1
|
Mugo JW, Day C, Choudhury A, Deetlefs M, Freercks R, Geraty S, Panieri A, Cotchobos C, Ribeiro M, Engelbrecht A, Micklesfield LK, Ramsay M, Pedretti S, Peter J. A GWAS of angiotensin-converting enzyme inhibitor-induced angioedema in a South African population. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2025; 4:100464. [PMID: 40290521 PMCID: PMC12022653 DOI: 10.1016/j.jacig.2025.100464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/15/2025] [Accepted: 02/02/2025] [Indexed: 04/30/2025]
Abstract
Background Angiotensin-converting enzyme inhibitor-induced angioedema (AE-ACEI) is a life-threatening adverse event; globally, it is the most common cause of emergency presentations with angioedema. Several genome-wide association studies (GWASs) have found genomic associations with AE-ACEI. However, despite African Americans having a 5-fold increased risk of AE-ACEI, there are no published GWASs from Africa. Objective The aim of this study was to conduct a GWAS of AE-ACEI in a South African population and perform a meta-analysis with an African American and European American population. Methods The GWAS included 202 South African adults with a history of AE-ACEI and 513 controls without angioedema following angiotensin-converting enzyme inhibitor (ACEI) treatment for at least 2 years. A meta-analysis was conducted with GWAS summary statistics from an African American and European American cohort (from the Vanderbilt-Marshfield cohort, which consisted of 174 case patients and 489 controls). Results No single-nucleotide polymorphisms (SNPs) attained genome-wide significance; however, 26 SNPs in the postimputation standard GWAS of the South African cohort and 73 SNPs in the meta-analysis attained suggestive thresholds (P < 5.0 × 10-06). Some of these SNPs were found to be located close to the genes PRKCQ (protein kinase C theta), RAD51B (RAD51 Paralog B), and RIMS1 (regulating synaptic membrane exocytosis 1), which were previously linked with drug-induced angioedema, and also close to the CSMD1 (CUB and sushi multiple domains 1) gene, which has been linked to ACEI cough, providing replication at the gene level but with novel lead SNPs. The study also replicated SNP rs500766 on chromosome 10, which was previously found to be associated with AE-ACEI. Conclusions Our results highlight the importance of African populations for detection of novel variants in replication studies. Further increased sampling across the continent and matched functional work are needed to confirm the importance of genetic variation in understanding the biology of AE-ACEI.
Collapse
Affiliation(s)
- Jacquiline W. Mugo
- Division of Allergy and Clinical Immunology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Johannesburg, South Africa
| | - Cascia Day
- Division of Allergy and Clinical Immunology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Johannesburg, South Africa
- Allergy and Immunology Unit, University of Cape Town Lung Institute (Pty) Ltd, Johannesburg, South Africa
| | - Ananyo Choudhury
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Maria Deetlefs
- Division of Allergy and Clinical Immunology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Johannesburg, South Africa
| | - Robert Freercks
- Faculty of Health Sciences, Department of Medicine, Nelson Mandela University, Gqeberhal, South Africa
| | - Sian Geraty
- Faculty of Health Sciences, Department of Medicine, Nelson Mandela University, Gqeberhal, South Africa
| | - Angelica Panieri
- Faculty of Health Sciences, Department of Medicine, Nelson Mandela University, Gqeberhal, South Africa
| | - Christian Cotchobos
- Faculty of Health Sciences, Department of Medicine, Nelson Mandela University, Gqeberhal, South Africa
| | - Melissa Ribeiro
- Allergy and Immunology Unit, University of Cape Town Lung Institute (Pty) Ltd, Johannesburg, South Africa
| | | | - Lisa K. Micklesfield
- South African Medical Research Council/Wits Developmental Pathways for Health Research Unit, Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Michèle Ramsay
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sarah Pedretti
- Allergy and Immunology Unit, University of Cape Town Lung Institute (Pty) Ltd, Johannesburg, South Africa
| | - Jonny Peter
- Division of Allergy and Clinical Immunology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Johannesburg, South Africa
- Allergy and Immunology Unit, University of Cape Town Lung Institute (Pty) Ltd, Johannesburg, South Africa
| |
Collapse
|
2
|
Wang J, Wang Q, Wang D, Gao Y, Li S, Wang T, Liu H, Wang H, Hu X, Wan C. Blunted Niacin Skin Flushing Response in Schizophrenia: A Meta-analysis. Schizophr Bull 2025:sbaf069. [PMID: 40401804 DOI: 10.1093/schbul/sbaf069] [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] [Indexed: 05/23/2025]
Abstract
BACKGROUND AND HYPOTHESIS The multifactorial pathogenesis of schizophrenia (SZ) hinders the diagnosis and treatment of this disorder. Niacin skin flushing response (NSFR) has been identified as an endophenotype for SZ, but the proportion of blunted NSFR (BNR) varied between studies. This study aims to clarify the relationship between NSFR and SZ through a meta-analysis. STUDY DESIGN PubMed, Embase, Web of Science, Cochrane, and Scopus databases were searched for articles published until May 2024, and 32 studies were eligible. Using random-effects models, we examined the characteristics of NSFR in SZ, including the reaction degree, speed, sensitivity, and risk and prevalence of BNR. Subgroup analyses and regression analyses were performed to investigate the relevant effect factors of NSFR. STUDY RESULTS The reaction degree (SMD = -0.90; CI, -1.08 to -0.72), speed (SMD = 0.64; CI, 0.02-1.25), and sensitivity (SMD = 0.89; CI, 0.49-1.29) of NSFR was significantly reduced in SZ compared to healthy controls (HC). Moreover, we observed a positive association between BNR and SZ (OR = 8.50; CI, 5.93-12.19). The overall prevalence of BNR was 58.5% in SZ (CI, 49.3%-67.8%) compared to 11.8% in HC (CI, 7.7%-15.9%). In addition, NSFR detection method, geographical regions, and age were found to have effects on reaction degree and prevalence of BNR. CONCLUSIONS This study confirmed a significantly abnormal NSFR and higher prevalence of BNR in SZ, which highlights the potential facilitation of the diagnosis and personalized intervention of SZ subgroups. In addition, the study points to a need to establish a standardized method for NSFR assessment.
Collapse
Affiliation(s)
- Jinfeng Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Qian Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Dandan Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Gao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Shuhui Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Tianqi Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Hao Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Huiling Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiaowen Hu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Chunling Wan
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
3
|
Leventhal MJ, Zanella CA, Kang B, Peng J, Gritsch D, Liao Z, Bukhari H, Wang T, Pao PC, Danquah S, Benetatos J, Nehme R, Farhi S, Tsai LH, Dong X, Scherzer CR, Feany MB, Fraenkel E. An integrative systems-biology approach defines mechanisms of Alzheimer's disease neurodegeneration. Nat Commun 2025; 16:4441. [PMID: 40393985 PMCID: PMC12092734 DOI: 10.1038/s41467-025-59654-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 04/28/2025] [Indexed: 05/22/2025] Open
Abstract
Despite years of intense investigation, the mechanisms underlying neuronal death in Alzheimer's disease, remain incompletely understood. To define relevant pathways, we conducted an unbiased, genome-scale forward genetic screen for age-associated neurodegeneration in Drosophila. We also measured proteomics, phosphoproteomics, and metabolomics in Drosophila models of Alzheimer's disease and identified Alzheimer's genetic variants that modify gene expression in disease-vulnerable neurons in humans. We then used a network model to integrate these data with previously published Alzheimer's disease proteomics, lipidomics and genomics. Here, we computationally predict and experimentally confirm how HNRNPA2B1 and MEPCE enhance toxicity of the tau protein, a pathological feature of Alzheimer's disease. Furthermore, we demonstrated that the screen hits CSNK2A1 and NOTCH1 regulate DNA damage in Drosophila and human stem cell-derived neural progenitor cells. Our study identifies candidate pathways that could be targeted to ameliorate neurodegeneration in Alzheimer's disease.
Collapse
Affiliation(s)
- Matthew J Leventhal
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Camila A Zanella
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Byunguk Kang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jiajie Peng
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - David Gritsch
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Zhixiang Liao
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Hassan Bukhari
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Tao Wang
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Ping-Chieh Pao
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Serwah Danquah
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Joseph Benetatos
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ralda Nehme
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Samouil Farhi
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Li-Huei Tsai
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Xianjun Dong
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Clemens R Scherzer
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Stephen and Denise Adams Center of Yale School of Medicine, New Haven, CT, USA
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ernest Fraenkel
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, MA, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
4
|
Lehmann B, Bräuninger L, Cho Y, Falck F, Jayadeva S, Katell M, Nguyen T, Perini A, Tallman S, Mackintosh M, Silver M, Kuchenbäcker K, Leslie D, Chatterjee N, Holmes C. Methodological opportunities in genomic data analysis to advance health equity. Nat Rev Genet 2025:10.1038/s41576-025-00839-w. [PMID: 40369311 DOI: 10.1038/s41576-025-00839-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2025] [Indexed: 05/16/2025]
Abstract
The causes and consequences of inequities in genomic research and medicine are complex and widespread. However, it is widely acknowledged that underrepresentation of diverse populations in human genetics research risks exacerbating existing health disparities. Efforts to improve diversity are ongoing, but an often-overlooked source of inequity is the choice of analytical methods used to process, analyse and interpret genomic data. This choice can influence all areas of genomic research, from genome-wide association studies and polygenic score development to variant prioritization and functional genomics. New statistical and machine learning techniques to understand, quantify and correct for the impact of biases in genomic data are emerging within the wider genomic research and genomic medicine ecosystems. At this crucial time point, it is important to clarify where improvements in methods and practices can, or cannot, have a role in improving equity in genomics. Here, we review existing approaches to promote equity and fairness in statistical analysis for genomics, and propose future methodological developments that are likely to yield the most impact for equity.
Collapse
Affiliation(s)
- Brieuc Lehmann
- Department of Statistical Science, University College London, London, UK.
| | - Leandra Bräuninger
- Department of Statistical Science, University College London, London, UK
- The Alan Turing Institute, London, UK
| | - Yoonsu Cho
- Genomics England, London, UK
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Fabian Falck
- The Alan Turing Institute, London, UK
- Department of Statistics, University of Oxford, Oxford, UK
| | | | | | | | | | | | | | - Matt Silver
- Genomics England, London, UK
- Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Karoline Kuchenbäcker
- Genomics England, London, UK
- Division of Psychiatry, University College London, London, UK
| | | | - Nilanjan Chatterjee
- Department of Biostatistics, Johns Hopkins University, Baltimore, MD, USA
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Chris Holmes
- Department of Statistics, University of Oxford, Oxford, UK
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
5
|
Yuan L, Su Y, Zhao J, Cho M, Wang D, Yuan L, Li M, Zheng D, Piao H, Wang Y, Zhu Z, Li D, Wang T, Ha KT, Park W, Liu K. Investigating the shared genetic architecture between obesity and depression: a large-scale genomewide cross-trait analysis. Front Endocrinol (Lausanne) 2025; 16:1578944. [PMID: 40405979 PMCID: PMC12094978 DOI: 10.3389/fendo.2025.1578944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
Introduction Increasing evidence suggests that individuals with obesity are at a higher risk of developing depression, and conversely, depression can contribute to the onset of obesity, creating a detrimental cycle. This study aims to investigate the potential shared biological pathways between obesity and depression by examining genetic correlations, identifying common polymorphisms, and conducting cross-trait genetic analyses. Methods We assessed the genetic correlation between obesity and depression using linkage disequilibrium score regression and high-density lipoprotein levels. We combined two different sources of obesity data using METAL and employed bidirectional Mendelian randomization to determine the causal relationship between obesity and depression. Additionally, we conducted multivariate trait analysis using the MTAG method to improve statistical robustness and identify novel genetic associations. Furthermore, we performed a thorough investigation of independent risk loci using GCTA-COJO, PLACO, MAGMA, POPS, and SMR, integrating different QTL information and methods to further identify risk genes and proteins. Results Our analysis revealed genetic correlations and bidirectional positive causal relationships between obesity and depression, highlighting shared risk SNP (rs10789340). We identified RPL31P12, NEGR1, and DCC as common risk genes for obesity and depression. Using the BLISS method, we identified SCG3 and FLRT2 as potential drug targets. Limitation Most of our data sources are from Europe, which may limit the generalization of our findings to other ethnic populations. Conclusion This study demonstrates the genetic causal relationship and common risk SNPs, genes, proteins, and pathways between obesity and depression. These findings contribute to a deeper understanding of their pathogenesis and the identification of potential therapeutic targets.
Collapse
Affiliation(s)
- Lei Yuan
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Yale Su
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jiangqi Zhao
- Department of Dermatology, The Second Hospital of Jilin University, Changchun, China
| | - Minkyoung Cho
- Department of Parasitology and Tropical Medicine, and Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Duo Wang
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Long Yuan
- School of Acupuncture, Moxibustion and Tuina, The Third Clinical Medical School of Henan University of Chinese Medicine, Zhengzhou, China
| | - Mixia Li
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Dongdong Zheng
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Hulin Piao
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Yong Wang
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Zhicheng Zhu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Dan Li
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Tiance Wang
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Ki-Tae Ha
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, Republic of Korea
| | - Wonyoung Park
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, Republic of Korea
| | - Kexiang Liu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
6
|
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
|
7
|
Yang S, Zheng C, Xia C, Kang J, Gu L. Detection of positive selection on depression-associated genes. Heredity (Edinb) 2025; 134:263-272. [PMID: 40075226 PMCID: PMC12056014 DOI: 10.1038/s41437-025-00753-1] [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: 11/08/2024] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Although depression significantly impacts fitness, some hypotheses suggest that it may offer a survival benefit. However, there has been limited systematic investigation into the selection pressures acting on genes associated with depression at the genomic level. Here, we conducted comparative genomic analyses and computational molecular evolutionary analyses on 320 depression-associated genes at two levels, i.e., across the primate phylogeny (long timescale selection) and in modern human populations (recent selection). We identified seven genes under positive selection in the human lineage, and 46 genes under positive selection in modern human populations. Most positively selected variants in modern human populations were at UTR regions and non-coding exons, indicating the importance of gene expression regulation in the evolution of depression-associated genes. Positively selected genes are not only related to immune responses, but also function in reproduction and dietary adaptation. Notably, the proportion of depression-associated genes under positive selection was significantly higher than the positively selected genes at the genome-wide average level in African, East Asian, and South Asian populations. We also identified two positively selected loci that happened to be associated with depression in the South Asian population. Our study revealed that depression-associated genes are subject to varying selection pressures across different populations. We suggest that, in precision medicine-particularly in gene therapy-it is crucial to consider the specific functions of genes within distinct populations.
Collapse
Affiliation(s)
- Shiyu Yang
- Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, China
- The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510180, China
| | - Chenqing Zheng
- State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China
| | - Canwei Xia
- Ministry of Education Key Laboratory for Biodiversity and Ecological Engineering, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
| | - Jihui Kang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Langyu Gu
- State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, 510275, China.
| |
Collapse
|
8
|
Shuai Z, Jie MS, Wen XK, Xu H, Yuan L. Effects of exercise intervention on exercise capacity and cardiopulmonary function in patients with atrial fibrillation: A randomized controlled trial systematic review and meta-analysis. Med Clin (Barc) 2025; 164:106908. [PMID: 40220475 DOI: 10.1016/j.medcli.2025.106908] [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: 06/24/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 04/14/2025]
Abstract
BACKGROUND Atrial fibrillation (AF) is a common cardiac arrhythmia that significantly impacts the cardiopulmonary function and quality of life of patients. Despite various treatment strategies, non-pharmacological interventions, particularly exercise interventions, have gained attention in recent years. OBJECTIVE Through systematic review and meta-analysis, this study explores the impact of physical activity on the exercise capacity and quality of life of AF patients. It assesses the safety, clinical outcomes, and physiological mechanisms of exercise intervention in the treatment of AF. METHODS The systematic review and individual patient data (IPD) meta-analysis method were employed, following the PRISMA-IPD guidelines, for literature selection, data extraction, and quality assessment. The analysis focused on the impact of exercise on the cardiopulmonary function and quality of life of AF patients in randomized controlled trials. RESULTS A total of 12 randomized controlled trials involving 287 AF patients were included. Meta-analysis demonstrated a significant improvement in the 6-minute walk test capacity (SMD=87.87, 95% CI [42.23, 133.51]), static heart rate improvement (SMD=-7.63, 95% CI [-11.42, -3.85]), and cardiopulmonary function enhancement (SMD=2.37, 95% CI [0.96, 3.77]) due to exercise. There was also a significant improvement in the quality of life (SMD=0.720, 95% CI [0.038, 1.402]). CONCLUSION Exercise has a significant effect on improving exercise capacity and cardiopulmonary function in patients with atrial fibrillation. Particularly, high-intensity exercise training has a more significant impact on improving cardiopulmonary function and exercise capacity, emphasizing the importance of personalized exercise plans in enhancing the cardiopulmonary health of AF patients. Further research is needed to explore the effects of exercise on improving the quality of life in the future. PROSPERO ID CRD2023493917.
Collapse
Affiliation(s)
- Zhang Shuai
- Graduate Development, Harbin Sport University, Harbin, Heilongjiang, China
| | - Mao Su Jie
- Graduate Development, Harbin Sport University, Harbin, Heilongjiang, China
| | - Xiao Kai Wen
- Chinese Fencing Academy, Nanjing Sport Institute, Nanjing, Jiangsu, China
| | - Hong Xu
- Nanjing Polytechnic Institute Sports Work Department, Nanjing, Jiangsu, China.
| | - Lu Yuan
- Nanjing Polytechnic Institute Sports Work Department, Nanjing, Jiangsu, China
| |
Collapse
|
9
|
Hatzikotoulas K, Southam L, Stefansdottir L, Boer CG, McDonald ML, Pett JP, Park YC, Tuerlings M, Mulders R, Barysenka A, Arruda AL, Tragante V, Rocco A, Bittner N, Chen S, Horn S, Srinivasasainagendra V, To K, Katsoula G, Kreitmaier P, Tenghe AMM, Gilly A, Arbeeva L, Chen LG, de Pins AM, Dochtermann D, Henkel C, Höijer J, Ito S, Lind PA, Lukusa-Sawalena B, Minn AKK, Mola-Caminal M, Narita A, Nguyen C, Reimann E, Silberstein MD, Skogholt AH, Tiwari HK, Yau MS, Yue M, Zhao W, Zhou JJ, Alexiadis G, Banasik K, Brunak S, Campbell A, Cheung JTS, Dowsett J, Faquih T, Faul JD, Fei L, Fenstad AM, Funayama T, Gabrielsen ME, Gocho C, Gromov K, Hansen T, Hudjashov G, Ingvarsson T, Johnson JS, Jonsson H, Kakehi S, Karjalainen J, Kasbohm E, Lemmelä S, Lin K, Liu X, Loef M, Mangino M, McCartney D, Millwood IY, Richman J, Roberts MB, Ryan KA, Samartzis D, Shivakumar M, Skou ST, Sugimoto S, Suzuki K, Takuwa H, Teder-Laving M, Thomas L, Tomizuka K, Turman C, Weiss S, Wu TT, Zengini E, Zhang Y, Ferreira MAR, Babis G, Baras A, Barker T, Carey DJ, Cheah KSE, Chen Z, Cheung JPY, Daly M, de Mutsert R, Eaton CB, et alHatzikotoulas K, Southam L, Stefansdottir L, Boer CG, McDonald ML, Pett JP, Park YC, Tuerlings M, Mulders R, Barysenka A, Arruda AL, Tragante V, Rocco A, Bittner N, Chen S, Horn S, Srinivasasainagendra V, To K, Katsoula G, Kreitmaier P, Tenghe AMM, Gilly A, Arbeeva L, Chen LG, de Pins AM, Dochtermann D, Henkel C, Höijer J, Ito S, Lind PA, Lukusa-Sawalena B, Minn AKK, Mola-Caminal M, Narita A, Nguyen C, Reimann E, Silberstein MD, Skogholt AH, Tiwari HK, Yau MS, Yue M, Zhao W, Zhou JJ, Alexiadis G, Banasik K, Brunak S, Campbell A, Cheung JTS, Dowsett J, Faquih T, Faul JD, Fei L, Fenstad AM, Funayama T, Gabrielsen ME, Gocho C, Gromov K, Hansen T, Hudjashov G, Ingvarsson T, Johnson JS, Jonsson H, Kakehi S, Karjalainen J, Kasbohm E, Lemmelä S, Lin K, Liu X, Loef M, Mangino M, McCartney D, Millwood IY, Richman J, Roberts MB, Ryan KA, Samartzis D, Shivakumar M, Skou ST, Sugimoto S, Suzuki K, Takuwa H, Teder-Laving M, Thomas L, Tomizuka K, Turman C, Weiss S, Wu TT, Zengini E, Zhang Y, Ferreira MAR, Babis G, Baras A, Barker T, Carey DJ, Cheah KSE, Chen Z, Cheung JPY, Daly M, de Mutsert R, Eaton CB, Erikstrup C, Furnes ON, Golightly YM, Gudbjartsson DF, Hailer NP, Hayward C, Hochberg MC, Homuth G, Huckins LM, Hveem K, Ikegawa S, Ishijima M, Isomura M, Jones M, Kang JH, Kardia SLR, Kloppenburg M, Kraft P, Kumahashi N, Kuwata S, Lee MTM, Lee PH, Lerner R, Li L, Lietman SA, Lotta L, Lupton MK, Mägi R, Martin NG, McAlindon TE, Medland SE, Michaëlsson K, Mitchell BD, Mook-Kanamori DO, Morris AP, Nabika T, Nagami F, Nelson AE, Ostrowski SR, Palotie A, Pedersen OB, Rosendaal FR, Sakurai-Yageta M, Schmidt CO, Sham PC, Singh JA, Smelser DT, Smith JA, Song YQ, Sørensen E, Tamiya G, Tamura Y, Terao C, Thorleifsson G, Troelsen A, Tsezou A, Uchio Y, Uitterlinden AG, Ullum H, Valdes AM, van Heel DA, Walters RG, Weir DR, Wilkinson JM, Winsvold BS, Yamamoto M, Zwart JA, Stefansson K, Meulenbelt I, Teichmann SA, van Meurs JBJ, Styrkarsdottir U, Zeggini E. Translational genomics of osteoarthritis in 1,962,069 individuals. Nature 2025:10.1038/s41586-025-08771-z. [PMID: 40205036 DOI: 10.1038/s41586-025-08771-z] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 02/11/2025] [Indexed: 04/11/2025]
Abstract
Osteoarthritis is the third most rapidly growing health condition associated with disability, after dementia and diabetes1. By 2050, the total number of patients with osteoarthritis is estimated to reach 1 billion worldwide2. As no disease-modifying treatments exist for osteoarthritis, a better understanding of disease aetiopathology is urgently needed. Here we perform a genome-wide association study meta-analyses across up to 489,975 cases and 1,472,094 controls, establishing 962 independent associations, 513 of which have not been previously reported. Using single-cell multiomics data, we identify signal enrichment in embryonic skeletal development pathways. We integrate orthogonal lines of evidence, including transcriptome, proteome and epigenome profiles of primary joint tissues, and implicate 700 effector genes. Within these, we find rare coding-variant burden associations with effect sizes that are consistently higher than common frequency variant associations. We highlight eight biological processes in which we find convergent involvement of multiple effector genes, including the circadian clock, glial-cell-related processes and pathways with an established role in osteoarthritis (TGFβ, FGF, WNT, BMP and retinoic acid signalling, and extracellular matrix organization). We find that 10% of the effector genes express a protein that is the target of approved drugs, offering repurposing opportunities, which can accelerate translation.
