analysis.
Collapse
Grants
- U01 DK085524 NIDDK NIH HHS
- HHSN268201800001I U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R01 DK078616 NIDDK NIH HHS
- U01 HL054472 NHLBI NIH HHS
- R01 HL071025 NHLBI NIH HHS
- UL1 RR033176 NCRR NIH HHS
- R01 HL112064 NHLBI NIH HHS
- K26 DK138425 NIDDK NIH HHS
- 75N92020D00002 NHLBI NIH HHS
- R01 HL113323 NHLBI NIH HHS
- U01-HG012064 U.S. Department of Health & Human Services | NIH | National Human Genome Research Institute (NHGRI)
- N01-HC-95160 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R01-HL071251 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R35 CA197449 NCI NIH HHS
- 75N92020D00005 NHLBI NIH HHS
- R01 HL104135 NHLBI NIH HHS
- HHSN268201600002C NHLBI NIH HHS
- N01HC95160 NHLBI NIH HHS
- R01-DK117445 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R01 HL071251 NHLBI NIH HHS
- R01 HL120393 NHLBI NIH HHS
- R01 HL087698 NHLBI NIH HHS
- R01 HL046380 NHLBI NIH HHS
- R01 HL071259 NHLBI NIH HHS
- N01-HC-95163 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- U19 CA203654 NCI NIH HHS
- N01HC95163 NHLBI NIH HHS
- R01-HL071259 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- UL1 TR001079 NCATS NIH HHS
- R01 HL175681 NHLBI NIH HHS
- U01 HG012064 NHGRI NIH HHS
- N01-HC-95169 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R01 HL087660 NHLBI NIH HHS
- DK063491 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R01 AR048797 NIAMS NIH HHS
- R01-HL071205 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R01 HL092577 NHLBI NIH HHS
- N01-HC-95166 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- N01HC95169 NHLBI NIH HHS
- U01 HL054509 NHLBI NIH HHS
- 75N92020D00001 NHLBI NIH HHS
- U01 HL120393 NHLBI NIH HHS
- R01 HL113338 NHLBI NIH HHS
- R01 DK117445 NIDDK NIH HHS
- R01 HL153805 NHLBI NIH HHS
- R01 AG058921 NIA NIH HHS
- R01 HL071250 NHLBI NIH HHS
- R01-HL104135-04S1 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- UL1-TR-000040 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- N01-HC-95162 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- UL1-TR001881 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R01 NS058700 NINDS NIH HHS
- R01 HL127564 NHLBI NIH HHS
- R01 HL076784 NHLBI NIH HHS
- N01-HC-95167 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- N01HC95164 NHLBI NIH HHS
- R01-HL113338 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL163972 NHLBI NIH HHS
- HHSN268201600004C NHLBI NIH HHS
- HHSN268201700005I NHLBI NIH HHS
- R03-HL154284 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01-HL142711 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 75N92020D00003 NHLBI NIH HHS
- F32 HL085989 NHLBI NIH HHS
- R01 MH078111 NIMH NIH HHS
- N01HC95162 NHLBI NIH HHS
- U01 HL054464 NHLBI NIH HHS
- R01 HL119443 NHLBI NIH HHS
- R01 HL105756 NHLBI NIH HHS
- N01HC95168 NHLBI NIH HHS
- NHLBI TOPMed Fellowship 75N92021F00229 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HHSN268201500003I NHLBI NIH HHS
- HHSN268201700004I NHLBI NIH HHS
- R01-HL071051 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R01 HL067348 NHLBI NIH HHS
- 1R01AG086379-01 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R01 HL142711 NHLBI NIH HHS
- R35 HL135818 NHLBI NIH HHS
- R01-HL071250 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R35-CA197449 U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
- U01 HL072524 NHLBI NIH HHS
- DK078616 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- P30 DK063491 NIDDK NIH HHS
- R01 HL071051 NHLBI NIH HHS
- N01-HC-95161 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- U01 HL054457 NHLBI NIH HHS
- N01HC95165 NHLBI NIH HHS
- N01HC95159 NHLBI NIH HHS
- M01 RR000052 NCRR NIH HHS
- HHSN268201700003I NHLBI NIH HHS
- N01HC95161 NHLBI NIH HHS
- UL1 TR001420 NCATS NIH HHS
- R01 HL049762 NHLBI NIH HHS
- HL046389 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P01 HL045522 NHLBI NIH HHS
- U01-HG009088 U.S. Department of Health & Human Services | NIH | National Human Genome Research Institute (NHGRI)
- 75N92020D00004 NHLBI NIH HHS
- R00 HG012956 NHGRI NIH HHS
- 75N92020D00007 NHLBI NIH HHS
- U01 HL072518 NHLBI NIH HHS
- U19-CA203654 U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
- U01 DK078616 NIDDK NIH HHS
- N01-HC-95168 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- HHSN268201700001I NHLBI NIH HHS
- 1R35-HL135818 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U01 HL137162 NHLBI NIH HHS
- M01 RR007122 NCRR NIH HHS
- R01 HL059684 NHLBI NIH HHS
- U54 HG013247 NHGRI NIH HHS
- HHSN268201600018C NHLBI NIH HHS
- R01 AG086379 NIA NIH HHS
- R01 MH078143 NIMH NIH HHS
- R01 DK071891 NIDDK NIH HHS
- N01HC95167 NHLBI NIH HHS
- R01 HG013163 NHGRI NIH HHS
- N01HC25195 NHLBI NIH HHS
- R01-MD012765 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R01 HL071205 NHLBI NIH HHS
- U01 HL054481 NHLBI NIH HHS
- 75N92019D00031 NHLBI NIH HHS
- R03 HL154284 NHLBI NIH HHS