Collapse
Affiliation(s)
- Konstantinos Hatzikotoulas
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Lorraine Southam
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | | | - Cindy G Boer
- Department of Internal Medicine, Erasmus MC Medical Center, Rotterdam, The Netherlands
| | - Merry-Lynn McDonald
- Birmingham Veterans Affairs Healthcare System (BVAHS), Birmingham, AL, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
- Department of Epidemiology, School of Public Health, UAB, Birmingham, AL, USA
- Department of Genetics, School of Medicine, UAB, Birmingham, AL, USA
| | - J Patrick Pett
- Wellcome Sanger Institute, Cellular Genetics Programme, Wellcome Genome Campus, Cambridge, UK
| | - Young-Chan Park
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Margo Tuerlings
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rick Mulders
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Andrei Barysenka
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Ana Luiza Arruda
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | | | - Alison Rocco
- Birmingham Veterans Affairs Healthcare System (BVAHS), Birmingham, AL, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, School of Medicine, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Norbert Bittner
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Shibo Chen
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Susanne Horn
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Vinodh Srinivasasainagendra
- Birmingham Veterans Affairs Healthcare System (BVAHS), Birmingham, AL, USA
- Department of Biostatistics, School of Public Health, UAB, Birmingham, AL, USA
| | - Ken To
- Wellcome Sanger Institute, Cellular Genetics Programme, Wellcome Genome Campus, Cambridge, UK
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Surgery, University of Cambridge, Cambridge, UK
| | - Georgia Katsoula
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Peter Kreitmaier
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Amabel M M Tenghe
- Centre for Genetic Epidemiology, Institute for Clinical Epidemiology and Applied Biometry, University of Tubingen, Tübingen, Germany
| | | | - Liubov Arbeeva
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lane G Chen
- Department of Psychiatry, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | | | - Daniel Dochtermann
- Birmingham Veterans Affairs Healthcare System (BVAHS), Birmingham, AL, USA
| | - Cecilie Henkel
- Clinical Orthopaedic Research Hvidovre (CORH), Department of Orthopaedic Surgery, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Jonas Höijer
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Shuji Ito
- Department of Orthopedic Surgery, Shimane University Faculty of Medicine, Izumo, Japan
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Penelope A Lind
- Psychiatric Genetics, Brain and Mental Health Research Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | | | - Aye Ko Ko Minn
- Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Marina Mola-Caminal
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Akira Narita
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Chelsea Nguyen
- Birmingham Veterans Affairs Healthcare System (BVAHS), Birmingham, AL, USA
| | - Ene Reimann
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Micah D Silberstein
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Anne-Heidi Skogholt
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Hemant K Tiwari
- Department of Biostatistics, School of Public Health, UAB, Birmingham, AL, USA
| | - Michelle S Yau
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew SeniorLife, Harvard Medical School, Boston, MA, USA
| | - Ming Yue
- School of Biomedical Sciences, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Wei Zhao
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Jin J Zhou
- Department of Biostatistics, UCLA Fielding School of Public Health, Los Angeles, CA, USA
| | - George Alexiadis
- 1st Department of Orthopaedics, KAT General Hospital, Athens, Greece
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, UK
| | | | - Joseph Dowsett
- Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Tariq Faquih
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Lijiang Fei
- Wellcome Sanger Institute, Cellular Genetics Programme, Wellcome Genome Campus, Cambridge, UK
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Anne Marie Fenstad
- The Norwegian Arthroplasty Register, Department of Orthopaedic Surgery, Haukeland University Hospital, Bergen, Norway
| | | | - Maiken E Gabrielsen
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Chinatsu Gocho
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Kirill Gromov
- Clinical Orthopaedic Research Hvidovre (CORH), Department of Orthopaedic Surgery, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Thomas Hansen
- Danish Headache Center, Department of Neurology, Copenhagen University Hospital, Rigshospitalet - Glostrup, Glostrup, Denmark
| | - Georgi Hudjashov
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Thorvaldur Ingvarsson
- Department of Orthopedic Surgery, Akureyri Hospital, Akureyri, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Jessica S Johnson
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Helgi Jonsson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
- Department of Medicine, Landspitali The National University Hospital of Iceland, Reykjavik, Iceland
| | - Saori Kakehi
- Department of Metabolism & Endocrinology, Sportology Center, Juntendo University Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Juha Karjalainen
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Elisa Kasbohm
- Institute for Community Medicine, SHIP-KEF, University Medicine Greifswald, Greifswald, Germany
| | - Susanna Lemmelä
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Kuang Lin
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Xiaoxi Liu
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Marieke Loef
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, UK
| | - Daniel McCartney
- Centre for Genomic and Experimental Medicine, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, UK
| | - Iona Y Millwood
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Joshua Richman
- Birmingham Veterans Affairs Healthcare System (BVAHS), Birmingham, AL, USA
- Department of Surgery, School of Medicine, UAB, Birmingham, AL, USA
| | - Mary B Roberts
- Center for Primary Care & Prevention, Brown University, Pawtucket, RI, USA
| | - Kathleen A Ryan
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dino Samartzis
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong SAR, China
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Manu Shivakumar
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Søren T Skou
- Research Unit for Musculoskeletal Function and Physiotherapy, Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
- The Research and Implementation Unit PROgrez, Department of Physiotherapy and Occupational Therapy, Næstved-Slagelse-Ringsted Hospitals, Slagelse, Denmark
| | - Sachiyo Sugimoto
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Ken Suzuki
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, University of Manchester, Manchester, UK
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hiroshi Takuwa
- Department of Orthopedic Surgery, Shimane University Faculty of Medicine, Izumo, Japan
| | - Maris Teder-Laving
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Laurent Thomas
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kohei Tomizuka
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Constance Turman
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Stefan Weiss
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Tian T Wu
- Department of Psychiatry, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Eleni Zengini
- 4th Psychiatric Department, Dromokaiteio Psychiatric Hospital, Haidari, Athens, Greece
| | - Yanfei Zhang
- Genomic Medicine Institute, Geisinger Health System, Danville, PA, USA
| | | | - George Babis
- 2nd Department of Orthopaedics, National and Kapodistrian University of Athens, Medical School, 'Konstantopouleio' Hospital, Athens, Greece
| | - Aris Baras
- Regeneron Genetics Center, Tarrytown, NY, USA
| | - Tyler Barker
- Sports Medicine Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Intermountain Healthcare, Precision Genomics, Salt Lake City, UT, USA
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA
| | - David J Carey
- Department of Genomic Health, Geisinger, Danville, PA, USA
| | - Kathryn S E Cheah
- School of Biomedical Sciences, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Zhengming Chen
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Jason Pui-Yin Cheung
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong SAR, China
| | - Mark Daly
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Charles B Eaton
- Center for Primary Care & Prevention, Brown University, Pawtucket, RI, USA
- Department of Family Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Department of Epidemiology, School of Public Health, Brown University, Providence, RI, USA
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Ove Nord Furnes
- The Norwegian Arthroplasty Register, Department of Orthopaedic Surgery, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Yvonne M Golightly
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- University of Nebraska Medical Center, Omaha, NE, USA
| | - Daniel F Gudbjartsson
- deCODE Genetics/Amgen, Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | - Nils P Hailer
- Orthopedics, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Caroline Hayward
- MRC Human Genetics Unit IGC, University of Edinburgh, Edinburgh, UK
| | - Marc C Hochberg
- Department of Epidemiology and Public Health, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Georg Homuth
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Laura M Huckins
- Department of Psychiatry, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Kristian Hveem
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Center, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Levanger, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Shiro Ikegawa
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Muneaki Ishijima
- Department of Medicine for Orthopaedics and Motor Organ, Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Minoru Isomura
- Faculty of Human Sciences, Shimane University, Matsue, Japan
| | | | - Jae H Kang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Margreet Kloppenburg
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Nobuyuki Kumahashi
- Department of Orthopedic Surgery, Matsue Red Cross Hospital, Matsue, Japan
| | - Suguru Kuwata
- Department of Orthopedic Surgery, Shimane University Faculty of Medicine, Izumo, Japan
| | | | - Phil H Lee
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, USA
| | - Robin Lerner
- Blizard Institute, Queen Mary University of London, London, UK
| | - Liming Li
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness & Response, Beijing, China
| | | | - Luca Lotta
- Regeneron Genetics Center, Tarrytown, NY, USA
| | - Michelle K Lupton
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
- Neurogenetics and Dementia, Brain and Mental Health Research Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Reedik Mägi
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Nicholas G Martin
- Genetic Epidemiology, Brain and Mental Health Research Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Timothy E McAlindon
- Division of Rheumatology, Allergy, & Immunology, Tufts Medical Center, Boston, MA, USA
| | - Sarah E Medland
- Psychiatric Genetics, Brain and Mental Health Research Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Psychology, University of Queensland, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Karl Michaëlsson
- Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Braxton D Mitchell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Andrew P Morris
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, University of Manchester, Manchester, UK
| | - Toru Nabika
- Department of Functional Pathology, Shimane University School of Medicine, Izumo, Japan
| | - Fuji Nagami
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Amanda E Nelson
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Ole Birger Pedersen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Immunology, Zealand University Hospital - Køge, Køge, Denmark
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Carsten Oliver Schmidt
- Institute for Community Medicine, SHIP-KEF, University Medicine Greifswald, Greifswald, Germany
| | - Pak Chung Sham
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Jasvinder A Singh
- Birmingham Veterans Affairs Healthcare System (BVAHS), Birmingham, AL, USA
- Department of Epidemiology, School of Public Health, UAB, Birmingham, AL, USA
- Division of Rheumatology and Clinical Immunology, Department of Medicine at the School of Medicine, UAB, Birmingham, AL, USA
- Medicine Service, Michale E. DeBakey VA Medical Center, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | | | - Jennifer A Smith
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - You-Qiang Song
- School of Biomedical Sciences, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Erik Sørensen
- Department of Clinical Immunology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Gen Tamiya
- Tohoku University Graduate School of Medicine, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Statistical Genetics Team, Center for Advanced Intelligence Project, RIKEN, Tokyo, Japan
| | - Yoshifumi Tamura
- Department of Metabolism & Endocrinology, Sportology Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan
- The Department of Applied Genetics, The School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | | | - Anders Troelsen
- Clinical Academic Group: Research OsteoArthritis Denmark (CAG ROAD), Department of Orthopaedic Surgery, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Aspasia Tsezou
- Laboratory of Cytogenetics and Molecular Genetics, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Yuji Uchio
- Department of Orthopedic Surgery, Shimane University Faculty of Medicine, Izumo, Japan
| | - A G Uitterlinden
- Department of Internal Medicine, Erasmus MC Medical Center, Rotterdam, The Netherlands
| | | | - Ana M Valdes
- Faculty of Medicine & Health Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | | | - Robin G Walters
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - J Mark Wilkinson
- School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
| | - Bendik S Winsvold
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital and University of Oslo, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Masayuki Yamamoto
- Tohoku University Graduate School of Medicine, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - John-Anker Zwart
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Research, Innovation and Education, Division of Clinical Neuroscience, Oslo University Hospital and University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kari Stefansson
- deCODE Genetics/Amgen, Reykjavik, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Ingrid Meulenbelt
- Department of Biomedical Data Sciences, Section Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sarah A Teichmann
- Department of Medicine & Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Joyce B J van Meurs
- Department of Internal Medicine, Erasmus MC Medical Center, Rotterdam, The Netherlands
| | | | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
- TUM School of Medicine and Health, Technical University of Munich and Klinikum Rechts der Isar, Munich, Germany.
| |
Collapse
|
10
|
Ji Y, Liu N, Yang Y, Wang M, Cheng J, Zhu W, Qiu S, Geng Z, Cui G, Yu Y, Liao W, Zhang H, Gao B, Xu X, Han T, Yao Z, Zhang Q, Qin W, Liu F, Liang M, Wang S, Xu Q, Xu J, Fu J, Zhang P, Li W, Shi D, Wang C, Lui S, Yan Z, Chen F, Zhang J, Shen W, Miao Y, Wang D, Gao JH, Zhang X, Xu K, Zuo XN, Zhang L, Ye Z, Li MJ, Xian J, Zhang B, Yu C. Cross-ancestry and sex-stratified genome-wide association analyses of amygdala and subnucleus volumes. Nat Genet 2025; 57:839-850. [PMID: 40097784 DOI: 10.1038/s41588-025-02136-y] [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: 11/14/2023] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
The amygdala is a small but critical multi-nucleus structure for emotion, cognition and neuropsychiatric disorders. Although genetic associations with amygdala volumetric traits have been investigated in sex-combined European populations, cross-ancestry and sex-stratified analyses are lacking. Here we conducted cross-ancestry and sex-stratified genome-wide association analyses for 21 amygdala volumetric traits in 6,923 Chinese and 48,634 European individuals. We identified 191 variant-trait associations (P < 2.38 × 10-9), including 47 new associations (12 new loci) in sex-combined univariate analyses and seven additional new loci in sex-combined and sex-stratified multivariate analyses. We identified 12 ancestry-specific and two sex-specific associations. The identified genetic variants include 16 fine-mapped causal variants and regulate amygdala and fetal brain gene expression. The variants were enriched for brain development and colocalized with mood, cognition and neuropsychiatric disorders. These results indicate that cross-ancestry and sex-stratified genetic association analyses may provide insight into the genetic architectures of amygdala and subnucleus volumes.
Collapse
Affiliation(s)
- Yuan Ji
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Nana Liu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yunjun Yang
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Meiyun Wang
- Department of Radiology, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou, China
- Biomedical Institute, Henan Academy of Sciences, Zhengzhou, China
| | - Jingliang Cheng
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenzhen Zhu
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shijun Qiu
- Department of Medical Imaging, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Zuojun Geng
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guangbin Cui
- Functional and Molecular Imaging Key Lab of Shaanxi Province & Department of Radiology, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Yongqiang Yu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Weihua Liao
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
- Molecular Imaging Research Center of Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hui Zhang
- Department of Radiology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Bo Gao
- Department of Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- Department of Radiology, Yantai Yuhuangding Hospital, Yantai, China
| | - Xiaojun Xu
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Tong Han
- Department of Radiology, Tianjin Huanhu Hospital, Tianjin, China
| | - Zhenwei Yao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Quan Zhang
- Department of Radiology, Characteristic Medical Center of Chinese People's Armed Police Force, Tianjin, China
| | - Wen Qin
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Feng Liu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Meng Liang
- School of Medical Imaging and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin, China
| | - Sijia Wang
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Qiang Xu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiayuan Xu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jilian Fu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Peng Zhang
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Wei Li
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Dapeng Shi
- Department of Radiology, Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou, China
| | - Caihong Wang
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Su Lui
- Department of Radiology, Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital of Sichuan University, Chengdu, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, China
| | - Zhihan Yan
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Feng Chen
- Department of Radiology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, China
| | - Jing Zhang
- Department of Magnetic Resonance, Lanzhou University Second Hospital, Lanzhou, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou, China
| | - Wen Shen
- Department of Radiology, Tianjin First Center Hospital, Tianjin, China
| | - Yanwei Miao
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dawei Wang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jia-Hong Gao
- Center for MRI Research, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Xiaochu Zhang
- Division of Life Science and Medicine, University of Science & Technology of China, Hefei, China
| | - Kai Xu
- Department of Radiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xi-Nian Zuo
- Developmental Population Neuroscience Research Center at IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
- Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Longjiang Zhang
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Mulin Jun Li
- Department of Bioinformatics, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Junfang Xian
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| | - Bing Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Chunshui Yu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging and State Key Laboratory of Experimental Hematology, Tianjin Medical University General Hospital, Tianjin, China.
- School of Medical Imaging and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
11
|
Martin-Gonzalez E, Perez-Garcia J, Herrera-Luis E, Martin-Almeida M, Kebede-Merid S, Hernandez-Pacheco N, Lorenzo-Diaz F, González-Pérez R, Sardón O, Hernández-Pérez JM, Poza-Guedes P, Sánchez-Machín I, Mederos-Luis E, Corcuera P, López-Fernández L, Román-Bernal B, Toncheva AA, Harner S, Wolff C, Brandstetter S, Abdel-Aziz MI, Hashimoto S, Vijverberg SJH, Kraneveld AD, Potočnik U, Kabesch M, Maitland-van der Zee AH, Villar J, Melén E, Pino-Yanes M. Epigenome-Wide Association Study of Asthma Exacerbations in Europeans. Allergy 2025; 80:1086-1099. [PMID: 39907155 DOI: 10.1111/all.16490] [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: 07/10/2024] [Revised: 12/03/2024] [Accepted: 01/01/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Asthma exacerbations (AEs) represent the major contributor to the global asthma burden. Although genetic and environmental factors have been associated with AEs, the role of epigenetics remains uncovered. OBJECTIVE This study aimed to identify whole blood DNA methylation (DNAm) markers associated with AEs in Europeans. METHODS DNAm was assessed in 406 blood samples from Spanish individuals using the Infinium MethylationEPIC microarray (Illumina). An epigenome-wide association study was conducted to test the association of DNAm with AEs at differentially methylated positions, regions, and epigenetic modules. CpGs suggestively associated with AEs (false discovery rate [FDR] < 0.1) were followed up for replication in 222 European individuals, and the genome-wide significance (p < 9 × 10-8) was declared after meta-analyzing the discovery and replication samples. Additional assessment was performed using nasal tissue DNAm data from 155 Spanish individuals. The effects of genetic variation on DNAm were assessed through cis-methylation quantitative trait loci (meQTL) analysis. Enrichment analyses of previous EWAS signals were conducted. RESULTS Four CpGs were associated with AEs, and two were replicated and reached genomic significance in the meta-analysis (annotated to ZBTB16 and BAIAP2). Of those, CpG cg25345365 (ZBTB16) was cross-tissue validated in nasal epithelium (p= 0.003) and associated with five independent meQTLs (FDR < 0.05). Additionally, four differentially methylated regions and one module were significantly associated with AEs. Enrichment analyses revealed an overrepresentation of prior epigenetic associations with prenatal and environmental exposures, immune-mediated diseases, and mortality. CONCLUSIONS DNAm in whole blood and nasal samples may contribute to AEs in Europeans, capturing genetic and environmental risk factors.
Collapse
Affiliation(s)
- Elena Martin-Gonzalez
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), La Laguna, Spain
| | - Javier Perez-Garcia
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), La Laguna, Spain
| | - Esther Herrera-Luis
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Mario Martin-Almeida
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), La Laguna, Spain
| | - Simon Kebede-Merid
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Natalia Hernandez-Pacheco
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Fabian Lorenzo-Diaz
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), La Laguna, Spain
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias, Universidad de La Laguna (ULL), La Laguna, Spain
| | - Ruperto González-Pérez
- Allergy Department, Hospital Universitario de Canarias, La Laguna, Spain
- Severe Asthma Unit, Allergy Department, Hospital Universitario de Canarias, La Laguna, Spain
| | - Olaia Sardón
- Division of Pediatric Respiratory Medicine, Hospital Universitario Donostia, San Sebastián, Spain
- Department of Pediatrics, University of the Basque Country (UPV/EHU), San Sebastián, Spain
| | - José M Hernández-Pérez
- Department of Respiratory Medicine, Hospital Universitario de N.S de Candelaria, Santa Cruz de Tenerife, Spain
- Respiratory Medicine, Hospital Universitario de La Palma, Santa Cruz de Tenerife, Spain
| | - Paloma Poza-Guedes
- Allergy Department, Hospital Universitario de Canarias, La Laguna, Spain
- Severe Asthma Unit, Allergy Department, Hospital Universitario de Canarias, La Laguna, Spain
| | | | - Elena Mederos-Luis
- Allergy Department, Hospital Universitario de Canarias, La Laguna, Spain
| | - Paula Corcuera
- Division of Pediatric Respiratory Medicine, Hospital Universitario Donostia, San Sebastián, Spain
| | - Leyre López-Fernández
- Division of Pediatric Respiratory Medicine, Hospital Universitario Donostia, San Sebastián, Spain
| | | | - Antoaneta A Toncheva
- University Children's Hospital Regensburg (KUNO), Hospital St. Hedwig of the Order of St. John, University of Regensburg, Regensburg, Germany
| | - Susanne Harner
- University Children's Hospital Regensburg (KUNO), Hospital St. Hedwig of the Order of St. John, University of Regensburg, Regensburg, Germany
| | - Christine Wolff
- University Children's Hospital Regensburg (KUNO), Hospital St. Hedwig of the Order of St. John, University of Regensburg, Regensburg, Germany
| | - Susanne Brandstetter
- University Children's Hospital Regensburg (KUNO), Hospital St. Hedwig of the Order of St. John, University of Regensburg, Regensburg, Germany
| | - Mahmoud Ibrahim Abdel-Aziz
- Pulmonary Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, the Netherlands
- Department of Clinical Pharmacy, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Simone Hashimoto
- Pulmonary Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, the Netherlands
- Pediatric Pulmonology, Emma's Childrens Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Susanne J H Vijverberg
- Pulmonary Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Aletta D Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Uroš Potočnik
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Faculty of Chemistry and Chemical Engineering, University of Maribor, Maribor, Slovenia
- Department for Science and Research, University Medical Centre Maribor, Maribor, Slovenia
| | - Michael Kabesch
- Department of Pediatric Pneumology and Allergy, University Children's Hospital Regensburg (KUNO), Regensburg, Germany
- Research and Development Campus Regensburg (WECARE) at the Hospital St. Hedwig of the Order of St. John, Regensburg, Germany
| | - Anke H Maitland-van der Zee
- Pulmonary Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, the Netherlands
- Pediatric Pulmonology, Emma's Childrens Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jesús Villar
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Research Unit, Hospital Universitario Dr. Negrín, Fundación Canaria Instituto de Investigación Sanitaria de Canarias, Las Palmas de Gran Canaria, Spain
- Faculty of Health Sciences, Universidad del Atlántico Medio, Las Palmas, Spain
- Li Ka Shing Knowledge Institute at St Michael's Hospital, Toronto, Canada
| | - Erik Melén
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Maria Pino-Yanes
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology, and Genetics, Universidad de La Laguna (ULL), La Laguna, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna (ULL), San Cristóbal de La Laguna, Spain
| |
Collapse
|
12
|
Cai N, Verhulst B, Andreassen OA, Buitelaar J, Edenberg HJ, Hettema JM, Gandal M, Grotzinger A, Jonas K, Lee P, Mallard TT, Mattheisen M, Neale MC, Nurnberger JI, Peyrot WJ, Tucker-Drob EM, Smoller JW, Kendler KS. Assessment and ascertainment in psychiatric molecular genetics: challenges and opportunities for cross-disorder research. Mol Psychiatry 2025; 30:1627-1638. [PMID: 39730880 PMCID: PMC11919726 DOI: 10.1038/s41380-024-02878-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 11/07/2024] [Accepted: 12/16/2024] [Indexed: 12/29/2024]
Abstract
Psychiatric disorders are highly comorbid, heritable, and genetically correlated [1-4]. The primary objective of cross-disorder psychiatric genetics research is to identify and characterize both the shared genetic factors that contribute to convergent disease etiologies and the unique genetic factors that distinguish between disorders [4, 5]. This information can illuminate the biological mechanisms underlying comorbid presentations of psychopathology, improve nosology and prediction of illness risk and trajectories, and aid the development of more effective and targeted interventions. In this review we discuss how estimates of comorbidity and identification of shared genetic loci between disorders can be influenced by how disorders are measured (phenotypic assessment) and the inclusion or exclusion criteria in individual genetic studies (sample ascertainment). Specifically, the depth of measurement, source of diagnosis, and time frame of disease trajectory have major implications for the clinical validity of the assessed phenotypes. Further, biases introduced in the ascertainment of both cases and controls can inflate or reduce estimates of genetic correlations. The impact of these design choices may have important implications for large meta-analyses of cohorts from diverse populations that use different forms of assessment and inclusion criteria, and subsequent cross-disorder analyses thereof. We review how assessment and ascertainment affect genetic findings in both univariate and multivariate analyses and conclude with recommendations for addressing them in future research.
Collapse
Affiliation(s)
- Na Cai
- Helmholtz Pioneer Campus, Helmholtz Munich, Neuherberg, Germany
- Computational Health Centre, Helmholtz Munich, Neuherberg, Germany
- School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Brad Verhulst
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, College Station, TX, USA
| | - Ole A Andreassen
- Centre of Precision Psychiatry, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo, Oslo, Norway
| | - Jan Buitelaar
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
- Karakter Child and Adolescent University Center, Nijmegen, The Netherlands
| | - Howard J Edenberg
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - John M Hettema
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Michael Gandal
- Departments of Psychiatry and Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Lifespan Brain Institute at Penn Med and the Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Andrew Grotzinger
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO, USA
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Katherine Jonas
- Department of Psychiatry & Behavioral Health, Stony Brook University, Stony Brook, NY, USA
| | - Phil Lee
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Travis T Mallard
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Manuel Mattheisen
- Department of Community Health and Epidemiology and Faculty of Computer Science, Dalhousie University, Halifax, NS, Canada
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital of Munich, Munich, Germany
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Michael C Neale
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - John I Nurnberger
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Wouter J Peyrot
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Public Health, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | | | - Jordan W Smoller
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kenneth S Kendler
- Virginia Institute for Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, USA.
- Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
13
|
Leonard HL. Novel Parkinson's Disease Genetic Risk Factors Within and Across European Populations. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.14.24319455. [PMID: 40166558 PMCID: PMC11957085 DOI: 10.1101/2025.03.14.24319455] [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
Introduction We conducted a meta-analysis of Parkinson's disease genome-wide association study summary statistics, stratified by source (clinically-recruited case-control cohorts versus population biobanks) and by general European versus European isolate ancestries. This study included 63,555 cases, 17,700 proxy cases with a family history of Parkinson's disease, and 1,746,386 controls, making it the largest investigation of Parkinson's disease genetic risk to date. Methods Meta-analyses were performed using standard fixed and random effect models for the European sub-populations, the case-control studies, and the population biobanks separately. Finally, all of the European ancestries for all study types as well as proxy cases were combined in our final cross-European meta-analysis. We estimated heritable risk across ancestry groups, investigated tissue and cell-type enrichment, and prioritized risk genes using public data to facilitate functional follow-up efforts. Results The final combined cross-European meta-analysis identified 134 risk loci (59 novel), with a total of 157 independent signals, significantly expanding our understanding of Parkinson's disease risk. Multi-omic data integration revealed that expression of the nominated risk genes are highly enriched in brain tissues, particularly in neuronal and astrocyte cell types. Additionally, we prioritized 33 high-confidence genes across these 134 loci for future follow-up studies. Conclusions By integrating diverse European populations and leveraging harmonized data from the Global Parkinson's Genetics Program (GP2), we reveal new insight into the genetic architecture of Parkinson's disease. We identified a total of 134 risk loci, expanding the number of known loci associated with PD by approximately 24%. We also provided an initial layer of biological context to these results through follow-up analyses in an effort to facilitate follow-up studies and precision medicine efforts with the goal of advancing Parkinson's disease research.
Collapse
|
14
|
Yata T, Sato G, Ogawa K, Naito T, Sonehara K, Saiki R, Edahiro R, Namba S, Watanabe M, Shirai Y, Yamamoto K, NamKoong H, Nakanishi T, Yamamoto Y, Hosokawa A, Yamamoto M, Oguro-Igashira E, Nii T, Maeda Y, Nakajima K, Nishikawa R, Tanaka H, Nakayamada S, Matsuda K, Nishigori C, Sano S, Kinoshita M, Koike R, Kimura A, Imoto S, Miyano S, Fukunaga K, Mihara M, Shimizu Y, Kawachi I, Miyamoto K, Tanaka Y, Kumanogoh A, Niino M, Nakatsuji Y, Ogawa S, Matsushita T, Kira JI, Mochizuki H, Isobe N, Okuno T, Okada Y. Contribution of germline and somatic mutations to risk of neuromyelitis optica spectrum disorder. CELL GENOMICS 2025; 5:100776. [PMID: 39986280 PMCID: PMC11960548 DOI: 10.1016/j.xgen.2025.100776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 09/27/2024] [Accepted: 01/25/2025] [Indexed: 02/24/2025]
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is a rare autoimmune disease characterized by optic neuritis and transverse myelitis, with an unclear genetic background. A genome-wide meta-analysis of NMOSD in Japanese individuals (240 patients and 50,578 controls) identified significant associations with the major histocompatibility complex region and a common variant close to CCR6 (rs12193698; p = 1.8 × 10-8, odds ratio [OR] = 1.73). In single-cell RNA sequencing (scRNA-seq) analysis (25 patients and 101 controls), the CCR6 risk variant showed disease-specific expression quantitative trait loci effects in CD4+ T (CD4T) cell subsets. Furthermore, we detected somatic mosaic chromosomal alterations (mCAs) in various autoimmune diseases and found that mCAs increase the risk of NMOSD (OR = 3.37 for copy number alteration). In scRNA-seq data, CD4T cells with 21q loss, a recurrently observed somatic event in NMOSD, showed dysregulation of type I interferon-related genes. Our integrated study identified novel germline and somatic mutations associated with NMOSD pathogenesis.
Collapse
Affiliation(s)
- Tomohiro Yata
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan; Department of Neurology, Osaka University Graduate School of Medicine, Suita, Japan; Department of Neurology, National Hospital Organization Osaka Toneyama Medical Center, Toyonaka, Japan
| | - Go Sato
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan; Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan; Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Tsurumi, Japan
| | - Kotaro Ogawa
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Tatsuhiko Naito
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan; Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Tsurumi, Japan
| | - Kyuto Sonehara
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan; Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Tsurumi, Japan; Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Japan
| | - Ryunosuke Saiki
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryuya Edahiro
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan; Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Tsurumi, Japan; Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shinichi Namba
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan; Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Tsurumi, Japan; Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mitsuru Watanabe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 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
| | - Kenichi Yamamoto
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan; Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan; Laboratory of Children's Health and Genetics, Division of Health Sciences, Osaka University Graduate School of Medicine, Suita, Japan
| | - Ho NamKoong
- Department of Infectious Diseases, Keio University School of Medicine, Tokyo, Japan
| | - Tomoko Nakanishi
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Tsurumi, Japan; Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Research Fellow, Japan Society for the Promotion of Science, Tokyo, Japan
| | - 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
| | - Akiko Hosokawa
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, Japan; Department of Neurology, Suita Municipal Hospital, Suita, Japan
| | - Mamoru Yamamoto
- Department of Neurology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Eri Oguro-Igashira
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan; Laboratory of Immune Regulation, Department of Microbiology and Immunology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Takuro Nii
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan; Department of Respiratory Medicine, National Hospital Organization Osaka Toneyama Medical Center, Toyonaka, Japan
| | - Yuichi Maeda
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Japan; Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan; Laboratory of Immune Regulation, Department of Microbiology and Immunology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kimiko Nakajima
- Department of Dermatology, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Rika Nishikawa
- Division of Dermatology, Department of Internal Related, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroaki Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Shingo Nakayamada
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Koichi Matsuda
- Laboratory of Clinical Genome Sequencing, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Chikako Nishigori
- Division of Dermatology, Department of Internal Related, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shigetoshi Sano
- Department of Dermatology, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Makoto Kinoshita
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Ryuji Koike
- Health Science Research and Development Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akinori Kimura
- Institute of Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, the Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Satoru Miyano
- M&D Data Science Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Masahito Mihara
- Department of Neurology, Kawasaki Medical School, Kurashiki, Japan
| | - Yuko Shimizu
- Department of Neurology, Tokyo Women's Medical University, Tokyo, Japan; Department of Medical Safety, Tokyo Women's Medical University, Tokyo, Japan
| | - Izumi Kawachi
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan; Medical Education Center, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Katsuichi Miyamoto
- Department of Neurology, Kindai University School of Medicine, Osakasayama, Japan; Department of Neurology, Wakayama Medical University, Wakayama, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Atsushi Kumanogoh
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Japan; 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; Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Japan
| | - Masaaki Niino
- Department of Clinical Research, National Hospital Organization Hokkaido Medical Center, Sapporo, Japan
| | - Yuji Nakatsuji
- Department of Neurology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan; Center for Hematology and Regenerative Medicine, Department of Medicine (MedH), Karolinska Institutet, Huddinge, Sweden
| | - Takuya Matsushita
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Neurology, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Jun-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Neurology, Brain and Nerve Center, Fukuoka Central Hospital, Fukuoka, Japan; Translational Neuroscience Center, Graduate School of Medicine, and School of Pharmacy at Fukuoka, International University of Health and Welfare, Fukuoka, Japan
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Noriko Isobe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Tatsusada Okuno
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, Japan.
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan; Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Tsurumi, Japan; Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan; Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Japan.
| |
Collapse
|
15
|
Mitsumori R, Asanomi Y, Morizono T, Shigemizu D, Niida S, Ozaki K. A genome-wide association study identifies a novel East Asian-specific locus for dementia with Lewy bodies in Japanese subjects. Mol Med 2025; 31:87. [PMID: 40045203 PMCID: PMC11884146 DOI: 10.1186/s10020-025-01115-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 02/04/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Dementia with Lewy bodies (DLB) is the second most common type of degenerative dementia in older patients. As with other multifactorial diseases, the pathogenesis results from interactions of environmental and genetic factors. The genetic basis of DLB is not yet fully understood. Recent genomic analyses of DLB in Caucasian cohorts identified genetic susceptibility loci for DLB, but the comprehensive genomic analysis in Asians was still not performed. METHODS We conducted a genome-wide association study (GWAS) in Japanese subjects (211 DLB cases and 6113 controls) to clarify the genetic architecture of DLB pathogenesis. RESULTS We identified the East Asian-specific DHTKD1 locus (rs138587229) on chromosome 10 with genome-wide significance (GWS; P = 3.27 × 10-8) and the ICOS/PARD3B locus on chromosome 2 with suggestive significance (P = 3.95 × 10-7) as novel DLB genetic risk loci. We also confirmed the APOE locus (rs429358, P < 5.0 × 10-8), a known risk locus for DLB and Alzheimer's disease in Caucasians. The DHTKD1 locus was associated with the gene expression of SEC61A2 and showed a causal relationship with cholinesterase levels. In a trans-ethnic meta-analysis that included Japanese, UK Biobank, and other Caucasian GWAS, we confirmed the risk for DLB at APOE and SNCA loci with GWS. Transcriptome-wide association analysis identified ZNF155 and ZNF284 in the brain cortex and GPRIN3 in the substantia nigra as putative causal genes for DLB. CONCLUSIONS This is the first GWAS for DLB in East Asians, and our findings provide new biological and clinical insights into the pathogenesis of DLB.
Collapse
Affiliation(s)
- Risa Mitsumori
- Medical Genome Center, National Center for Geriatrics and Gerontology, Research Institute, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
| | - Yuya Asanomi
- Medical Genome Center, National Center for Geriatrics and Gerontology, Research Institute, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
| | - Takashi Morizono
- Medical Genome Center, National Center for Geriatrics and Gerontology, Research Institute, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
| | - Daichi Shigemizu
- Medical Genome Center, National Center for Geriatrics and Gerontology, Research Institute, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan
| | - Shumpei Niida
- National Center for Geriatrics and Gerontology, Research Institute, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan
| | - Kouichi Ozaki
- Medical Genome Center, National Center for Geriatrics and Gerontology, Research Institute, 7-430 Morioka-Cho, Obu, Aichi, 474-8511, Japan.
- Department of Cardiovascular Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3 Kasumi, Minami-Ku, Hiroshima, 734-8553, Japan.
- RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-Cho, Tsurumi-Ku, Yokohama, Kanagawa, 230-0045, Japan.
| |
Collapse
|
16
|
Vialle RA, de Paiva Lopes K, Li Y, Ng B, Schneider JA, Buchman AS, Wang Y, Farfel JM, Barnes LL, Wingo AP, Wingo TS, Seyfried NT, De Jager PL, Gaiteri C, Tasaki S, Bennett DA. Structural variants linked to Alzheimer's disease and other common age-related clinical and neuropathologic traits. Genome Med 2025; 17:20. [PMID: 40038788 PMCID: PMC11881306 DOI: 10.1186/s13073-025-01444-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 02/24/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a complex neurodegenerative disorder with substantial genetic influence. While genome-wide association studies (GWAS) have identified numerous risk loci for late-onset AD (LOAD), the functional mechanisms underlying most of these associations remain unresolved. Large genomic rearrangements, known as structural variants (SVs), represent a promising avenue for elucidating such mechanisms within some of these loci. METHODS By leveraging data from two ongoing cohort studies of aging and dementia, the Religious Orders Study and Rush Memory and Aging Project (ROS/MAP), we performed genome-wide association analysis testing 20,205 common SVs from 1088 participants with whole genome sequencing (WGS) data. A range of Alzheimer's disease and other common age-related clinical and neuropathologic traits were examined. RESULTS First, we mapped SVs across 81 AD risk loci and discovered 22 SVs in linkage disequilibrium (LD) with GWAS lead variants and directly associated with the phenotypes tested. The strongest association was a deletion of an Alu element in the 3'UTR of the TMEM106B gene, in high LD with the respective AD GWAS locus and associated with multiple AD and AD-related disorders (ADRD) phenotypes, including tangles density, TDP-43, and cognitive resilience. The deletion of this element was also linked to lower TMEM106B protein abundance. We also found a 22-kb deletion associated with depression in ROS/MAP and bearing similar association patterns as GWAS SNPs at the IQCK locus. In addition, we leveraged our catalog of SV-GWAS to replicate and characterize independent findings in SV-based GWAS for AD and five other neurodegenerative diseases. Among these findings, we highlight the replication of genome-wide significant SVs for progressive supranuclear palsy (PSP), including markers for the 17q21.31 MAPT locus inversion and a 1483-bp deletion at the CYP2A13 locus, along with other suggestive associations, such as a 994-bp duplication in the LMNTD1 locus, suggestively linked to AD and a 3958-bp deletion at the DOCK5 locus linked to Lewy body disease (LBD) (P = 3.36 × 10-4). CONCLUSIONS While still limited in sample size, this study highlights the utility of including analysis of SVs for elucidating mechanisms underlying GWAS loci and provides a valuable resource for the characterization of the effects of SVs in neurodegenerative disease pathogenesis.
Collapse
Affiliation(s)
- Ricardo A Vialle
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA.
| | - Katia de Paiva Lopes
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Yan Li
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Bernard Ng
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Aron S Buchman
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Yanling Wang
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Jose M Farfel
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Lisa L Barnes
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Aliza P Wingo
- Department of Psychiatry, University of California, Davis, Davis, CA, USA
- VA Northern California Health Care System, Davis, CA, USA
| | - Thomas S Wingo
- Department of Neurology, University of California, Davis, Davis, CA, USA
| | - Nicholas T Seyfried
- Department of Neurology and Department of Biochemistry, Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Chris Gaiteri
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Shinya Tasaki
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| |
Collapse
|
17
|
Flinn C, McInerney A, Nearchou F. The prevalence of comorbid mental health difficulties in young people with chronic skin conditions: A systematic review and meta-analysis. J Health Psychol 2025; 30:652-679. [PMID: 38812260 PMCID: PMC11927027 DOI: 10.1177/13591053241252216] [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: 05/31/2024] Open
Abstract
Chronic skin conditions can have psychosocial and somatic implications, influencing well-being and quality of life. This systematic review and meta-analysis aimed to synthesise evidence on the prevalence of comorbid mental health difficulties in 0-25-year-olds with chronic skin conditions. A secondary aim included identifying factors associated with resilience. The narrative synthesis included 45 studies. Four meta-analyses were performed with moderate-high quality studies, one for each outcome: diagnosed mental disorders; mental health symptoms; suicidal behaviour; socio-emotional and behavioural difficulties. The pooled prevalence of diagnosed mental disorders was 1.2% (95% CI = 0.2-6.1); of mental health symptoms was 22.6% (95% CI = 18.9-26.7); of suicidal behaviour was 7.8% (95% CI = 1.4-3.1); of socio-emotional and behavioural difficulties was 20.9% (95% CI = 14.7-28.8). Findings demonstrate the pooled prevalence of comorbid mental health difficulties in youth with chronic skin conditions.