- R01 MD012765 NIMHD NIH HHS
- R00HG012956-02 U.S. Department of Health & Human Services | NIH | National Human Genome Research Institute (NHGRI)
- UL1 TR000040 NCATS NIH HHS
- HL105756 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U01-HL054472 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HHSN268201700002I NHLBI NIH HHS
- R01 HL151855 NHLBI NIH HHS
- U01 HG009088 NHGRI NIH HHS
- UM1 DK078616 NIDDK NIH HHS
- R01 MH083824 NIMH NIH HHS
- R01 HL117626 NHLBI NIH HHS
- N01-HC-95159 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- 75N92020D00006 NHLBI NIH HHS
- HHSN268201600001C NHLBI NIH HHS
- N01HC95166 NHLBI NIH HHS
- U01-HL054473 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- N01-HC-95164 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R01 AG028321 NIA NIH HHS
- U01-HL054509 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- UL1-TR-001420 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- U01-HL054495 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U01-HL137162 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01-HL071258 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- HHSN268201600003C NHLBI NIH HHS
- UL1-TR-001079 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- UL1 TR001881 NCATS NIH HHS
- UL1-RR033176 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- N01-HC-95165 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- U01 HL054495 NHLBI NIH HHS
- R01 HL071258 NHLBI NIH HHS
- R01-HL153805 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL055673 NHLBI NIH HHS
- R01-HL055673-18S1 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL092301 NHLBI NIH HHS
- U01 HL054473 NHLBI NIH HHS
- HL151855 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01-HL127564 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U01-HL072524 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
Collapse
Affiliation(s)
- Xihao Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Han Chen
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Margaret Sunitha Selvaraj
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Eric Van Buren
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Hufeng Zhou
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Yuxuan Wang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Ryan Sun
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zachary R McCaw
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zhi Yu
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Clinical and Translational Epidemiology Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Min-Zhi Jiang
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biostatistics, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Daniel DiCorpo
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Sheila M Gaynor
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Rounak Dey
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Donna K Arnett
- Provost Office, University of South Carolina, Columbia, SC, USA
| | - Emelia J Benjamin
- Section of Cardiovascular Medicine, Boston Medical Center, Boston University Chobanian & 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
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - John Blangero
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Donald W Bowden
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Brian E Cade
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - April P Carson
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jenna C Carlson
- Department of Human Genetics and Department of Biostatistics and Health Data Science, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nathalie Chami
- The 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
| | - 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
| | - Joanne E Curran
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Myriam Fornage
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Barry I Freedman
- Department of Internal Medicine, Nephrology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Charles Gu
- Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Nancy L Heard-Costa
- Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
- Translational Science Institute, Tulane University, New Orleans, LA, USA
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern University, Chicago, IL, USA
| | - Yi-Jen Hung
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Marguerite R Irvin
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Tanika N Kelly
- Department of Medicine, Division of Nephrology, University of Illinois Chicago, Chicago, IL, USA
| | - Iain Konigsberg
- Department of Biomedical Informatics, University of Colorado, Aurora, CO, USA
| | - Charles Kooperberg
- Department of Medicine, Division of Nephrology, University of Illinois Chicago, Chicago, IL, USA
| | - Brian G Kral