Collapse
|
18
|
Ajasa AA, Gjøen HM, Boison SA, Lillehammer M. Genome-wide association analysis using multiple Atlantic salmon populations. Genet Sel Evol 2025; 57:9. [PMID: 40016680 PMCID: PMC11869457 DOI: 10.1186/s12711-025-00959-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 02/11/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND In a previous study, we found low persistence of linkage disequilibrium (LD) phase across breeding populations of Atlantic salmon. Accordingly, we observed no increase in accuracy from combining these populations for genomic prediction. In this study, we aimed to examine if the same were true for detection power in genome-wide association studies (GWAS), in terms of reduction in p-values, and if the precision of mapping quantitative trait loci (QTL) would improve from such analysis. Since individual records may not always be available, e.g. due to proprietorship or confidentiality, we also compared mega-analysis and meta-analysis. Mega-analysis needs access to all individual records, whereas meta-analysis utilizes parameters, such as p-values or allele substitution effects, from multiple studies or populations. Furthermore, different methods for determining the presence or absence of independent or secondary signals, such as conditional association analysis, approximate conditional and joint analysis (COJO), and the clumping approach, were assessed. RESULTS Mega-analysis resulted in increased detection power, in terms of reduction in p-values, and increased precision, compared to the within-population GWAS. Only one QTL was detected using conditional association analysis, both within populations and in mega-analysis, while the number of QTL detected with COJO and the clumping approach ranged from 1 to 19. The allele substitution effect and -log10p-values obtained from mega-analysis were highly correlated with the corresponding values from various meta-analysis methods. Compared to mega-analysis, a higher detection power and reduced precision were obtained with the meta-analysis methods. CONCLUSIONS Our results show that combining multiple datasets or populations in a mega-analysis can increase detection power and mapping precision. With meta-analysis, a higher detection power was obtained compared to mega-analysis. However, care must be taken in the interpretation of the meta-analysis results from multiple populations because their test statistics might be inflated due to population structure or cryptic relatedness.
Collapse
Affiliation(s)
- Afees A Ajasa
- Department of Breeding and Genetics, Nofima (Norwegian Institute of Food, Fisheries and Aquaculture Research), P. O. Box 210, N-1431, Ås, Norway.
- Department of Animal and Aquacultural Sciences, Norwegian University of Life Sciences, 5003 NMBU, N-1432, Ås, Norway.
| | - Hans M Gjøen
- Department of Animal and Aquacultural Sciences, Norwegian University of Life Sciences, 5003 NMBU, N-1432, Ås, Norway
| | | | - Marie Lillehammer
- Department of Breeding and Genetics, Nofima (Norwegian Institute of Food, Fisheries and Aquaculture Research), P. O. Box 210, N-1431, Ås, Norway
| |
Collapse
|
19
|
Singh M, Chatzinakos C, Barr PB, Gentry AE, Bigdeli TB, Webb BT, Peterson RE. Trans-ancestry Genome-Wide Analyses in UK Biobank Yield Novel Risk Loci for Major Depression. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.22.25322721. [PMID: 40061314 PMCID: PMC11888526 DOI: 10.1101/2025.02.22.25322721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/17/2025]
Abstract
Most genome-wide association studies (GWASs) of depression focus on broad, heterogeneous outcomes, limiting the discovery of genomic risk loci specific to major depressive disorder (MDD). Previous UK Biobank (UKB) studies had limited ability to pinpoint MDD-associated loci due to a smaller sample with strictly defined MDD outcomes and further exclusion of many participants based on ancestry or relatedness, significantly underutilizing this resource's potential for elucidating the genetic architecture of MDD. Here, we present novel genomic insights into MDD by fully utilizing existing UKB data through (1) a trans-ancestry GWAS pipeline using two complementary approaches controlling for population structure and relatedness and (2) an increased sample with MDD symptom-level data across two mental health assessments. We identified strict MDD outcomes among 211,535 participants, representing a 38% increase in eligible participants from prior studies with only one assessment. Ancestrally inclusive analyses yielded 61 genomic risk loci across depression phenotypes, compared to 47 in the analyses restricted to participants genetically similar to European ancestry. Fourteen of these loci, including five novel, were associated with strict MDD phenotypes, whereas only one locus has been previously reported in UKB. MDD-associated genomic loci and predicted gene expression levels showed little overlap with broad depression, indicating higher specificity. Notably, polygenic scores based on these results were significantly associated with depression diagnoses across ancestry groups in the All of Us Research Program, highlighting the shared genetic architecture across populations. While the trans-ancestry analyses, which included non-European participants, increased the number of associated loci, the discovery of non-European ancestry-specific loci was limited, underscoring the need for larger, globally representative studies of MDD. Importantly, beyond these results, our GWAS pipeline will facilitate inclusive analyses of other traits and disorders, helping improve statistical power, representation, and generalizability in genomic studies.
Collapse
Affiliation(s)
- Madhurbain Singh
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, USA
| | - Chris Chatzinakos
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - Peter B Barr
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Amanda Elswick Gentry
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - Tim B Bigdeli
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Bradley T Webb
- GenOmics and Translational Research Center, RTI International, Research Triangle Park, NC, USA
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| | - Roseann E Peterson
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
- Virginia Institute for Psychiatric and Behavioral Genetics, Department of Psychiatry, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
20
|
Križanac AM, Reimer C, Heise J, Liu Z, Pryce JE, Bennewitz J, Thaller G, Falker-Gieske C, Tetens J. Sequence-based GWAS in 180,000 German Holstein cattle reveals new candidate variants for milk production traits. Genet Sel Evol 2025; 57:3. [PMID: 39905301 PMCID: PMC11796172 DOI: 10.1186/s12711-025-00951-9] [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: 06/25/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Milk production traits are complex and influenced by many genetic and environmental factors. Although extensive research has been performed for these traits, with many associations unveiled thus far, due to their crucial economic importance, complex genetic architecture, and the fact that causal variants in cattle are still scarce, there is a need for a better understanding of their genetic background. In this study, we aimed to identify new candidate loci associated with milk production traits in German Holstein cattle, the most important dairy breed in Germany and worldwide. For that purpose, 180,217 cattle were imputed to the sequence level and large-scale genome-wide association study (GWAS) followed by fine-mapping and evolutionary and functional annotation were carried out to identify and prioritize new association signals. RESULTS Using the imputed sequence data of a large cattle dataset, we identified 50,876 significant variants, confirming many known and identifying previously unreported candidate variants for milk (MY), fat (FY), and protein yield (PY). Genome-wide significant signals were fine-mapped with the Bayesian approach that determines the credible variant sets and generates the probability of causality for each signal. The variants with the highest probabilities of being causal were further classified using external information about the function and evolution, making the prioritization for subsequent validation experiments easier. The top potential causal variants determined with fine-mapping explained a large percentage of genetic variance compared to random ones; 178 variants explained 11.5%, 104 explained 7.7%, and 68 variants explained 3.9% of the variance for MY, FY, and PY, respectively, demonstrating the potential for causality. CONCLUSIONS Our findings proved the power of large samples and sequence-based GWAS in detecting new association signals. In order to fully exploit the power of GWAS, one should aim at very large samples combined with whole-genome sequence data. These can also come with both computational and time burdens, as presented in our study. Although milk production traits in cattle are comprehensively investigated, the genetic background of these traits is still not fully understood, with the potential for many new associations to be revealed, as shown. With constantly growing sample sizes, we expect more insights into the genetic architecture of milk production traits in the future.
Collapse
Affiliation(s)
- Ana-Marija Križanac
- Department of Animal Sciences, University of Goettingen, Burckhardtweg 2, 37077, Göttingen, Germany.
- Center for Integrated Breeding Research, Department of Animal Sciences, University of Goettingen, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany.
| | - Christian Reimer
- Center for Integrated Breeding Research, Department of Animal Sciences, University of Goettingen, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, 31535, Neustadt, Germany
| | - Johannes Heise
- Vereinigte Informationssysteme Tierhaltung w.V. (VIT), 27283, Verden, Germany
| | - Zengting Liu
- Vereinigte Informationssysteme Tierhaltung w.V. (VIT), 27283, Verden, Germany
| | - Jennie E Pryce
- Agriculture Victoria Research, AgriBio, Centre for AgriBioscience, Bundoora, VIC, 3083, Australia
- School of Applied Systems Biology, La Trobe University, Bundoora, VIC, 3083, Australia
| | - Jörn Bennewitz
- Institute of Animal Science, University of Hohenheim, 70599, Stuttgart, Germany
| | - Georg Thaller
- Institute of Animal Breeding and Husbandry, Christian-Albrechts-University, 24118, Kiel, Germany
| | - Clemens Falker-Gieske
- Department of Animal Sciences, University of Goettingen, Burckhardtweg 2, 37077, Göttingen, Germany
- Center for Integrated Breeding Research, Department of Animal Sciences, University of Goettingen, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany
| | - Jens Tetens
- Department of Animal Sciences, University of Goettingen, Burckhardtweg 2, 37077, Göttingen, Germany
- Center for Integrated Breeding Research, Department of Animal Sciences, University of Goettingen, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany
| |
Collapse
|
21
|
Kuang A, Hivert MF, Hayes MG, Lowe WL, Scholtens DM. Multi-ancestry genome-wide association analyses: a comparison of meta- and mega-analyses in the Hyperglycemia and Adverse Pregnancy Outcome (HAPO) study. BMC Genomics 2025; 26:65. [PMID: 39849370 PMCID: PMC11755808 DOI: 10.1186/s12864-025-11229-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 01/08/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND There is increasing need for effective incorporation of high-dimensional genetics data from individuals with varied ancestry in genome-wide association (GWAS) analyses. Classically, multi-ancestry GWAS analyses are performed using statistical meta-analysis to combine results conducted within homogeneous ancestry groups. The emergence of cosmopolitan reference panels makes collective preprocessing of GWAS data possible, but impact on downstream GWAS results in a mega-analysis framework merits investigation. We utilized GWAS data from the multi-national Hyperglycemia and Adverse Pregnancy Outcome Study to investigate differences in GWAS findings using a homogeneous ancestry meta-analysis versus a heterogeneous ancestry mega-analysis pipeline. Maternal fasting and 1-hr glucose and metabolomics measured during a 2-hr 75-gram oral glucose tolerance test during early third trimester pregnancy were evaluated as phenotypes. RESULTS For the homogeneous ancestry meta-analysis pipeline, variant data were prepared by identifying sets of individuals with similar ancestry and imputing to ancestry-specific reference panels. GWAS was conducted within each ancestry group and results were combined using random-effects meta-analysis. For the heterogeneous ancestry mega-analysis pipeline, data for all individuals were collectively imputed to the Trans-Omics for Precision Medicine (TOPMed) cosmopolitan reference panel, and GWAS was conducted using a unified mega-analysis. The meta-analysis pipeline identified genome-wide significant associations for 15 variants in a region close to GCK on chromosome 7 with maternal fasting glucose and no significant findings for 1-hr glucose. Associations in this same region were identified using the mega-analysis pipeline, along with a well-documented association at MTNR1B on chromosome 11 with both fasting and 1-hr maternal glucose. For metabolomics analyses, the number of significant findings in the heterogeneous ancestry mega-analysis far exceeded those from the homogeneous ancestry meta-analysis and confirmed many previously documented associations, but genomic inflation factors were much more variable. CONCLUSIONS For multi-ancestry GWAS, heterogeneous ancestry mega-analysis generates a rich set of variants for analysis using a cosmopolitan reference panel and results in vastly more significant, biologically credible and previously documented associations than a homogeneous ancestry meta-analysis approach. Genomic inflation factors do indicate that findings from the mega-analysis pipeline may merit cautious interpretation and further follow-up.
Collapse
Affiliation(s)
- Alan Kuang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Marie-France Hivert
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - M Geoffrey Hayes
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - William L Lowe
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Denise M Scholtens
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
22
|
Liang X, Mounier N, Apfel N, Khalid S, Frayling TM, Bowden J. Using clustering of genetic variants in Mendelian randomization to interrogate the causal pathways underlying multimorbidity from a common risk factor. Genet Epidemiol 2025; 49:e22582. [PMID: 39138631 PMCID: PMC11647065 DOI: 10.1002/gepi.22582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/17/2024] [Accepted: 07/09/2024] [Indexed: 08/15/2024]
Abstract
Mendelian randomization (MR) is an epidemiological approach that utilizes genetic variants as instrumental variables to estimate the causal effect of an exposure on a health outcome. This paper investigates an MR scenario in which genetic variants aggregate into clusters that identify heterogeneous causal effects. Such variant clusters are likely to emerge if they affect the exposure and outcome via distinct biological pathways. In the multi-outcome MR framework, where a shared exposure causally impacts several disease outcomes simultaneously, these variant clusters can provide insights into the common disease-causing mechanisms underpinning the co-occurrence of multiple long-term conditions, a phenomenon known as multimorbidity. To identify such variant clusters, we adapt the general method of agglomerative hierarchical clustering to multi-sample summary-data MR setup, enabling cluster detection based on variant-specific ratio estimates. Particularly, we tailor the method for multi-outcome MR to aid in elucidating the causal pathways through which a common risk factor contributes to multiple morbidities. We show in simulations that our "MR-AHC" method detects clusters with high accuracy, outperforming the existing methods. We apply the method to investigate the causal effects of high body fat percentage on type 2 diabetes and osteoarthritis, uncovering interconnected cellular processes underlying this multimorbid disease pair.
Collapse
Affiliation(s)
- Xiaoran Liang
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Ninon Mounier
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life SciencesUniversity of ExeterExeterUK
| | - Nicolas Apfel
- Department of EconomicsUniversity of SouthamptonSouthamptonUK
| | - Sara Khalid
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal SciencesUniversity of OxfordOxfordUK
| | - Timothy M. Frayling
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life SciencesUniversity of ExeterExeterUK
- Department of Genetic Medicine and Development, Faculty of MedicineCMUGenevaSwitzerland
| | - Jack Bowden
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life SciencesUniversity of ExeterExeterUK
| |
Collapse
|
23
|
Li R, Li M, Zhao N. A Mixed-Effect Kernel Machine Regression Model for Integrative Analysis of Alpha Diversity in Microbiome Studies. Genet Epidemiol 2025; 49:e22596. [PMID: 39350346 DOI: 10.1002/gepi.22596] [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: 11/10/2023] [Revised: 08/22/2024] [Accepted: 09/05/2024] [Indexed: 12/20/2024]
Abstract
Increasing evidence suggests that human microbiota plays a crucial role in many diseases. Alpha diversity, a commonly used summary statistic that captures the richness and/or evenness of the microbial community, has been associated with many clinical conditions. However, individual studies that assess the association between alpha diversity and clinical conditions often provide inconsistent results due to insufficient sample size, heterogeneous study populations and technical variability. In practice, meta-analysis tools have been applied to integrate data from multiple studies. However, these methods do not consider the heterogeneity caused by sequencing protocols, and the contribution of each study to the final model depends mainly on its sample size (or variance estimate). To combine studies with distinct sequencing protocols, a robust statistical framework for integrative analysis of microbiome datasets is needed. Here, we propose a mixed-effect kernel machine regression model to assess the association of alpha diversity with a phenotype of interest. Our approach readily incorporates the study-specific characteristics (including sequencing protocols) to allow for flexible modeling of microbiome effect via a kernel similarity matrix. Within the proposed framework, we provide three hypothesis testing approaches to answer different questions that are of interest to researchers. We evaluate the model performance through extensive simulations based on two distinct data generation mechanisms. We also apply our framework to data from HIV reanalysis consortium to investigate gut dysbiosis in HIV infection.
Collapse
Affiliation(s)
- Runzhe Li
- Department of Biostatistics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Mo Li
- Department of Mathematics, University of Louisiana at Lafayette, Lafayette, Louisiana, USA
| | - Ni Zhao
- Department of Biostatistics, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
24
|
Akutsu Y, Ota M, Itamiya T, Mori M, Morio T, Yamamoto K, Okamura T, Fujio K. Effect of Epstein-Barr Virus infection on gene regulation in immune cells of patients with Immune-Mediated Diseases. J Autoimmun 2025; 150:103355. [PMID: 39787676 DOI: 10.1016/j.jaut.2024.103355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/20/2024] [Accepted: 12/21/2024] [Indexed: 01/12/2025]
Abstract
It has been known that Epstein-Barr virus (EBV) can latently infect immune cells after the initial infection, and epidemiological studies have suggested its association with the onset of immune-mediated diseases (IMDs). However, the specific impact of EBV infection on IMDs pathology remains unclear. We quantified EBV load of B cell subsets (Naïve B cells, Unswitched memory B cells, Switched memory B cells, Double negative B cells, and Plasmablasts) in IMD patients as well as healthy control (HC) using bulk RNA sequencing data of 504 donors. The EBV load was clearly higher in IMD patients compared to HC. Furthermore, the wide range of EBV load in this dataset enabled us to assess the impact of EBV load on gene regulation. We found many examples of expression quantitative trait loci (eQTL) whose effects were associated with EBV load. Expression QTLs that exhibited larger effects with increasing EBV load were significantly overlapped with binding sites of transcription factors derived from the EBV genome. These EBV load-associated eQTLs exhibited high enrichment of systemic lupus erythematosus (SLE) GWAS signals, suggesting the mechanical link of EBV infection and the onset of the disease via gene regulation. These findings provide the first evidence of the influence of EBV infection on gene regulation in human primary cells and its association with the SLE onset and/or progression.
Collapse
Affiliation(s)
- Yuko Akutsu
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mineto Ota
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Takahiro Itamiya
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masaaki Mori
- Department of Lifetime Clinical Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomohiro Morio
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kazuhiko Yamamoto
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Tomohisa Okamura
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
25
|
Guo X, Feng Y, Ji X, Jia N, Maimaiti A, Lai J, Wang Z, Yang S, Hu S. Shared genetic architecture and bidirectional clinical risks within the psycho-metabolic nexus. EBioMedicine 2025; 111:105530. [PMID: 39731856 PMCID: PMC11743124 DOI: 10.1016/j.ebiom.2024.105530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 12/12/2024] [Accepted: 12/12/2024] [Indexed: 12/30/2024] Open
Abstract
BACKGROUND Increasing evidence suggests a complex interplay between psychiatric disorders and metabolic dysregulations. However, most research has been limited to specific disorder pairs, leaving a significant gap in our understanding of the broader psycho-metabolic nexus. METHODS This study leveraged large-scale cohort data and genome-wide association study (GWAS) summary statistics, covering 8 common psychiatric disorders and 43 metabolic traits. We introduced a comprehensive analytical strategy to identify shared genetic bases sequentially, from key genetic correlation regions to local pleiotropy and pleiotropic genes. Finally, we developed polygenic risk score (PRS) models to translate these findings into clinical applications. FINDINGS We identified significant bidirectional clinical risks between psychiatric disorders and metabolic dysregulations among 310,848 participants from the UK Biobank. Genetic correlation analysis confirmed 104 robust trait pairs, revealing 1088 key genomic regions, including critical hotspots such as chr3: 47588462-50387742. Cross-trait meta-analysis uncovered 388 pleiotropic single nucleotide variants (SNVs) and 126 shared causal variants. Among variants, 45 novel SNVs were associated with psychiatric disorders and 75 novel SNVs were associated with metabolic traits, shedding light on new targets to unravel the mechanism of comorbidity. Notably, RBM6, a gene involved in alternative splicing and cellular stress response regulation, emerged as a key pleiotropic gene. When psychiatric and metabolic genetic information were integrated, PRS models demonstrated enhanced predictive power. INTERPRETATION The study highlights the intertwined genetic and clinical relationships between psychiatric disorders and metabolic dysregulations, emphasising the need for integrated approaches in diagnosis and treatment. FUNDING The National Key Research and Development Program of China (2023YFC2506200, SHH). The National Natural Science Foundation of China (82273741, SY).