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Changwei Li
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
- Translational Science Institute, Tulane University, New Orleans, LA, USA
| | - Yun Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Honghuang Lin
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Ruth J F Loos
- The 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, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael C Mahaney
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Lisa W Martin
- School of Medicine and Health Sciences, George Washington University, Washington, DC, USA
| | - Rasika A Mathias
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Braxton D Mitchell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - May E Montasser
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Take Naseri
- Naseri & Associates Public Health Consultancy Firm and Family Health Clinic, Apia, Samoa
- Department of Epidemiology, Brown University, Providence, RI, USA
| | - Kari E North
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nicholette D Palmer
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Patricia A Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Departments of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Alexander P Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Departments of Epidemiology, University of Washington, Seattle, WA, USA
| | - Stephen S Rich
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
| | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 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
| | - Hemant K Tiwari
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ramachandran S Vasan
- Framingham Heart Study, Framingham, MA, USA
- Department of Quantitative and Qualitative Health Sciences, UT Health San Antonio School of Public Health, San Antonia, TX, USA
| | - Satupa'itea Viali
- School of Medicine, National University of Samoa, Apia, Samoa
- Department of Chronic Disease Epidemiology, Yale University School of Public Health, New Haven, CT, USA
- Oceania University of Medicine, Apia, Samoa
| | - Zhe Wang
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jennifer Wessel
- Department of Epidemiology, Fairbanks School of Public Health, Indiana University, Indianapolis, IN, USA
- Diabetes Translational Research Center, Indiana University, Indianapolis, IN, USA
| | - Lisa R Yanek
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bing Yu
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, QC, Canada
| | - James B Meigs
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Paul L Auer
- Division of Biostatistics, Data Science Institute, and Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alisa K Manning
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Metabolism Program, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA, USA
| | - Kenneth M Rice
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Gina M Peloso
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Pradeep Natarajan
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Zilin Li
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Zhonghua Liu
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA.
| | - Xihong Lin
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Department of Statistics, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
45
|
Schipper M, de Leeuw CA, Maciel BAPC, Wightman DP, Hubers N, Boomsma DI, O'Donovan MC, Posthuma D. Prioritizing effector genes at trait-associated loci using multimodal evidence. Nat Genet 2025; 57:323-333. [PMID: 39930082 DOI: 10.1038/s41588-025-02084-7] [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: 01/29/2024] [Accepted: 01/08/2025] [Indexed: 02/14/2025]
Abstract
Genome-wide association studies (GWAS) yield large numbers of genetic loci associated with traits and diseases. Predicting the effector genes that mediate these locus-trait associations remains challenging. Here we present the FLAMES (fine-mapped locus assessment model of effector genes) framework, which predicts the most likely effector gene in a locus. FLAMES creates machine learning predictions from biological data linking single-nucleotide polymorphisms to genes, and then evaluates these scores together with gene-centric evidence of convergence of the GWAS signal in functional networks. We benchmark FLAMES on gene-locus pairs derived by expert curation, rare variant implication and domain knowledge of molecular traits. We demonstrate that combining single-nucleotide-polymorphism-based and convergence-based modalities outperforms prioritization strategies using a single line of evidence. Applying FLAMES, we resolve the FSHB locus in the GWAS for dizygotic twinning and further leverage this framework to find schizophrenia risk genes that converge with rare coding evidence and are relevant in different stages of life.
Collapse
Affiliation(s)
- Marijn Schipper
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| | - Christiaan A de Leeuw
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Bernardo A P C Maciel
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Douglas P Wightman
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Nikki Hubers
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Netherlands Twin Register, Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development (AR&D) research institute, Amsterdam, The Netherlands
| | - Dorret I Boomsma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development (AR&D) research institute, Amsterdam, The Netherlands
| | - Michael C O'Donovan
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine & Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Danielle Posthuma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, 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
| |
Collapse
|
46
|
Degner KN, Bell JL, Jones SD, Won H. Just a SNP away: The future of in vivo massively parallel reporter assay. CELL INSIGHT 2025; 4:100214. [PMID: 39618480 PMCID: PMC11607654 DOI: 10.1016/j.cellin.2024.100214] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/03/2024] [Accepted: 10/06/2024] [Indexed: 04/03/2025]
Abstract
The human genome is largely noncoding, yet the field is still grasping to understand how noncoding variants impact transcription and contribute to disease etiology. The massively parallel reporter assay (MPRA) has been employed to characterize the function of noncoding variants at unprecedented scales, but its application has been largely limited by the in vitro context. The field will benefit from establishing a systemic platform to study noncoding variant function across multiple tissue types under physiologically relevant conditions. However, to date, MPRA has been applied to only a handful of in vivo conditions. Given the complexity of the central nervous system and its widespread interactions with all other organ systems, our understanding of neuropsychiatric disorder-associated noncoding variants would be greatly advanced by studying their functional impact in the intact brain. In this review, we discuss the importance, technical considerations, and future applications of implementing MPRA in the in vivo space with the focus on neuropsychiatric disorders.
Collapse
Affiliation(s)
- Katherine N. Degner
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jessica L. Bell
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sean D. Jones
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hyejung Won
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
47
|
Greenwood E, Cao M, Lee CM, Liu A, Moyo B, Bao G, Gibson G. Haplotype rather than single causal variants effects contribute to regulatory gene expression associations in human myeloid cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635675. [PMID: 39975189 PMCID: PMC11838257 DOI: 10.1101/2025.01.30.635675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Genome-wide association studies typically identify hundreds to thousands of loci, many of which harbor multiple independent peaks, each parsimoniously assumed to be due to the activity of a single causal variant. Fine-mapping of such variants has become a priority and since most associations are located within regulatory regions, it is also assumed that they colocalize with regulatory variants that influence the expression of nearby genes. Here we examine these assumptions by using a moderate throughput expression CROPseq protocol in which Cas9 nuclease is used to induce small insertions and deletions across the credible set of SNPs that may account for expression quantitative trait loci (eQTL) for genes associated with inflammatory bowel disease (IBD). Of the 4,384 SNPs targeted in 88 loci (an average of 50 per locus), 439 were significant and further examined for validation. From these, 98 significantly altered target gene expression in HL-60 myeloid cell line, 74 in induced macrophages from these HL-60 cells, and 78 in induced neutrophils for a total of 201 validated effects (46%), 43 of which were observed in at least two of the cell types. Considering the observed sensitivity and specificity of the controls, we estimate that there are at least 150 true positives per cell type, an average of almost 2.4 for each of the 64 eQTL for which putative causal variants have been fine-mapped. This implies that haplotype effects are likely to explain many of the associations. We also demonstrate that the same approach can be used to investigate the activity of very rare variants in regulatory regions for 89 genes, providing a rapid strategy for establishing clinical relevance of non-coding mutations.