Collapse
Affiliation(s)
- Xiaonan Guo
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yu Feng
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Department of Psychiatry, Melbourne Neuropsychiatry Centre, The University of Melbourne, Carlton South, VIC, Australia
| | - Xiaolong Ji
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ningning Jia
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Aierpati Maimaiti
- Department of Neurosurgery, Xinjiang Medical University Affiliated First Hospital, Urumqi, Xinjiang, China
| | - Jianbo Lai
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zheng Wang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Sheng Yang
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Shaohua Hu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Nanhu Brain-Computer Interface Institute, Hangzhou, Zhejiang, China; Zhejiang Key Laboratory of Precision Psychiatry, Hangzhou, 310003, China; Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 311121, China; Brain Research Institute of Zhejiang University, Hangzhou, 310058, China; MOE Frontier Science Center for Brain Science and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, 310058, China; Department of Psychology and Behavioral Sciences, Graduate School, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
26
|
Jones SC, Cardone KM, Bradford Y, Tishkoff SA, Ritchie MD. The Impact of Ancestry on Genome-Wide Association Studies. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2025; 30:251-267. [PMID: 39670375 PMCID: PMC11694900 DOI: 10.1142/9789819807024_0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Genome-wide association studies (GWAS) are an important tool for the study of complex disease genetics. Decisions regarding the quality control (QC) procedures employed as part of a GWAS can have important implications on the results and their biological interpretation. Many GWAS have been conducted predominantly in cohorts of European ancestry, but many initiatives aim to increase the representation of diverse ancestries in genetic studies. The question of how these data should be combined and the consequences that genetic variation across ancestry groups might have on GWAS results warrants further investigation. In this study, we focus on several commonly used methods for combining genetic data across diverse ancestry groups and the impact these decisions have on the outcome of GWAS summary statistics. We ran GWAS on two binary phenotypes using ancestry-specific, multi-ancestry mega-analysis, and meta-analysis approaches. We found that while multi-ancestry mega-analysis and meta-analysis approaches can aid in identifying signals shared across ancestries, they can diminish the signal of ancestry-specific associations and modify their effect sizes. These results demonstrate the potential impact on downstream post-GWAS analyses and follow-up studies. Decisions regarding how the genetic data are combined has the potential to mask important findings that might serve individuals of ancestries that have been historically underrepresented in genetic studies. New methods that consider ancestry-specific variants in conjunction with the shared variants need to be developed.
Collapse
Affiliation(s)
- Steven Christopher Jones
- Genomics and Computational Biology Graduate Group, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Katie M Cardone
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Yuki Bradford
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Sarah A Tishkoff
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Marylyn D Ritchie
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA,
| |
Collapse
|
27
|
Mu Z, Randolph HE, Aguirre-Gamboa R, Ketter E, Dumaine A, Locher V, Brandolino C, Liu X, Kaufmann DE, Barreiro LB, Li YI. Impact of disease-associated chromatin accessibility QTLs across immune cell types and contexts. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.05.24318552. [PMID: 39711700 PMCID: PMC11661428 DOI: 10.1101/2024.12.05.24318552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Only a third of immune-associated loci from genome-wide association studies (GWAS) colocalize with expression quantitative trait loci (eQTLs). To learn about causal genes and mechanisms at the remaining loci, we created a unified single-cell chromatin accessibility (scATAC-seq) map in peripheral blood comprising a total of 282,424 cells from 48 individuals. Clustering and topic modeling of scATAC data identified discrete cell-types and continuous cell states, which helped reveal disease-relevant cellular contexts, and allowed mapping of genetic effects on chromatin accessibility across these contexts. We identified 37,390 chromatin accessibility QTLs (caQTL) at 10% FDR across eight cell groups and observed extensive sharing of caQTLs across immune cell contexts, finding that fewer than 20% of caQTLs are specific to a single cell type. Notably, caQTLs colocalized with ∼50% more GWAS loci compared to eQTLs, helping to nominate putative causal genes for many unexplained loci. However, most GWAS-caQTL colocalizations had no detectable downstream regulatory effects on gene expression levels in the same cell type. We find evidence that the higher rates of colocalization between caQTLs and GWAS signals reflect missing disease-relevant cellular contexts among existing eQTL studies. Thus, there remains a pressing need for identifying disease-causing cellular contexts and for mapping gene regulatory variation in these cells.
Collapse
|
28
|
Gorman BR, Voloudakis G, Igo RP, Kinzy T, Halladay CW, Bigdeli TB, Zeng B, Venkatesh S, Cooke Bailey JN, Crawford DC, Markianos K, Dong F, Schreiner PA, Zhang W, Hadi T, Anger MD, Stockwell A, Melles RB, Yin J, Choquet H, Kaye R, Patasova K, Patel PJ, Yaspan BL, Jorgenson E, Hysi PG, Lotery AJ, Gaziano JM, Tsao PS, Fliesler SJ, Sullivan JM, Greenberg PB, Wu WC, Assimes TL, Pyarajan S, Roussos P, Peachey NS, Iyengar SK. Genome-wide association analyses identify distinct genetic architectures for age-related macular degeneration across ancestries. Nat Genet 2024; 56:2659-2671. [PMID: 39623103 DOI: 10.1038/s41588-024-01764-0] [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: 09/02/2022] [Accepted: 04/22/2024] [Indexed: 12/12/2024]
Abstract
To effectively reduce vision loss due to age-related macular generation (AMD) on a global scale, knowledge of its genetic architecture in diverse populations is necessary. A critical element, AMD risk profiles in African and Hispanic/Latino ancestries, remains largely unknown. We combined data in the Million Veteran Program with five other cohorts to conduct the first multi-ancestry genome-wide association study of AMD and discovered 63 loci (30 novel). We observe marked cross-ancestry heterogeneity at major risk loci, especially in African-ancestry populations which demonstrate a primary signal in a major histocompatibility complex class II haplotype and reduced risk at the established CFH and ARMS2/HTRA1 loci. Dissecting local ancestry in admixed individuals, we find significantly smaller marginal effect sizes for CFH risk alleles in African ancestry haplotypes. Broadening efforts to include ancestrally distinct populations helped uncover genes and pathways that boost risk in an ancestry-dependent manner and are potential targets for corrective therapies.
Collapse
Affiliation(s)
- Bryan R Gorman
- Center for Data and Computational Sciences (C-DACS), VA Cooperative Studies Program, VA Boston Healthcare System, Boston, MA, USA
- Booz Allen Hamilton, McLean, VA, USA
| | - Georgios Voloudakis
- Center for Disease Neurogenomics, Department of Psychiatry; Friedman Brain Institute; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Precision Medicine and Translational Therapeutics, VISN 2 Mental Illness Research, Education, and Clinical Center (MIRECC), James J. Peters Veterans Affairs Medical Center, New York/New Jersey VA Health Care Network, Bronx, NY, USA
| | - Robert P Igo
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Tyler Kinzy
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Christopher W Halladay
- Center of Innovation in Long Term Services and Supports, VA Providence Healthcare System, Providence, RI, USA
| | - Tim B Bigdeli
- Research Service, VA New York Harbor Healthcare System, Brooklyn, NY, USA
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Biao Zeng
- Center for Disease Neurogenomics, Department of Psychiatry; Friedman Brain Institute; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sanan Venkatesh
- Center for Disease Neurogenomics, Department of Psychiatry; Friedman Brain Institute; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Precision Medicine and Translational Therapeutics, VISN 2 Mental Illness Research, Education, and Clinical Center (MIRECC), James J. Peters Veterans Affairs Medical Center, New York/New Jersey VA Health Care Network, Bronx, NY, USA
| | - Jessica N Cooke Bailey
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
- Department of Genetics & Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Dana C Crawford
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
- Department of Genetics & Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Kyriacos Markianos
- Center for Data and Computational Sciences (C-DACS), VA Cooperative Studies Program, VA Boston Healthcare System, Boston, MA, USA
| | - Frederick Dong
- Center for Data and Computational Sciences (C-DACS), VA Cooperative Studies Program, VA Boston Healthcare System, Boston, MA, USA
- Booz Allen Hamilton, McLean, VA, USA
| | - Patrick A Schreiner
- Center for Data and Computational Sciences (C-DACS), VA Cooperative Studies Program, VA Boston Healthcare System, Boston, MA, USA
- Booz Allen Hamilton, McLean, VA, USA
| | - Wen Zhang
- Center for Disease Neurogenomics, Department of Psychiatry; Friedman Brain Institute; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tamer Hadi
- Eye Clinic, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
- Department of Ophthalmology and Visual Sciences, University Hospitals Eye Institute, Cleveland, OH, USA
| | - Matthew D Anger
- Eye Clinic, VA Western NY Healthcare System, Buffalo, NY, USA
- Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, SUNY-University at Buffalo, Buffalo, NY, USA
| | - Amy Stockwell
- Department of Human Genetics, Genentech, South San Francisco, CA, USA
| | - Ronald B Melles
- Department of Ophthalmology, Kaiser Permanente Northern California, Redwood City, CA, USA
| | - Jie Yin
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Hélène Choquet
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Rebecca Kaye
- Southampton Eye Unit, University Hospital Southampton National Health Service Foundation Trust, Southampton, UK
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Karina Patasova
- Section of Ophthalmology, School of Life Course Sciences, King's College London, London, UK
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Praveen J Patel
- National Institute for Health and Care Research Biomedical Research Centre, Moorfields Eye Hospital National Health Service Foundation Trust, London, UK
- Institute of Ophthalmology, University College London, London, UK
| | - Brian L Yaspan
- Department of Human Genetics, Genentech, South San Francisco, CA, USA
| | | | - Pirro G Hysi
- Section of Ophthalmology, School of Life Course Sciences, King's College London, London, UK
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- UCL Great Ormond Street Institute of Child Health, King's College London, London, UK
- Sørlandet Sykehus Arendal, Arendal Hospital, Arendal, Norway
| | - Andrew J Lotery
- Southampton Eye Unit, University Hospital Southampton National Health Service Foundation Trust, Southampton, UK
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - J Michael Gaziano
- Million Veteran Program Coordinating Center, VA Boston Healthcare System, Boston, MA, USA
- Division of Aging, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Philip S Tsao
- VA Palo Alto Epidemiology Research and Information Center for Genomics, VA Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Steven J Fliesler
- Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, SUNY-University at Buffalo, Buffalo, NY, USA
- Research Service, VA Western NY Healthcare System, Buffalo, NY, USA
- Biochemistry, Jacobs School of Medicine and Biomedical Sciences, SUNY-University at Buffalo, Buffalo, NY, USA
- Graduate Program in Neurosciences, Jacobs School of Medicine and Biomedical Sciences, SUNY-University at Buffalo, Buffalo, NY, USA
| | - Jack M Sullivan
- Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, SUNY-University at Buffalo, Buffalo, NY, USA
- Research Service, VA Western NY Healthcare System, Buffalo, NY, USA
- Graduate Program in Neurosciences, Jacobs School of Medicine and Biomedical Sciences, SUNY-University at Buffalo, Buffalo, NY, USA
| | - Paul B Greenberg
- Section of Ophthalmology, VA Providence Healthcare System, Providence, RI, USA
- Division of Ophthalmology, Alpert Medical School, Brown University, Providence, RI, USA
| | - Wen-Chih Wu
- Section of Cardiology, Medical Service, VA Providence Healthcare System, Providence, RI, USA
- Division of Cardiology, Department of Medicine, Alpert Medical School, Brown University, Providence, RI, USA
| | - Themistocles L Assimes
- VA Palo Alto Epidemiology Research and Information Center for Genomics, VA Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Saiju Pyarajan
- Center for Data and Computational Sciences (C-DACS), VA Cooperative Studies Program, VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Panos Roussos
- Center for Disease Neurogenomics, Department of Psychiatry; Friedman Brain Institute; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Precision Medicine and Translational Therapeutics, VISN 2 Mental Illness Research, Education, and Clinical Center (MIRECC), James J. Peters Veterans Affairs Medical Center, New York/New Jersey VA Health Care Network, Bronx, NY, USA.
| | - Neal S Peachey
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH, USA.
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA.
| | - Sudha K Iyengar
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH, USA.
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA.
- Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA.
- Department of Genetics & Genome Sciences, Case Western Reserve University, Cleveland, OH, USA.
- Department of Ophthalmology and Visual Sciences, University Hospitals Eye Institute, Cleveland, OH, USA.
| |
Collapse
|
29
|
Lee D, Baek JH, Kim Y, Lee BD, Cho EY, Joo EJ, Ahn YM, Kim SH, Chung YC, Rami FZ, Kim SJ, Kim SW, Myung W, Ha TH, Lee HJ, Oh H, Lee KY, Kim MJ, Kang CY, Jeon S, Jo A, Yu H, Jeong S, Ha K, Kim B, Shim I, Cho C, Huang H, Won HH, Hong KS. Genome-wide association study and polygenic risk score analysis for schizophrenia in a Korean population. Asian J Psychiatr 2024; 102:104203. [PMID: 39293130 DOI: 10.1016/j.ajp.2024.104203] [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: 04/17/2024] [Revised: 08/13/2024] [Accepted: 08/28/2024] [Indexed: 09/20/2024]
Abstract
Although large-scale genome-wide association studies (GWASs) have revealed the genetic architecture of schizophrenia, these studies have mainly focused on populations of European ancestry. This study aimed to identify common genetic variants associated with schizophrenia in the Korean population and evaluate the performance of polygenic risk scores (PRSs) derived from large-scale GWASs across ancestries. In the Korean psychiatric GWAS project (KPGP), seven academic institutes and their affiliated hospitals across South Korea recruited a cohort of 1670 patients with DSM-IV-defined schizophrenia and 2271 healthy controls. A total of 6690,822 SNPs were tested for association with schizophrenia. We identified one previously unreported genome-wide significant locus rs2423464 (P = 2.83 × 10-11; odds ratio = 1.65; 95 % confidence interval = 1.43-1.91, minor allele frequency = 0.126). This variant was also associated with increased lysosomal-associated membrane protein family member 5 (LAMP5) gene expression. The PRS derived from the meta-analysis results of East Asian and European GWASs explained a larger proportion of the phenotypic variance in the Korean schizophrenia sample than the PRS of an East Asian or European GWAS. (R2 = 0.073 for meta-analysis; 0.028 for East Asian GWAS; 0.037 for European GWAS). GWASs involving diverse ethnic groups will expand our understanding of the genetic architecture of schizophrenia.
Collapse
Affiliation(s)
- Dongbin Lee
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Ji Hyun Baek
- Department of Psychiatry, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yujin Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Byung Dae Lee
- Pusan National University Research Park, Busan, South Korea
| | - Eun-Young Cho
- Samsung Biomedical Research Institute, Seoul, South Korea
| | - Eun-Jeong Joo
- Department of Psychiatry, Euijeongbu Eulji University Hospital, Euijeongbu, South Korea; Department of Neuropsychiatry, Eulji University, School of Medicine, Daejeon, South Korea
| | - Yong Min Ahn
- Department of Psychiatry, Seoul National University, College of Medicine, Seoul, South Korea; Department of Neuropsychiatry, Seoul National University Hospital, Seoul, South Korea
| | - Se Hyun Kim
- Department of Psychiatry, Seoul National University, College of Medicine, Seoul, South Korea; Department of Neuropsychiatry, Seoul National University Hospital, Seoul, South Korea
| | - Young-Chul Chung
- Department of Psychiatry, Jeonbuk National University Medical School, Jeonju, South Korea; Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea
| | - Fatima Zahra Rami
- Department of Psychiatry, Jeonbuk National University Medical School, Jeonju, South Korea; Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea
| | - Se Joo Kim
- Department of Psychiatry and Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung-Wan Kim
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, South Korea
| | - Woojae Myung
- Department of Psychiatry, Seoul National University, College of Medicine, Seoul, South Korea; Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Tae Hyon Ha
- Department of Psychiatry, Seoul National University, College of Medicine, Seoul, South Korea; Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Heon-Jeong Lee
- Department of Psychiatry, Korea University College of Medicine, Seoul, South Korea
| | - Hayoung Oh
- Pusan National University Research Park, Pusan, South Korea
| | - Kyu Young Lee
- Department of Neuropsychiatry, Eulji University, School of Medicine, Daejeon, South Korea; Department of Psychiatry, Nowon Eulji University Hospital, Seoul, South Korea
| | - Min Ji Kim
- Department of Psychiatry, Seoul National University, College of Medicine, Seoul, South Korea; Department of Neuropsychiatry, Seoul National University Hospital, Seoul, South Korea
| | - Chae Yeong Kang
- Department of Psychiatry, Jeonbuk National University Medical School, Jeonju, South Korea; Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea
| | - Sumoa Jeon
- Institute of Behavioral Science in Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Anna Jo
- Department of Psychiatry, Chonnam National University Medical School, Gwangju, South Korea
| | - Hyeona Yu
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Seunghwa Jeong
- Department of Psychiatry, Korea University College of Medicine, Seoul, South Korea
| | - Kyooseob Ha
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada; Department of Psychiatry, Lions Gate Hospital - Vancouver Coastal Health Authority, British Columbia, Canada
| | - Beomsu Kim
- Department of Precision Medicine, Sungkyunkwan University, School of Medicine, Seoul, South Korea
| | - Injeong Shim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Chamlee Cho
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea
| | - Hailiang Huang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| | - Hong-Hee Won
- Department of Digital Health, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Samsung Medical Center, Seoul, South Korea.
| | - Kyung Sue Hong
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada; Department of Psychiatry, Lions Gate Hospital - Vancouver Coastal Health Authority, British Columbia, Canada.
| |
Collapse
|
30
|
Liu X, Li D, Gao W, Liu H, Chen P, Zhao Y, Zhao W, Dong G. Shared genetic architecture between COVID-19 and irritable bowel syndrome: a large-scale genome-wide cross-trait analysis. Front Immunol 2024; 15:1442693. [PMID: 39620219 PMCID: PMC11604633 DOI: 10.3389/fimmu.2024.1442693] [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: 06/02/2024] [Accepted: 10/30/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND It has been reported that COVID-19 patients have an increased risk of developing IBS; however, the underlying genetic mechanisms of these associations remain largely unknown. The aim of this study was to investigate potential shared SNPs, genes, proteins, and biological pathways between COVID-19 and IBS by assessing pairwise genetic correlations and cross-trait genetic analysis. MATERIALS AND METHODS We assessed the genetic correlation between three COVID-19 phenotypes and IBS using linkage disequilibrium score regression (LDSC) and high-definition likelihood (HDL) methods. Two different sources of IBS data were combined using METAL, and the Multi-trait analysis of GWAS (MTAG) method was applied for multi-trait analysis to enhance statistical robustness and discover new genetic associations. Independent risk loci were examined using genome-wide complex trait analysis (GCTA)-conditional and joint analysis (COJO), multi-marker analysis of genomic annotation (MAGMA), and functional mapping and annotation (FUMA), integrating various QTL information and methods to further identify risk genes and proteins. Gene set variation analysis (GSVA) was employed to compute pleiotropic gene scores, and combined with immune infiltration algorithms, IBS patients were categorized into high and low immune infiltration groups. RESULTS We found a positive genetic correlation between COVID-19 infection, COVID-19 hospitalization, and IBS. Subsequent multi-trait analysis identified nine significantly associated genomic loci. Among these, eight genetic variants were closely related to the comorbidity of IBS and COVID-19. The study also highlighted four genes and 231 proteins associated with the susceptibility to IBS identified through various analytical strategies and a stratification approach for IBS risk populations. CONCLUSIONS Our study reveals a shared genetic architecture between these two diseases, providing new insights into potential biological mechanisms and laying the groundwork for more effective interventions.
Collapse
Affiliation(s)
- Xianqiang Liu
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Dingchang Li
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Wenxing Gao
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hao Liu
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Peng Chen
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yingjie Zhao
- Medical School of Chinese PLA, Beijing, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Wen Zhao
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Guanglong Dong
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
31
|
Wang S, Ojewunmi OO, Kamiza A, Ramsay M, Morris AP, Chikowore T, Fatumo S, Asimit JL. Accounting for heterogeneity due to environmental sources in meta-analysis of genome-wide association studies. Commun Biol 2024; 7:1512. [PMID: 39543362 PMCID: PMC11564974 DOI: 10.1038/s42003-024-07236-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024] Open
Abstract
Meta-analysis of genome-wide association studies (GWAS) across diverse populations offers power gains to identify loci associated with complex traits and diseases. Often heterogeneity in effect sizes across populations will be correlated with genetic ancestry and environmental exposures (e.g. lifestyle factors). We present an environment-adjusted meta-regression model (env-MR-MEGA) to detect genetic associations by adjusting for and quantifying environmental and ancestral heterogeneity between populations. In simulations, env-MR-MEGA has similar or greater association power than MR-MEGA, with notable gains when the environmental factor has a greater correlation with the trait than ancestry. In our analysis of low-density lipoprotein cholesterol in ~19,000 individuals across twelve sex-stratified GWAS from Africa, adjusting for sex, BMI, and urban status, we identify additional heterogeneity beyond ancestral effects for seven variants. Env-MR-MEGA provides an approach to account for environmental effects using summary-level data, making it a useful tool for meta-analyses without the need to share individual-level data.
Collapse
Affiliation(s)
- Siru Wang
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK.
| | - Oyesola O Ojewunmi
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Abram Kamiza
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- The African Computational Genomic (TACG) Research Group, MRC/UVRI and LSHTM, Entebbe, Uganda
- Malawi Epidemiology and Intervention Research Unit, Lilongwe, Malawi
| | - Michele Ramsay
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK
| | - Tinashe Chikowore
- MRC/Wits Developmental Pathways for Health Research Unit, Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Segun Fatumo
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
- The African Computational Genomic (TACG) Research Group, MRC/UVRI and LSHTM, Entebbe, Uganda
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
| | | |
Collapse
|
32
|
Liu X, Li D, Zhang Y, Liu H, Chen P, Zhao Y, Ruscitti P, Zhao W, Dong G. Identifying Common Genetic Etiologies Between Inflammatory Bowel Disease and Related Immune-Mediated Diseases. Biomedicines 2024; 12:2562. [PMID: 39595128 PMCID: PMC11592296 DOI: 10.3390/biomedicines12112562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/25/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Patients with inflammatory bowel disease (IBD) have an increased risk of developing immune-mediated diseases. However, the genetic basis of IBD is complex, and an integrated approach should be used to elucidate the complex genetic relationship between IBD and immune-mediated diseases. METHODS The genetic relationship between IBD and 16 immune-mediated diseases was examined using linkage disequilibrium score regression. GWAS data were synthesized from two IBD databases using the METAL, and multi-trait analysis of genome-wide association studies was performed to enhance statistical robustness and identify novel genetic associations. Independent risk loci were meticulously examined using conditional and joint genome-wide multi-trait analysis, multi-marker analysis of genomic annotation, and functional mapping and annotation of significant genetic loci, integrating the information of quantitative trait loci and different methodologies to identify risk-related genes and proteins. RESULTS The results revealed four immune-mediated diseases (AS, psoriasis, iridocyclitis, and PsA) with a significant relationship with IBD. The multi-trait analysis revealed 909 gene loci of statistical significance. Of these loci, 28 genetic variants were closely related to IBD, and 7 single-nucleotide polymorphisms represented novel independent risk loci. In addition, 14 genes and 514 proteins were found to be associated with susceptibility to immune-mediated diseases. Notably, IL1RL1 emerged as a key player, present within pleiotropic genes across multiple protein databases, highlighting its potential as a therapeutic target. CONCLUSIONS This study suggests that the common polygenic determinants between IBD and immune-mediated diseases are widely distributed across the genome. The findings not only support a shared genetic relationship between IBD and immune-mediated diseases but also provide novel therapeutic targets for these diseases.
Collapse
Affiliation(s)
- Xianqiang Liu
- Medical School of Chinese PLA, Beijing 100853, China; (X.L.)
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Dingchang Li
- Medical School of Chinese PLA, Beijing 100853, China; (X.L.)
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Yue Zhang
- Medical School of Chinese PLA, Beijing 100853, China; (X.L.)
| | - Hao Liu
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Peng Chen
- Medical School of Chinese PLA, Beijing 100853, China; (X.L.)
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Yingjie Zhao
- Medical School of Chinese PLA, Beijing 100853, China; (X.L.)
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Piero Ruscitti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Wen Zhao
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Guanglong Dong
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, No. 28 Fuxing Road, Haidian District, Beijing 100853, China
| |
Collapse
|
33
|
Patel Y, Shin J, Sliz E, Tang A, Mishra A, Xia R, Hofer E, Rajula HSR, Wang R, Beyer F, Horn K, Riedl M, Yu J, Völzke H, Bülow R, Völker U, Frenzel S, Wittfeld K, Van der Auwera S, Mosley TH, Bouteloup V, Lambert JC, Chêne G, Dufouil C, Tzourio C, Mangin JF, Gottesman RF, Fornage M, Schmidt R, Yang Q, Witte V, Scholz M, Loeffler M, Roshchupkin GV, Ikram MA, Grabe HJ, Seshadri S, Debette S, Paus T, Pausova Z. Genetic risk factors underlying white matter hyperintensities and cortical atrophy. Nat Commun 2024; 15:9517. [PMID: 39496600 PMCID: PMC11535513 DOI: 10.1038/s41467-024-53689-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 10/18/2024] [Indexed: 11/06/2024] Open
Abstract
White matter hyperintensities index structural abnormalities in the cerebral white matter, including axonal damage. The latter may promote atrophy of the cerebral cortex, a key feature of dementia. Here, we report a study of 51,065 individuals from 10 cohorts demonstrating that higher white matter hyperintensity volume associates with lower cortical thickness. The meta-GWAS of white matter hyperintensities-associated cortical 'atrophy' identifies 20 genome-wide significant loci, and enrichment in genes specific to vascular cell types, astrocytes, and oligodendrocytes. White matter hyperintensities-associated cortical 'atrophy' showed positive genetic correlations with vascular-risk traits and plasma biomarkers of neurodegeneration, and negative genetic correlations with cognitive functioning. 15 of the 20 loci regulated the expression of 54 genes in the cerebral cortex that, together with their co-expressed genes, were enriched in biological processes of axonal cytoskeleton and intracellular transport. The white matter hyperintensities-cortical thickness associations were most pronounced in cortical regions with higher expression of genes specific to excitatory neurons with long-range axons traversing through the white matter. The meta-GWAS-based polygenic risk score predicts vascular and all-cause dementia in an independent sample of 500,348 individuals. Thus, the genetics of white matter hyperintensities-related cortical atrophy involves vascular and neuronal processes and increases dementia risk.
Collapse
Affiliation(s)
- Yash Patel
- The Hospital for Sick Children, Toronto, Ontario, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Jean Shin
- The Hospital for Sick Children, Toronto, Ontario, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Eeva Sliz
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Ariana Tang
- The Hospital for Sick Children, Toronto, Ontario, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Aniket Mishra
- University of Bordeaux, INSERM, Bordeaux Population Health research center, UMR1219, Bordeaux, France
| | - Rui Xia
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Edith Hofer
- Institut für Medizinische Informatik, Statistik und Dokumentation, Graz, Austria
- Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
| | - Hema Sekhar Reddy Rajula
- University of Bordeaux, INSERM, Bordeaux Population Health research center, UMR1219, Bordeaux, France
| | - Ruiqi Wang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Frauke Beyer
- University of Bordeaux, INSERM, Bordeaux Population Health research center, UMR1219, Bordeaux, France
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Katrin Horn
- Institute for Medical Informatics, Statistics and Epidemiology; Leipzig University, Leipzig, Germany
| | - Max Riedl
- Institute for Medical Informatics, Statistics and Epidemiology; Leipzig University, Leipzig, Germany
| | - Jing Yu
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Robin Bülow
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Stefan Frenzel
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Sandra Van der Auwera
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
| | - Thomas H Mosley
- The MIND Center, The University of Mississippi Medical Center, Jackson, MS, USA
| | - Vincent Bouteloup
- University of Bordeaux, INSERM, Bordeaux Population Health research center, UMR1219, Bordeaux, France
- CHU Bordeaux, CIC 1401 EC, Pôle Santé Publique, Bordeaux, France
| | - Jean-Charles Lambert
- U1167-RID-AGE facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, INSERM, CHU Lille, Institut Pasteur de Lille, University of Lille, Lille, France
| | - Geneviève Chêne
- University of Bordeaux, INSERM, Bordeaux Population Health research center, UMR1219, Bordeaux, France
- Department of Public Health, CHU de Bordeaux, Bordeaux, France
| | - Carole Dufouil
- University of Bordeaux, INSERM, Bordeaux Population Health research center, UMR1219, Bordeaux, France
| | - Christophe Tzourio
- University of Bordeaux, INSERM, Bordeaux Population Health research center, UMR1219, Bordeaux, France
- Department of Public Health, CHU de Bordeaux, Bordeaux, France
| | | | - Rebecca F Gottesman
- National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, Maryland, USA
| | - Myriam Fornage
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Reinhold Schmidt
- Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Veronica Witte
- Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology; Leipzig University, Leipzig, Germany
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology; Leipzig University, Leipzig, Germany
- Leipzig Research Centre for Civilization Diseases; Leipzig University, Leipzig, Germany
| | - Gennady V Roshchupkin
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Hans J Grabe
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | | | - Stephanie Debette
- University of Bordeaux, INSERM, Bordeaux Population Health research center, UMR1219, Bordeaux, France
- Bordeaux University Hospital, Department of Neurology, Institute for Neurodegenerative Diseases, Bordeaux, France
| | - Tomas Paus
- Centre hospitalier universitaire Sainte-Justine, University of Montreal, Montreal, Canada.
- Departments of Psychiatry and Neuroscience, Faculty of Medicine, University of Montreal, Montreal, Canada.
- Department of Psychiatry, McGill University, Montreal, Canada.
- ECOGENE-21, Chicoutimi, Canada.
| | - Zdenka Pausova
- The Hospital for Sick Children, Toronto, Ontario, Canada.
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada.
- Centre hospitalier universitaire Sainte-Justine, University of Montreal, Montreal, Canada.
- ECOGENE-21, Chicoutimi, Canada.
- Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, Canada.
| |
Collapse
|
34
|
Cheng H, Zhang ZY, Han H, Wei R, Zhao W, Sun YC, Xu BB, Hou XL, Wang JL, He YQ, Fu Y, Wang QS, Pan YC, Zhang Z, Wang Z. Cross-ancestry meta-genome-wide association studies provide insights to the understanding of semen traits in pigs. Animal 2024; 18:101331. [PMID: 39405960 DOI: 10.1016/j.animal.2024.101331] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 11/18/2024] Open
Abstract
Semen traits play a crucial role in pig reproduction and fertility. However, limited data availability hinder a comprehensive understanding of the genetic mechanisms underlying these traits. In this study, we integrated 597 299 ejaculates and 3 596 sequence data to identify genetic variants and candidate genes related to four semen traits, including sperm progressive motility (MOT), semen volume, sperm concentration (CON), and effective sperm count (SUM). A cross-ancestry meta-genome-wide association study was conducted to detect 163 lead single nucleotide polymorphisms (SNPs) associated with MOT, CON, and SUM. Subsequently, transcriptome-wide association studies and colocalisation analyses were integrated to identify 176 candidate genes, many of which have documented roles in spermatogenesis or male mammal semen traits. Our analysis highlighted the potential involvement of CSM5, PDZD9, and LDAF1 in regulating semen traits through multiple methods. Finally, to validate the function of significant SNPs, we performed genomic feature best linear unbiased prediction in 348 independent pigs using identified trait-related SNP subsets as genomic features. We found that integrating the top 0.1, 1, and 5% significant SNPs as genomic features could enhance genomic prediction accuracy for CON and MOT compared to traditional genomic best linear unbiased prediction. This study contributes to a comprehensive understanding of the genetic mechanisms of boar semen traits and provides insight for developing genomic selection models.
Collapse
Affiliation(s)
- H Cheng
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China
| | - Z Y Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China
| | - H Han
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China
| | - R Wei
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China
| | - W Zhao
- SciGene Biotechnology Co., Ltd., Hefei 230031, China
| | - Y C Sun
- Haidian Foreign Language Academy, Beijing 100195, China
| | - B B Xu
- SciGene Biotechnology Co., Ltd., Hefei 230031, China
| | - X L Hou
- SciGene Biotechnology Co., Ltd., Hefei 230031, China
| | - J L Wang
- SciGene Biotechnology Co., Ltd., Hefei 230031, China
| | - Y Q He
- SciGene Biotechnology Co., Ltd., Hefei 230031, China
| | - Y Fu
- SciGene Biotechnology Co., Ltd., Hefei 230031, China
| | - Q S Wang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China; Hainan Institute, Zhejiang University, Yongyou Industry Park, Yazhou Bay Sci-Tech City, Sanya 572000, China
| | - Y C Pan
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China; Hainan Institute, Zhejiang University, Yongyou Industry Park, Yazhou Bay Sci-Tech City, Sanya 572000, China
| | - Z Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China
| | - Z Wang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Livestock and Poultry Resources Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Hangzhou 310058, China.
| |
Collapse
|
35
|
Jones AC, Patki A, Srinivasasainagendra V, Tiwari HK, Armstrong ND, Chaudhary NS, Limdi NA, Hidalgo BA, Davis B, Cimino JJ, Khan A, Kiryluk K, Lange LA, Lange EM, Arnett DK, Young BA, Diamantidis CJ, Franceschini N, Wassertheil-Smoller S, Rich SS, Rotter JI, Mychaleckyj JC, Kramer HJ, Chen YDI, Psaty BM, Brody JA, de Boer IH, Bansal N, Bis JC, Irvin MR. Single-Ancestry versus Multi-Ancestry Polygenic Risk Scores for CKD in Black American Populations. J Am Soc Nephrol 2024; 35:1558-1569. [PMID: 39073889 PMCID: PMC11543021 DOI: 10.1681/asn.0000000000000437] [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: 01/22/2024] [Accepted: 06/28/2024] [Indexed: 07/31/2024] Open
Abstract
Key Points The predictive performance of an African ancestry–specific polygenic risk score (PRS) was comparable to a European ancestry–derived PRS for kidney traits. However, multi-ancestry PRSs outperform single-ancestry PRSs in Black American populations. Predictive accuracy of PRSs for CKD was improved with the use of race-free eGFR. Background CKD is a risk factor of cardiovascular disease and early death. Recently, polygenic risk scores (PRSs) have been developed to quantify risk for CKD. However, African ancestry populations are underrepresented in both CKD genetic studies and PRS development overall. Moreover, European ancestry–derived PRSs demonstrate diminished predictive performance in African ancestry populations. Methods This study aimed to develop a PRS for CKD in Black American populations. We obtained score weights from a meta-analysis of genome-wide association studies for eGFR in the Million Veteran Program and Reasons for Geographic and Racial Differences in Stroke Study to develop an eGFR PRS. We optimized the PRS risk model in a cohort of participants from the Hypertension Genetic Epidemiology Network. Validation was performed in subsets of Black participants of the Trans-Omics in Precision Medicine Consortium and Genetics of Hypertension Associated Treatment Study. Results The prevalence of CKD—defined as stage 3 or higher—was associated with the PRS as a continuous predictor (odds ratio [95% confidence interval]: 1.35 [1.08 to 1.68]) and in a threshold-dependent manner. Furthermore, including APOL1 risk status—a putative variant for CKD with higher prevalence among those of sub-Saharan African descent—improved the score's accuracy. PRS associations were robust to sensitivity analyses accounting for traditional CKD risk factors, as well as CKD classification based on prior eGFR equations. Compared with previously published PRS, the predictive performance of our PRS was comparable with a European ancestry–derived PRS for kidney traits. However, single-ancestry PRSs were less predictive than multi-ancestry–derived PRSs. Conclusions In this study, we developed a PRS that was significantly associated with CKD with improved predictive accuracy when including APOL1 risk status. However, PRS generated from multi-ancestry populations outperformed single-ancestry PRS in our study.
Collapse
Affiliation(s)
- Alana C. Jones
- Medical Scientist Training Program, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama
| | - Amit Patki
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama
| | - Vinodh Srinivasasainagendra
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama
| | - Hemant K. Tiwari
- Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama
| | - Nicole D. Armstrong
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ninad S. Chaudhary
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama
| | - Nita A. Limdi
- Department of Neurology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Bertha A. Hidalgo
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama
| | - Brittney Davis
- Department of Neurology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - James J. Cimino
- Department of Biomedical Informatics and Data Science, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Atlas Khan
- Division of Nephrology, Department of Medicine, Columbia University Medical Center, New York, New York
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, Columbia University Medical Center, New York, New York
| | - Leslie A. Lange
- Department of Biomedical Informatics, University of Colorado-Anschutz, Aurora, Colorado
| | - Ethan M. Lange
- Department of Biomedical Informatics, University of Colorado-Anschutz, Aurora, Colorado
| | - Donna K. Arnett
- Office of the Provost, University of South Carolina, Columbia, South Carolina
| | - Bessie A. Young
- Division of Nephrology, University of Washington, Seattle, Washington
| | | | - Nora Franceschini
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Sylvia Wassertheil-Smoller
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, New York, New York
| | - Stephen S. Rich
- Department of Genome Sciences, University of Virginia, Charlottesville, Virginia
| | - Jerome I. Rotter
- Department of Pediatrics, The Institute for Translational Genomic and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbort-UCLA Medical Center, Torrance, California
| | - Josyf C. Mychaleckyj
- Department of Genome Sciences, University of Virginia, Charlottesville, Virginia
| | - Holly J. Kramer
- Departments of Public Health Sciences and Medicine, Loyola University Medical Center, Taywood, Illinois
| | - Yii-Der I. Chen
- Department of Pediatrics, The Institute for Translational Genomic and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbort-UCLA Medical Center, Torrance, California
| | - Bruce M. Psaty
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, Washington
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington
| | - Jennifer A. Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington
| | - Ian H. de Boer
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, Washington
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington
| | - Nisha Bansal
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington
| | - Joshua C. Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington
| | - Marguerite R. Irvin
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
36
|
Leventhal MJ, Zanella CA, Kang B, Peng J, Gritsch D, Liao Z, Bukhari H, Wang T, Pao PC, Danquah S, Benetatos J, Nehme R, Farhi S, Tsai LH, Dong X, Scherzer CR, Feany MB, Fraenkel E. An integrative systems-biology approach defines mechanisms of Alzheimer's disease neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.17.585262. [PMID: 38559190 PMCID: PMC10980014 DOI: 10.1101/2024.03.17.585262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Despite years of intense investigation, the mechanisms underlying neuronal death in Alzheimer's disease, the most common neurodegenerative disorder, remain incompletely understood. To define relevant pathways, we integrated the results of an unbiased, genome-scale forward genetic screen for age-associated neurodegeneration in Drosophila with human and Drosophila Alzheimer's disease-associated multi-omics. We measured proteomics, phosphoproteomics, and metabolomics in Drosophila models of Alzheimer's disease and identified Alzheimer's disease human genetic variants that modify expression in disease-vulnerable neurons. We used a network optimization approach to integrate these data with previously published Alzheimer's disease multi-omic data. We computationally predicted and experimentally demonstrated how HNRNPA2B1 and MEPCE enhance tau-mediated neurotoxicity. Furthermore, we demonstrated that the screen hits CSNK2A1 and NOTCH1 regulate DNA damage in Drosophila and human iPSC-derived neural progenitor cells. Our work identifies candidate pathways that could be targeted to ameliorate neurodegeneration in Alzheimer's disease.
Collapse
Affiliation(s)
- Matthew J Leventhal
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Camila A Zanella
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Byunguk Kang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
| | - Jiajie Peng
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - David Gritsch
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Zhixiang Liao
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Hassan Bukhari
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Tao Wang
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Present address: School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Ping-Chieh Pao
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Serwah Danquah
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Joseph Benetatos
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ralda Nehme
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Samouil Farhi
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
| | - Li-Huei Tsai
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Xianjun Dong
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Clemens R Scherzer
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Present address: Stephen and Denise Adams Center of Yale School of Medicine, CT, USA
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ernest Fraenkel
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Lead contact
| |
Collapse
|
37
|
Gorman BR, Ji SG, Francis M, Sendamarai AK, Shi Y, Devineni P, Saxena U, Partan E, DeVito AK, Byun J, Han Y, Xiao X, Sin DD, Timens W, Moser J, Muralidhar S, Ramoni R, Hung RJ, McKay JD, Bossé Y, Sun R, Amos CI, Pyarajan S. Multi-ancestry GWAS meta-analyses of lung cancer reveal susceptibility loci and elucidate smoking-independent genetic risk. Nat Commun 2024; 15:8629. [PMID: 39366959 PMCID: PMC11452618 DOI: 10.1038/s41467-024-52129-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 08/27/2024] [Indexed: 10/06/2024] Open
Abstract
Lung cancer remains the leading cause of cancer mortality, despite declining smoking rates. Previous lung cancer GWAS have identified numerous loci, but separating the genetic risks of lung cancer and smoking behavioral susceptibility remains challenging. Here, we perform multi-ancestry GWAS meta-analyses of lung cancer using the Million Veteran Program cohort (approximately 95% male cases) and a previous study of European-ancestry individuals, jointly comprising 42,102 cases and 181,270 controls, followed by replication in an independent cohort of 19,404 cases and 17,378 controls. We then carry out conditional meta-analyses on cigarettes per day and identify two novel, replicated loci, including the 19p13.11 pleiotropic cancer locus in squamous cell lung carcinoma. Overall, we report twelve novel risk loci for overall lung cancer, lung adenocarcinoma, and squamous cell lung carcinoma, nine of which are externally replicated. Finally, we perform PheWAS on polygenic risk scores for lung cancer, with and without conditioning on smoking. The unconditioned lung cancer polygenic risk score is associated with smoking status in controls, illustrating a reduced predictive utility in non-smokers. Additionally, our polygenic risk score demonstrates smoking-independent pleiotropy of lung cancer risk across neoplasms and metabolic traits.