Collapse
Affiliation(s)
- Emily Greenwood
- School of Biological Sciences, Georgia Institute of Technology, Atlanta GA 30332, USA
| | - Mingming Cao
- Department of Bioengineering, Rice University, Houston TX 77005, USA
| | - Ciaran M. Lee
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Aidi Liu
- Department of Bioengineering, Rice University, Houston TX 77005, USA
| | - Buhle Moyo
- Department of Bioengineering, Rice University, Houston TX 77005, USA
| | - Gang Bao
- Department of Bioengineering, Rice University, Houston TX 77005, USA
| | - Greg Gibson
- School of Biological Sciences, Georgia Institute of Technology, Atlanta GA 30332, USA
| |
Collapse
|
48
|
Pan Q, Tao Y, Cai T, Veluchamy A, Hebert HL, Zhu P, Haque M, Dottorini T, Colvin LA, Smith BH, Meng W. A genome-wide association study identifies genetic variants associated with hip pain in the UK Biobank cohort (N = 221,127). Sci Rep 2025; 15:2812. [PMID: 39843573 PMCID: PMC11754597 DOI: 10.1038/s41598-025-85871-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025] Open
Abstract
Hip pain is a common musculoskeletal complaint that leads many people to seek medical attention. We conducted a primary genome-wide association study (GWAS) on the hip pain phenotype within the UK Biobank cohort. Sex-stratified GWAS analysis approach was also performed to explore sex specific variants associated with hip pain. We found seven different loci associated with hip pain at GWAS significance level, with the most significant single nucleotide polymorphism (SNP) being rs77641763 within the EXD3 (p value = 2.20 × 10-13). We utilized summary statistics from the FinnGen cohort and a previous GWAS meta-analysis on hip osteoarthritis as replication cohorts. Four loci (rs509345, rs73581564, rs9597759, rs2018384) were replicated with a p value less than 0.05. Sex-stratified GWAS analyses revealed a unique locus within the CUL1 gene (rs4726995, p = 2.56 × 10-9) in males, and three unique loci in females: rs1651359966 on chromosome 7 (p = 1.15 × 10-8), rs552965738 on chromosome 9 (p = 2.72 × 10-8), and rs1978969 on chromosome 13 (p = 2.87 × 10-9). This study has identified seven genetic loci associated with hip pain. Sex-stratified analysis also revealed sex specific variants associated with hip pain in males and females. This study has provided a foundation for advancing research of hip pain and hip osteoarthritis.
Collapse
Affiliation(s)
- Qi Pan
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, 315100, China
| | - Yiwen Tao
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, 315100, China
| | - Tengda Cai
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, 315100, China
| | - Abi Veluchamy
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD2 4BF, UK
| | - Harry L Hebert
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD2 4BF, UK
| | - Peixi Zhu
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang, China
| | - Mainul Haque
- School of Mathematical Sciences, University of Nottingham Ningbo China, Ningbo, 315100, China
| | - Tania Dottorini
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, LE12 5RD, UK
| | - Lesley A Colvin
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD2 4BF, UK
| | - Blair H Smith
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD2 4BF, UK
| | - Weihua Meng
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo, 315100, China.
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD2 4BF, UK.
- Center for Public Health, Faculty of Medicine, Health and Life Sciences, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, BT12 6BA, UK.