Collapse
Affiliation(s)
- Bryan R Gorman
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, USA
- Booz Allen Hamilton, McLean, VA, USA
| | - Sun-Gou Ji
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, USA
- BridgeBio Pharma, Palo Alto, CA, USA
| | - Michael Francis
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, USA
- Booz Allen Hamilton, McLean, VA, USA
| | - Anoop K Sendamarai
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, USA
- Carbone Cancer Center, University of Wisconsin, Madison, WI, USA
| | - Yunling Shi
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, USA
| | - Poornima Devineni
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, USA
| | - Uma Saxena
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, USA
| | - Elizabeth Partan
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, USA
| | - Andrea K DeVito
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, USA
- Booz Allen Hamilton, McLean, VA, USA
| | - Jinyoung Byun
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Younghun Han
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Xiangjun Xiao
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Don D Sin
- The University of British Columbia Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, Canada
| | - Wim Timens
- University Medical Centre Groningen, GRIAC (Groningen Research Institute for Asthma and COPD), University of Groningen, Groningen, Netherlands
- Department of Pathology & Medical Biology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Jennifer Moser
- Office of Research and Development, Department of Veterans Affairs, Washington, DC, USA
| | - Sumitra Muralidhar
- Office of Research and Development, Department of Veterans Affairs, Washington, DC, USA
| | - Rachel Ramoni
- Office of Research and Development, Department of Veterans Affairs, Washington, DC, USA
| | - Rayjean J Hung
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, University of Toronto, Toronto, ON, Canada
| | - James D McKay
- Section of Genetics, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Yohan Bossé
- Institut universitaire de cardiologie et de pneumologie de Québec, Department of Molecular Medicine, Laval University, Quebec City, QC, Canada
| | - Ryan Sun
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher I Amos
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Saiju Pyarajan
- Center for Data and Computational Sciences (C-DACS), VA Boston Healthcare System, Boston, MA, USA.
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
38
|
Fu S, Wheeler W, Wang X, Hua X, Godbole D, Duan J, Zhu B, Deng L, Qin F, Zhang H, Shi J, Yu K. A comprehensive framework for trans-ancestry pathway analysis using GWAS summary data from diverse populations. PLoS Genet 2024; 20:e1011322. [PMID: 39441834 PMCID: PMC11534268 DOI: 10.1371/journal.pgen.1011322] [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: 05/30/2024] [Revised: 11/04/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024] Open
Abstract
As more multi-ancestry GWAS summary data become available, we have developed a comprehensive trans-ancestry pathway analysis framework that effectively utilizes this diverse genetic information. Within this framework, we evaluated various strategies for integrating genetic data at different levels-SNP, gene, and pathway-from multiple ancestry groups. Through extensive simulation studies, we have identified robust strategies that demonstrate superior performance across diverse scenarios. Applying these methods, we analyzed 6,970 pathways for their association with schizophrenia, incorporating data from African, East Asian, and European populations. Our analysis identified over 200 pathways significantly associated with schizophrenia, even after excluding genes near genome-wide significant loci. This approach substantially enhances detection efficiency compared to traditional single-ancestry pathway analysis and the conventional approach that amalgamates single-ancestry pathway analysis results across different ancestry groups. Our framework provides a flexible and effective tool for leveraging the expanding pool of multi-ancestry GWAS summary data, thereby improving our ability to identify biologically relevant pathways that contribute to disease susceptibility.
Collapse
Affiliation(s)
- Sheng Fu
- School of Statistics and Data Science, Nankai University, Tianjin, China
- Key Laboratory of Pure Mathematics and Combinatorics, Nankai University, Tianjin, China
| | - William Wheeler
- Information Management Services, Inc, Bethesda, Maryland, United States of America
| | - Xiaoyu Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc, Rockville, Maryland, United States of America
| | - Xing Hua
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc, Rockville, Maryland, United States of America
| | - Devika Godbole
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc, Rockville, Maryland, United States of America
| | - Jubao Duan
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, Illinois, United States of America
- Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, Illinois, United States of America
| | - Bin Zhu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Lu Deng
- School of Statistics and Data Science, Nankai University, Tianjin, China
| | - Fei Qin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Haoyu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Jianxin Shi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Kai Yu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| |
Collapse
|
39
|
Mugo JW, Day C, Choudhury A, Deetlefs M, Freercks R, Geraty S, Panieri A, Cotchbos C, Ribeiro M, Engelbrecht A, Lisa K. Micklesfield, Ramsay M, Sarah P, Peter J. A GWAS of ACE Inhibitor-Induced Angioedema in a South African Population. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.13.24313664. [PMID: 39314982 PMCID: PMC11419215 DOI: 10.1101/2024.09.13.24313664] [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: 09/25/2024]
Abstract
Background Angiotensin-converting enzyme inhibitor-induced angioedema (AE-ACEI) is a life-threatening adverse event and, globally, the commonest cause of emergency presentations with angioedema. Several large genome-wide association studies (GWAS) have found genomic associations with AE-ACEI. However, despite African Americans having a 5-fold increased risk of AE-ACEI, there are no published GWAS from Africa. The aim of this study was to conduct a case-control GWAS of AE-ACEI in a South African population and perform a meta-analysis with an African American and European American population. Methods The GWAS included 202 South African adults with a history of AE-ACEI and 513 controls without angioedema following angiotensin-converting enzyme inhibitor (ACEI) treatment for at least 2 years. A meta-analysis was conducted with GWAS summary statistics from an African American and European American cohort (from Vanderbilt/Marshfield with 174 cases and 489 controls). Results No SNPs attained genome-wide significance. However, 26 SNPs in the post-imputation standard GWAS of the South African cohort and 37 SNPs in the meta-analysis were associated to AE-ACEI with suggestive threshold(p-value<5.0×10-06). Some of these SNPs were found to be located close to the genes PRKCQ and RIMS1, previously linked with drug-induced angioedema, and also close to the CSMD1 gene linked to ACEI cough, providing replication at the gene level, but with novel lead SNPs. Conclusions Our results highlight the importance of African populations to detect novel variants in replication studies. Further increased sampling across the continent and matched functional work are needed to confirm the importance of genetic variation in understanding the biology of AE-ACEI.
Collapse
Affiliation(s)
- Jacquiline W. Mugo
- Division of Allergy and Clinical Immunology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town, 7625, Western Cape, South Africa
| | - Cascia Day
- Division of Allergy and Clinical Immunology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town, 7625, Western Cape, South Africa
- Allergy and Immunology Unit, University of Cape Town Lung Institute (Pty) Ltd, George Street, Mowbray, Cape Town, 7700, Western Cape, South Africa
| | - Ananyo Choudhury
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, 1 Jan Smuts Avenue, Braamfontein, Johannesburg, 2000, South Africa
| | - Maria Deetlefs
- Division of Allergy and Clinical Immunology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town, 7625, Western Cape, South Africa
| | - Robert Freercks
- Faculty of Health Sciences, Department of Medicine, Nelson Mandela University, Gqeberha, South Africa
| | - Sian Geraty
- Faculty of Health Sciences, Department of Medicine, Nelson Mandela University, Gqeberha, South Africa
| | - Angelica Panieri
- Faculty of Health Sciences, Department of Medicine, Nelson Mandela University, Gqeberha, South Africa
| | - Christian Cotchbos
- Faculty of Health Sciences, Department of Medicine, Nelson Mandela University, Gqeberha, South Africa
| | - Melissa Ribeiro
- Allergy and Immunology Unit, University of Cape Town Lung Institute (Pty) Ltd, George Street, Mowbray, Cape Town, 7700, Western Cape, South Africa
| | - Adelein Engelbrecht
- Western Cape Department of Health, District 6 Day Hospital, 50 Caledon Street, Zonnebloem, Cape Town, Western Cape, South Africa
| | - Lisa K. Micklesfield
- South African Medical Research Council/Wits Developmental Pathways for Health Research Unit (DPHRU), Department of Paediatrics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Michèle Ramsay
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, 1 Jan Smuts Avenue, Braamfontein, Johannesburg, 2000, South Africa
| | - Pedretti Sarah
- Allergy and Immunology Unit, University of Cape Town Lung Institute (Pty) Ltd, George Street, Mowbray, Cape Town, 7700, Western Cape, South Africa
| | - Jonny Peter
- Division of Allergy and Clinical Immunology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town, 7625, Western Cape, South Africa
- Allergy and Immunology Unit, University of Cape Town Lung Institute (Pty) Ltd, George Street, Mowbray, Cape Town, 7700, Western Cape, South Africa
| |
Collapse
|
40
|
Akgun B, Feliciano-Astacio BE, Hamilton-Nelson KL, Scott K, Rivero J, Adams LD, Sanchez JJ, Valladares GS, Tejada S, Bussies PL, Silva-Vergara C, Rodriguez VC, Mena PR, Celis K, Whitehead PG, Prough M, Kosanovic C, Van Booven DJ, Schmidt MA, Acosta H, Griswold AJ, Dalgard CL, McInerney KF, Beecham GW, Cuccaro ML, Vance JM, Pericak-Vance MA, Rajabli F. Genome-wide association analysis and admixture mapping in a Puerto Rican cohort supports an Alzheimer disease risk locus on chromosome 12. Front Aging Neurosci 2024; 16:1459796. [PMID: 39295643 PMCID: PMC11408238 DOI: 10.3389/fnagi.2024.1459796] [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: 07/04/2024] [Accepted: 08/26/2024] [Indexed: 09/21/2024] Open
Abstract
Introduction Hispanic/Latino populations are underrepresented in Alzheimer Disease (AD) genetic studies. Puerto Ricans (PR), a three-way admixed (European, African, and Amerindian) population is the second-largest Hispanic group in the continental US. We aimed to conduct a genome-wide association study (GWAS) and comprehensive analyses to identify novel AD susceptibility loci and characterize known AD genetic risk loci in the PR population. Materials and methods Our study included Whole Genome Sequencing (WGS) and phenotype data from 648 PR individuals (345 AD, 303 cognitively unimpaired). We used a generalized linear-mixed model adjusting for sex, age, population substructure, and genetic relationship matrix. To infer local ancestry, we merged the dataset with the HGDP/1000G reference panel. Subsequently, we conducted univariate admixture mapping (AM) analysis. Results We identified suggestive signals within the SLC38A1 and SCN8A genes on chromosome 12q13. This region overlaps with an area of linkage of AD in previous studies (12q13) in independent data sets further supporting. Univariate African AM analysis identified one suggestive ancestral block (p = 7.2×10-6) located in the same region. The ancestry-aware approach showed that this region has both European and African ancestral backgrounds and both contributing to the risk in this region. We also replicated 11 different known AD loci -including APOE- identified in mostly European studies, which is likely due to the high European background of the PR population. Conclusion PR GWAS and AM analysis identified a suggestive AD risk locus on chromosome 12, which includes the SLC38A1 and SCN8A genes. Our findings demonstrate the importance of designing GWAS and ancestry-aware approaches and including underrepresented populations in genetic studies of AD.
Collapse
Affiliation(s)
- Bilcag Akgun
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| | | | - Kara L Hamilton-Nelson
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Kyle Scott
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Joe Rivero
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Larry D Adams
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Jose J Sanchez
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Glenies S Valladares
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Sergio Tejada
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Parker L Bussies
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Concepcion Silva-Vergara
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Vanessa C Rodriguez
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Pedro R Mena
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Katrina Celis
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Patrice G Whitehead
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Michael Prough
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Christina Kosanovic
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Derek J Van Booven
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Michael A Schmidt
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
| | | | - Anthony J Griswold
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Clifton L Dalgard
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Katalina F McInerney
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Gary W Beecham
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Michael L Cuccaro
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Jeffery M Vance
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Margaret A Pericak-Vance
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Farid Rajabli
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
41
|
Yuan K, Longchamps RJ, Pardiñas AF, Yu M, Chen TT, Lin SC, Chen Y, Lam M, Liu R, Xia Y, Guo Z, Shi W, Shen C, Daly MJ, Neale BM, Feng YCA, Lin YF, Chen CY, O'Donovan MC, Ge T, Huang H. Fine-mapping across diverse ancestries drives the discovery of putative causal variants underlying human complex traits and diseases. Nat Genet 2024; 56:1841-1850. [PMID: 39187616 PMCID: PMC11888783 DOI: 10.1038/s41588-024-01870-z] [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: 01/10/2023] [Accepted: 07/15/2024] [Indexed: 08/28/2024]
Abstract
Genome-wide association studies (GWAS) of human complex traits or diseases often implicate genetic loci that span hundreds or thousands of genetic variants, many of which have similar statistical significance. While statistical fine-mapping in individuals of European ancestry has made important discoveries, cross-population fine-mapping has the potential to improve power and resolution by capitalizing on the genomic diversity across ancestries. Here we present SuSiEx, an accurate and computationally efficient method for cross-population fine-mapping. SuSiEx integrates data from an arbitrary number of ancestries, explicitly models population-specific allele frequencies and linkage disequilibrium patterns, accounts for multiple causal variants in a genomic region and can be applied to GWAS summary statistics. We comprehensively assessed the performance of SuSiEx using simulations. We further showed that SuSiEx improves the fine-mapping of a range of quantitative traits available in both the UK Biobank and Taiwan Biobank, and improves the fine-mapping of schizophrenia-associated loci by integrating GWAS across East Asian and European ancestries.
Collapse
Affiliation(s)
- Kai Yuan
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, the Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Ryan J Longchamps
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, the Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Antonio F Pardiñas
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Mingrui Yu
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, the Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Tzu-Ting Chen
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Shu-Chin Lin
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Yu Chen
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, the Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Medical Genetics & Hunan Key Laboratory of Medical Genetics, School of Life Sciences and Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Max Lam
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Human Genetics, Genome Institute of Singapore, Singapore, Singapore
- Division of Psychiatry Research, the Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY, USA
- Research Division Institute of Mental Health Singapore, Singapore, Singapore
| | - Ruize Liu
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, the Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Yan Xia
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, the Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Zhenglin Guo
- Stanley Center for Psychiatric Research, the Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Wenzhao Shi
- Digital Health China Technologies Corp. Ltd, Beijing, China
| | - Chengguo Shen
- Digital Health China Technologies Corp. Ltd, Beijing, China
| | - Mark J Daly
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, the Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Benjamin M Neale
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, the Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yen-Chen A Feng
- Institute of Health Data Analytics and Statistics, College of Public Health, National Taiwan University, Taipei, Taiwan
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Yen-Feng Lin
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
- Department of Public Health & Medical Humanities, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Behavioral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | - Michael C O'Donovan
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Tian Ge
- Stanley Center for Psychiatric Research, the Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.
| | - Hailiang Huang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA.
- Stanley Center for Psychiatric Research, the Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
42
|
Shen J, Jiang L, Wang K, Wang A, Chen F, Newcombe PJ, Haiman CA, Conti DV. Hierarchical joint analysis of marginal summary statistics-Part I: Multipopulation fine mapping and credible set construction. Genet Epidemiol 2024; 48:241-257. [PMID: 38606643 PMCID: PMC11980956 DOI: 10.1002/gepi.22562] [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: 09/27/2023] [Revised: 02/27/2024] [Accepted: 03/27/2024] [Indexed: 04/13/2024]
Abstract
Recent advancement in genome-wide association studies (GWAS) comes from not only increasingly larger sample sizes but also the shift in focus towards underrepresented populations. Multipopulation GWAS increase power to detect novel risk variants and improve fine-mapping resolution by leveraging evidence and differences in linkage disequilibrium (LD) from diverse populations. Here, we expand upon our previous approach for single-population fine-mapping through Joint Analysis of Marginal SNP Effects (JAM) to a multipopulation analysis (mJAM). Under the assumption that true causal variants are common across studies, we implement a hierarchical model framework that conditions on multiple SNPs while explicitly incorporating the different LD structures across populations. The mJAM framework can be used to first select index variants using the mJAM likelihood with different feature selection approaches. In addition, we present a novel approach leveraging the ideas of mediation to construct credible sets for these index variants. Construction of such credible sets can be performed given any existing index variants. We illustrate the implementation of the mJAM likelihood through two implementations: mJAM-SuSiE (a Bayesian approach) and mJAM-Forward selection. Through simulation studies based on realistic effect sizes and levels of LD, we demonstrated that mJAM performs well for constructing concise credible sets that include the underlying causal variants. In real data examples taken from the most recent multipopulation prostate cancer GWAS, we showed several practical advantages of mJAM over other existing multipopulation methods.
Collapse
Affiliation(s)
- Jiayi Shen
- Department of Population and Public Health Sciences, Division of Biostatistics, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Lai Jiang
- Department of Population and Public Health Sciences, Division of Biostatistics, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Kan Wang
- Department of Population and Public Health Sciences, Division of Biostatistics, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Anqi Wang
- Department of Population and Public Health Science, Center for Genetic Epidemiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Fei Chen
- Department of Population and Public Health Science, Center for Genetic Epidemiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | | | - Christopher A. Haiman
- Department of Population and Public Health Science, Center for Genetic Epidemiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - David V. Conti
- Department of Population and Public Health Sciences, Division of Biostatistics, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Department of Population and Public Health Science, Center for Genetic Epidemiology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
43
|
Bigdeli TB, Chatzinakos C, Bendl J, Barr PB, Venkatesh S, Gorman BR, Clarence T, Genovese G, Iyegbe CO, Peterson RE, Kolokotronis SO, Burstein D, Meyers JL, Li Y, Rajeevan N, Sayward F, Cheung KH, Project Among African-Americans to Explore Risks for Schizophrenia (PAARTNERS), Consortium on the Genomics of Schizophrenia (COGS), Genomic Psychiatry Cohort (GPC) Investigators, DeLisi LE, Kosten TR, Zhao H, Achtyes E, Buckley P, Malaspina D, Lehrer D, Rapaport MH, Braff DL, Pato MT, Fanous AH, Pato CN, PsychAD Consortium, Cooperative Studies Program (CSP) #572, Million Veteran Program (MVP), Huang GD, Muralidhar S, Michael Gaziano J, Pyarajan S, Girdhar K, Lee D, Hoffman GE, Aslan M, Fullard JF, Voloudakis G, Harvey PD, Roussos P. Biological Insights from Schizophrenia-associated Loci in Ancestral Populations. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.27.24312631. [PMID: 39252912 PMCID: PMC11383513 DOI: 10.1101/2024.08.27.24312631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Large-scale genome-wide association studies of schizophrenia have uncovered hundreds of associated loci but with extremely limited representation of African diaspora populations. We surveyed electronic health records of 200,000 individuals of African ancestry in the Million Veteran and All of Us Research Programs, and, coupled with genotype-level data from four case-control studies, realized a combined sample size of 13,012 affected and 54,266 unaffected persons. Three genome-wide significant signals - near PLXNA4, PMAIP1, and TRPA1 - are the first to be independently identified in populations of predominantly African ancestry. Joint analyses of African, European, and East Asian ancestries across 86,981 cases and 303,771 controls, yielded 376 distinct autosomal loci, which were refined to 708 putatively causal variants via multi-ancestry fine-mapping. Utilizing single-cell functional genomic data from human brain tissue and two complementary approaches, transcriptome-wide association studies and enhancer-promoter contact mapping, we identified a consensus set of 94 genes across ancestries and pinpointed the specific cell types in which they act. We identified reproducible associations of schizophrenia polygenic risk scores with schizophrenia diagnoses and a range of other mental and physical health problems. Our study addresses a longstanding gap in the generalizability of research findings for schizophrenia across ancestral populations, underlining shared biological underpinnings of schizophrenia across global populations in the presence of broadly divergent risk allele frequencies.