| |
Collapse
|
49
|
Vashisht S, Parisi C, Winata CL. Computational analysis of congenital heart disease associated SNPs: unveiling their impact on the gene regulatory system. BMC Genomics 2025; 26:55. [PMID: 39838281 PMCID: PMC11749323 DOI: 10.1186/s12864-025-11232-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 01/09/2025] [Indexed: 01/23/2025] Open
Abstract
Congenital heart disease (CHD) is a prevalent condition characterized by defective heart development, causing premature death and stillbirths among infants. Genome-wide association studies (GWASs) have provided insights into the role of genetic variants in CHD pathogenesis through the identification of a comprehensive set of single-nucleotide polymorphisms (SNPs). Notably, 90-95% of these variants reside in the noncoding genome, complicating the understanding of their underlying mechanisms. Here, we developed a systematic computational pipeline for the identification and analysis of CHD-associated SNPs spanning both coding and noncoding regions of the genome. Initially, we curated a thorough dataset of SNPs from GWAS-catalog and ClinVar database and filtered them based on CHD-related traits. Subsequently, these CHD-SNPs were annotated and categorized into noncoding and coding regions based on their location. To study the functional implications of noncoding CHD-SNPs, we cross-validated them with enhancer-specific histone modification marks from developing human heart across 9 Carnegie stages and identified potential cardiac enhancers. This approach led to the identification of 2,056 CHD-associated putative enhancers (CHD-enhancers), 38.9% of them overlapping with known enhancers catalogued in human enhancer disease database. We identified heart-related transcription factor binding sites within these CHD-enhancers, offering insights into the impact of SNPs on TF binding. Conservation analysis further revealed that many of these CHD-enhancers were highly conserved across vertebrates, suggesting their evolutionary significance. Utilizing heart-specific expression quantitative trait loci data, we further identified a subset of 63 CHD-SNPs with regulatory potential distributed across various cardiac tissues. Concurrently, coding CHD-SNPs were represented as a protein interaction network and its subsequent binding energy analysis focused on a pair of proteins within this network, pinpointed a deleterious coding CHD-SNP, rs770030288, located in C2 domain of MYBPC3 protein. Overall, our findings demonstrate that SNPs have the potential to disrupt gene regulatory systems, either by affecting enhancer sequences or modulating protein-protein interactions, which can lead to abnormal developmental processes contributing to CHD pathogenesis.
Collapse
Affiliation(s)
- Shikha Vashisht
- International Institute of Molecular and Cell Biology in Warsaw, Laboratory of Zebrafish Developmental Genomics, Księcia Trojdena 4, Warsaw, 02-109, Poland
| | - Costantino Parisi
- International Institute of Molecular and Cell Biology in Warsaw, Laboratory of Zebrafish Developmental Genomics, Księcia Trojdena 4, Warsaw, 02-109, Poland
| | - Cecilia L Winata
- International Institute of Molecular and Cell Biology in Warsaw, Laboratory of Zebrafish Developmental Genomics, Księcia Trojdena 4, Warsaw, 02-109, Poland.
| |
Collapse
|
50
|
Kun E, Sohail M, Narasimhan VM. The trait-specific timing of accelerated genomic change in the human lineage. CELL GENOMICS 2025; 5:100740. [PMID: 39788103 PMCID: PMC11770217 DOI: 10.1016/j.xgen.2024.100740] [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: 05/01/2024] [Revised: 10/04/2024] [Accepted: 12/14/2024] [Indexed: 01/12/2025]
Abstract
Humans exhibit distinct characteristics compared to our primate and ancient hominin ancestors. To investigate genomic bursts in the evolution of these traits, we use two complementary approaches to examine enrichment among genome-wide association study loci spanning diseases and AI-based image-derived brain, heart, and skeletal tissue phenotypes with genomic regions reflecting four evolutionary divergence points. These regions cover epigenetic differences among humans and rhesus macaques, human accelerated regions (HARs), ancient selective sweeps, and Neanderthal-introgressed alleles. Skeletal traits such as pelvic width and limb proportions showed enrichment in evolutionary annotations that mirror morphological changes in the primate fossil record. Additionally, we observe enrichment of loci associated with the longitudinal fasciculus in human-gained epigenetic elements since macaques, the visual cortex in HARs, and the thalamus proper in Neanderthal-introgressed alleles, implying that associated cognitive functions such as language processing, decision-making, sensory signaling, and motor control are enriched at different evolutionary depths.
Collapse
Affiliation(s)
- Eucharist Kun
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
| | - Mashaal Sohail
- Centro de Ciencias Genómicas (CCG), Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Mexico.
| | - Vagheesh M Narasimhan
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA; Department of Statistics and Data Science, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|