Collapse
Affiliation(s)
- Tim B. Bigdeli
- VA New York Harbor Healthcare System, Brooklyn, NY
- Department of Psychiatry and Behavioral Sciences and SUNY Downstate Health Sciences University, Brooklyn, NY
- Institute for Genomics in Health (IGH), SUNY Downstate Health Sciences University, Brooklyn, NY
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Chris Chatzinakos
- Department of Psychiatry and Behavioral Sciences and SUNY Downstate Health Sciences University, Brooklyn, NY
- Institute for Genomics in Health (IGH), SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Jaroslav Bendl
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
| | - Peter B. Barr
- VA New York Harbor Healthcare System, Brooklyn, NY
- Department of Psychiatry and Behavioral Sciences and SUNY Downstate Health Sciences University, Brooklyn, NY
- Institute for Genomics in Health (IGH), SUNY Downstate Health Sciences University, Brooklyn, NY
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Sanan Venkatesh
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Bryan R. Gorman
- Massachusetts Area Veterans Epidemiology, Research, and Information Center (MAVERIC), Jamaica Plain, MA
| | - Tereza Clarence
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
| | - Giulio Genovese
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
| | - Conrad O. Iyegbe
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
| | - Roseann E. Peterson
- VA New York Harbor Healthcare System, Brooklyn, NY
- Department of Psychiatry and Behavioral Sciences and SUNY Downstate Health Sciences University, Brooklyn, NY
- Institute for Genomics in Health (IGH), SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Sergios-Orestis Kolokotronis
- Institute for Genomics in Health (IGH), SUNY Downstate Health Sciences University, Brooklyn, NY
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY
- Division of Infectious Diseases, Department of Medicine, College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY
- Department of Cell Biology, College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY
| | - David Burstein
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA
- Mental Illness Research, Education and Clinical Center VISN2, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Jacquelyn L. Meyers
- Department of Psychiatry and Behavioral Sciences and SUNY Downstate Health Sciences University, Brooklyn, NY
- Institute for Genomics in Health (IGH), SUNY Downstate Health Sciences University, Brooklyn, NY
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Yuli Li
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Yale University School of Medicine, New Haven, CT
| | - Nallakkandi Rajeevan
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Yale University School of Medicine, New Haven, CT
| | - Frederick Sayward
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Yale University School of Medicine, New Haven, CT
| | - Kei-Hoi Cheung
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Yale University School of Medicine, New Haven, CT
| | | | | | | | - Lynn E. DeLisi
- Department of Psychiatry, Cambridge Health Alliance, Cambridge, MA
| | - Thomas R. Kosten
- Michael E. DeBakey VA Medical Center, Houston, TX
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX
| | - Hongyu Zhao
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Yale University School of Medicine, New Haven, CT
| | - Eric Achtyes
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI
| | - Peter Buckley
- University of Tennessee Health Science Center in Memphis, TN
| | - Dolores Malaspina
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
| | - Douglas Lehrer
- Department of Psychiatry, Wright State University, Dayton, OH
| | - Mark H. Rapaport
- Huntsman Mental Health Institute, Department of Psychiatry, University of Utah, Salt Lake City, UT
| | - David L. Braff
- Department of Psychiatry, University of California, San Diego, CA
- VA San Diego Healthcare System, San Diego, CA
| | - Michele T. Pato
- Department of Psychiatry, Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Ayman H. Fanous
- Department of Psychiatry, University of Arizona College of Medicine-Phoenix, Phoenix, AZ
- Department of Psychiatry, VA Phoenix Healthcare System, Phoenix, AZ
| | - Carlos N. Pato
- Department of Psychiatry, Robert Wood Johnson Medical School, New Brunswick, NJ
| | | | | | | | - Grant D. Huang
- Office of Research and Development, Veterans Health Administration, Washington, DC
| | - Sumitra Muralidhar
- Office of Research and Development, Veterans Health Administration, Washington, DC
| | - J. Michael Gaziano
- Massachusetts Area Veterans Epidemiology, Research, and Information Center (MAVERIC), Jamaica Plain, MA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Saiju Pyarajan
- Massachusetts Area Veterans Epidemiology, Research, and Information Center (MAVERIC), Jamaica Plain, MA
| | - Kiran Girdhar
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
| | - Donghoon Lee
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
| | - Gabriel E. Hoffman
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA
- Mental Illness Research, Education and Clinical Center VISN2, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Mihaela Aslan
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Yale University School of Medicine, New Haven, CT
| | - John F. Fullard
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
| | - Georgios Voloudakis
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA
- Mental Illness Research, Education and Clinical Center VISN2, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Philip D. Harvey
- Bruce W. Carter Miami Veterans Affairs (VA) Medical Center, Miami, FL
- University of Miami School of Medicine, Miami, FL
| | - Panos Roussos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA
- Mental Illness Research, Education and Clinical Center VISN2, James J. Peters VA Medical Center, Bronx, NY, USA
| |
Collapse
|
44
|
Kamp M, Pain O, Lewis CM, Ramsay M. Ancestry-aligned polygenic scores combined with conventional risk factors improve prediction of cardiometabolic outcomes in African populations. Genome Med 2024; 16:106. [PMID: 39187845 PMCID: PMC11346299 DOI: 10.1186/s13073-024-01377-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 08/12/2024] [Indexed: 08/28/2024] Open
Abstract
BACKGROUND Cardiovascular diseases (CVD) are a major health concern in Africa. Improved identification and treatment of high-risk individuals can reduce adverse health outcomes. Current CVD risk calculators are largely unvalidated in African populations and overlook genetic factors. Polygenic scores (PGS) can enhance risk prediction by measuring genetic susceptibility to CVD, but their effectiveness in genetically diverse populations is limited by a European-ancestry bias. To address this, we developed models integrating genetic data and conventional risk factors to assess the risk of developing cardiometabolic outcomes in African populations. METHODS We used summary statistics from a genome-wide association meta-analysis (n = 14,126) in African populations to derive novel genome-wide PGS for 14 cardiometabolic traits in an independent African target sample (Africa Wits-INDEPTH Partnership for Genomic Research (AWI-Gen), n = 10,603). Regression analyses assessed relationships between each PGS and corresponding cardiometabolic trait, and seven CVD outcomes (CVD, heart attack, stroke, diabetes mellitus, dyslipidaemia, hypertension, and obesity). The predictive utility of the genetic data was evaluated using elastic net models containing multiple PGS (MultiPGS) and reference-projected principal components of ancestry (PPCs). An integrated risk prediction model incorporating genetic and conventional risk factors was developed. Nested cross-validation was used when deriving elastic net models to enhance generalisability. RESULTS Our African-specific PGS displayed significant but variable within- and cross- trait prediction (max.R2 = 6.8%, p = 1.86 × 10-173). Significantly associated PGS with dyslipidaemia included the PGS for total cholesterol (logOR = 0.210, SE = 0.022, p = 2.18 × 10-21) and low-density lipoprotein (logOR = - 0.141, SE = 0.022, p = 1.30 × 10-20); with hypertension, the systolic blood pressure PGS (logOR = 0.150, SE = 0.045, p = 8.34 × 10-4); and multiple PGS associated with obesity: body mass index (max. logOR = 0.131, SE = 0.031, p = 2.22 × 10-5), hip circumference (logOR = 0.122, SE = 0.029, p = 2.28 × 10-5), waist circumference (logOR = 0.013, SE = 0.098, p = 8.13 × 10-4) and weight (logOR = 0.103, SE = 0.029, p = 4.89 × 10-5). Elastic net models incorporating MultiPGS and PPCs significantly improved prediction over MultiPGS alone. Models including genetic data and conventional risk factors were more predictive than conventional risk models alone (dyslipidaemia: R2 increase = 2.6%, p = 4.45 × 10-12; hypertension: R2 increase = 2.6%, p = 2.37 × 10-13; obesity: R2 increase = 5.5%, 1.33 × 10-34). CONCLUSIONS In African populations, CVD and associated cardiometabolic trait prediction models can be improved by incorporating ancestry-aligned PGS and accounting for ancestry. Combining PGS with conventional risk factors further enhances prediction over traditional models based on conventional factors. Incorporating data from target populations can improve the generalisability of international predictive models for CVD and associated traits in African populations.
Collapse
Affiliation(s)
- Michelle Kamp
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, The University of the Witwatersrand, Johannesburg, South Africa.
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, Psychology & Neuroscience, London, UK.
| | - Oliver Pain
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
| | - Cathryn M Lewis
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, Psychology & Neuroscience, London, UK
- Department of Medical & Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Michèle Ramsay
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, The University of the Witwatersrand, Johannesburg, South Africa.
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
45
|
Vialle RA, de Paiva Lopes K, Li Y, Ng B, Schneider JA, Buchman AS, Wang Y, Farfel JM, Barnes LL, Wingo AP, Wingo TS, Seyfried NT, De Jager PL, Gaiteri C, Tasaki S, Bennett DA. Structural variants linked to Alzheimer's Disease and other common age-related clinical and neuropathologic traits. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.12.24311887. [PMID: 39185527 PMCID: PMC11343262 DOI: 10.1101/2024.08.12.24311887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Advances have led to a greater understanding of the genetics of Alzheimer's Disease (AD). However, the gap between the predicted and observed genetic heritability estimates when using single nucleotide polymorphisms (SNPs) and small indel data remains. Large genomic rearrangements, known as structural variants (SVs), have the potential to account for this missing genetic heritability. By leveraging data from two ongoing cohort studies of aging and dementia, the Religious Orders Study and Rush Memory and Aging Project (ROS/MAP), we performed genome-wide association analysis testing around 20,000 common SVs from 1,088 participants with whole genome sequencing (WGS) data. A range of Alzheimer's Disease and Related Disorders (AD/ADRD) clinical and pathologic traits were examined. Given the limited sample size, no genome-wide significant association was found, but we mapped SVs across 81 AD risk loci and discovered 22 SVs in linkage disequilibrium (LD) with GWAS lead variants and directly associated with AD/ADRD phenotypes (nominal P < 0.05). The strongest association was a deletion of an Alu element in the 3'UTR of the TMEM106B gene. This SV was in high LD with the respective AD GWAS locus and was associated with multiple AD/ADRD phenotypes, including tangle density, TDP-43, and cognitive resilience. The deletion of this element was also linked to lower TMEM106B protein abundance. We also found a 22 kb deletion associated with depression in ROSMAP and bearing similar association patterns as AD GWAS SNPs at the IQCK locus. In addition, genome-wide scans allowed the identification of 7 SVs, with no LD with SNPs and nominally associated with AD/ADRD traits. This result suggests potentially new ADRD risk loci not discoverable using SNP data. Among these findings, we highlight a 5.6 kb duplication of coding regions of the gene C1orf186 at chromosome 1 associated with indices of cognitive impairment, decline, and resilience. While further replication in independent datasets is needed to validate these findings, our results support the potential roles of common structural variations in the pathogenesis of AD/ADRD.
Collapse
Affiliation(s)
- Ricardo A Vialle
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Katia de Paiva Lopes
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Yan Li
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Bernard Ng
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Aron S Buchman
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Yanling Wang
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Jose M Farfel
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Lisa L Barnes
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Aliza P Wingo
- Department of Psychiatry, University of California, Davis CA, USA
- VA Northern California Health Care System, McClellan Park, CA, USA
| | - Thomas S Wingo
- Department of Neurology, University of California, Davis, CA, USA
| | - Nicholas T Seyfried
- Goizueta Alzheimer's Disease Research Center, Department of Neurology and Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Philip L De Jager
- Department of Neurology, College of Physicians and Surgeons, Columbia University and the New York Presbyterian Hospital, New York, NY, USA
| | - Chris Gaiteri
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Shinya Tasaki
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
46
|
Imtiaz MA, Melas K, Tin A, Talevi V, Chen H, Fornage M, Shrestha S, Gögele M, Emmert D, Pattaro C, Pramstaller P, Förster F, Horn K, Mosley TH, Fuchsberger C, Scholz M, Breteler MM, Aziz NA. Genome-Wide Association Study Meta-Analysis Uncovers Novel Genetic Variants Associated with Olfactory Dysfunction. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.09.24311665. [PMID: 39148842 PMCID: PMC11326328 DOI: 10.1101/2024.08.09.24311665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
IMPORTANCE Olfactory dysfunction is among the earliest signs of many age-related neurodegenerative diseases and has been associated with increased mortality in older adults; however, its genetic basis remains largely unknown. OBJECTIVE To identify the genetic loci associated with olfactory dysfunction in the general population. DESIGN SETTING AND PARTIICIPANTS This genome-wide association study meta-analysis (GWMA) included participants of European ancestry (N = 22,730) enrolled in four different large population-based studies, followed by a multi-ancestry GWMA including participants of African ancestry (N = 1,030). The data analysis was performed from March 2023 through June 2024. EXPOSURES Genome-wide single nucleotide polymorphisms. MAIN OUTCOMES AND MEASURES Olfactory dysfunction was the outcome and assessed using a 12-item smell identification test. RESULTS GWMA revealed a novel genome-wide significant locus (tagged by rs11228623 at 11q12) associated with olfactory dysfunction. Gene-based analysis revealed a high enrichment for olfactory receptor genes in this region. Phenome-wide association studies demonstrated associations between genetic variants related to olfactory dysfunction and blood cell counts, kidney function, skeletal muscle mass, cholesterol levels and cardiovascular disease. Using individual-level data, we also confirmed and quantified the strength of these associations on a phenotypic level. Moreover, employing two-sample Mendelian Randomization analyses, we found evidence for causal associations between olfactory dysfunction and these phenotypes. CONCLUSIONS These findings provide novel insights into the genetic architecture of the sense of smell and highlight its importance for many aspects of human health.
Collapse
Affiliation(s)
- Mohammed Aslam Imtiaz
- Population Health Sciences, German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Konstantinos Melas
- Population Health Sciences, German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Adrienne Tin
- Department of Medicine, University of Mississippi Medical Center, Jackson, 39216, MS, USA
| | - Valentina Talevi
- Population Health Sciences, German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Honglei Chen
- Department of Epidemiology and Biostatistics, Michigan State University, Michigan, USA
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science at Houston, Houston, TX 77030, USA
- Human Genetics Center, School of Public Health, University of Texas Health Science at Houston, Houston, TX 77030, USA
| | - Srishti Shrestha
- The Memory Impairment Neurodegenerative Dementia (MIND) Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Martin Gögele
- Eurac Research, Institute for Biomedicine, Via Volta 21, 39100, Bolzano, Italy
| | - David Emmert
- Eurac Research, Institute for Biomedicine, Via Volta 21, 39100, Bolzano, Italy
| | - Cristian Pattaro
- Eurac Research, Institute for Biomedicine, Via Volta 21, 39100, Bolzano, Italy
| | - Peter Pramstaller
- Eurac Research, Institute for Biomedicine, Via Volta 21, 39100, Bolzano, Italy
| | - Franz Förster
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), Medical Faculty, Leipzig University, Leipzig, Germany
| | - Katrin Horn
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), Medical Faculty, Leipzig University, Leipzig, Germany
| | - Thomas H. Mosley
- The Memory Impairment Neurodegenerative Dementia (MIND) Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | | | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), Medical Faculty, Leipzig University, Leipzig, Germany
- LIFE Research Center for Civilization Diseases, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Monique M.B. Breteler
- Population Health Sciences, German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Institute for Medical Biometry, Informatics and Epidemiology (IMBIE), Faculty of Medicine, University of Bonn, Germany
| | - N. Ahmad Aziz
- Population Health Sciences, German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, Faculty of Medicine, University of Bonn, Bonn, Germany
| |
Collapse
|
47
|
Rajabli F, Emekci A. Addressing overlapping sample challenges in genome-wide association studies: Meta-reductive approach. PLoS One 2024; 19:e0296207. [PMID: 39088468 PMCID: PMC11293628 DOI: 10.1371/journal.pone.0296207] [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: 12/06/2023] [Accepted: 06/10/2024] [Indexed: 08/03/2024] Open
Abstract
Polygenic risk scores (PRS) are instrumental in genetics, offering insights into an individual level genetic risk to a range of diseases based on accumulated genetic variations. These scores rely on Genome-Wide Association Studies (GWAS). However, precision in PRS is often challenged by the requirement of extensive sample sizes and the potential for overlapping datasets that can inflate PRS calculations. In this study, we present a novel methodology, Meta-Reductive Approach (MRA), that was derived algebraically to adjust GWAS results, aiming to neutralize the influence of select cohorts. Our approach recalibrates summary statistics using algebraic derivations. Validating our technique with datasets from Alzheimer disease studies, we showed that the summary statistics of the MRA and those derived from individual-level data yielded the exact same values. This innovative method offers a promising avenue for enhancing the accuracy of PRS, especially when derived from meta-analyzed GWAS data.
Collapse
Affiliation(s)
- Farid Rajabli
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Dr. John T Macdonald Foundation Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Azra Emekci
- Pioneer High School, San Jose, CA, United States of America
| |
Collapse
|
48
|
Yap CF, Morris AP. Methods for multiancestry genome-wide association study meta-analysis. Ann Hum Genet 2024. [PMID: 39022911 DOI: 10.1111/ahg.12572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/20/2024]
Abstract
Genome-wide association studies (GWAS) have significantly enhanced our understanding of the genetic basis of complex diseases. Despite the technological advancements, gaps in our understanding remain, partly due to small effect sizes and inadequate coverage of genetic variation. Multiancestry GWAS meta-analysis (MAGMA) addresses these challenges by integrating genetic data from diverse populations, thereby increasing power to detect loci and improving fine-mapping resolution to identify causal variants across different ancestry groups. This review provides an overview of the protocols, statistical methods, and software of MAGMA, as well as highlighting some challenges associated with this approach.
Collapse
Affiliation(s)
- Chuan Fu Yap
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, The University of Manchester, Manchester, UK
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, The University of Manchester, Manchester, UK
| |
Collapse
|
49
|
Tsetsos F, Topaloudi A, Jain P, Yang Z, Yu D, Kolovos P, Tumer Z, Rizzo R, Hartmann A, Depienne C, Worbe Y, Müller-Vahl KR, Cath DC, Boomsma DI, Wolanczyk T, Zekanowski C, Barta C, Nemoda Z, Tarnok Z, Padmanabhuni SS, Buxbaum JD, Grice D, Glennon J, Stefansson H, Hengerer B, Yannaki E, Stamatoyannopoulos JA, Benaroya-Milshtein N, Cardona F, Hedderly T, Heyman I, Huyser C, Mir P, Morer A, Mueller N, Munchau A, Plessen KJ, Porcelli C, Roessner V, Walitza S, Schrag A, Martino D, PGC TS Working Group, The TSAICG, The TSGeneSEE initiative, The EMTICS collaborative group, The TS-EUROTRAIN network, The TIC Genetics collaborative group, Tischfield JA, Heiman GA, Willsey AJ, Dietrich A, Davis LK, Crowley JJ, Mathews CA, Scharf JM, Georgitsi M, Hoekstra PJ, Paschou P. Genome-Wide Association Study Points to Novel Locus for Gilles de la Tourette Syndrome. Biol Psychiatry 2024; 96:114-124. [PMID: 36738982 PMCID: PMC10783199 DOI: 10.1016/j.biopsych.2023.01.023] [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: 05/11/2022] [Revised: 11/23/2022] [Accepted: 01/24/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND Tourette syndrome (TS) is a childhood-onset neurodevelopmental disorder of complex genetic architecture and is characterized by multiple motor tics and at least one vocal tic persisting for more than 1 year. METHODS We performed a genome-wide meta-analysis integrating a novel TS cohort with previously published data, resulting in a sample size of 6133 individuals with TS and 13,565 ancestry-matched control participants. RESULTS We identified a genome-wide significant locus on chromosome 5q15. Integration of expression quantitative trait locus, Hi-C (high-throughput chromosome conformation capture), and genome-wide association study data implicated the NR2F1 gene and associated long noncoding RNAs within the 5q15 locus. Heritability partitioning identified statistically significant enrichment in brain tissue histone marks, while polygenic risk scoring of brain volume data identified statistically significant associations with right and left thalamus volumes and right putamen volume. CONCLUSIONS Our work presents novel insights into the neurobiology of TS, thereby opening up new directions for future studies.
Collapse
Affiliation(s)
- Fotis Tsetsos
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - Apostolia Topaloudi
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Pritesh Jain
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Zhiyu Yang
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| | - Dongmei Yu
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Petros Kolovos
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - Zeynep Tumer
- Department of Clinical Genetics, Kennedy Center, Copenhagen University Hospital, Rigshospitalet, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen
| | - Renata Rizzo
- Child and Adolescent Neurology and Psychiatry, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Andreas Hartmann
- Department of Neurology, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Christel Depienne
- Institute for Human Genetics, University Hospital Essen, Essen, Germany
| | - Yulia Worbe
- Assistance Publique Hôpitaux de Paris, Hopital Saint Antoine, Paris France
- French Reference Centre for Gilles de la Tourette Syndrome, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Kirsten R. Müller-Vahl
- Department of Psychiatry, Social psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Danielle C. Cath
- Department of Clinical and health Psychology, Utrecht University, Utrecht, Netherlands
| | - Dorret I. Boomsma
- Institute for Anatomy and Cell Biology, Ulm University, Ulm, Germany
- EMGO+ Institute for Health and Care Research, VU University Medical Centre, Amsterdam, Netherlands
| | - Tomasz Wolanczyk
- Department of Child Psychiatry, Medical University of Warsaw, Warsaw, Poland
| | - Cezary Zekanowski
- Laboratory of Neurogenetics, Department of Neurodegenerative Disorders, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Csaba Barta
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Zsofia Nemoda
- Department of Molecular Biology, Institute of Biochemistry and Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Zsanett Tarnok
- Vadaskert Clinic for Child and Adolescent Psychiatry, Hungary
| | | | - Joseph D. Buxbaum
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, USA
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, USA
| | - Dorothy Grice
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, USA
- Division of Tics, OCD, and Related Disorders, Icahn School of Medicine at Mount Sinai, USA
| | - Jeffrey Glennon
- Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Netherlands
| | | | - Bastian Hengerer
- Boehringer Ingelheim Pharma GmbH & Co. KG, CNS Research, Germany
| | - Evangelia Yannaki
- Hematology Department- Hematopoietic Cell Transplantation Unit, Gene and Cell Therapy Center, George Papanikolaou Hospital, Greece
- Department of Medicine, University of Washington, WA, USA
| | - John A. Stamatoyannopoulos
- Altius Institute for Biomedical Sciences, WA, USA
- Department of Genome Sciences, University of Washington, WA, USA
- Department of Medicine, Division of Oncology, University of Washington, WA, USA
| | - Noa Benaroya-Milshtein
- Child and Adolescent Psychiatry Department, Schneider Children’s Medical Centre of Israel, Petah-Tikva. Affiliated to Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - Francesco Cardona
- Department of Human Neurosciences, University La Sapienza of Rome, Rome, Italy
| | - Tammy Hedderly
- Evelina London Children’s Hospital GSTT, Kings Health Partners AHSC, London, UK
| | - Isobel Heyman
- Psychological Medicine, Great Ormond Street Hospital NHS Foundation Trust, Great Ormond Street, London, UK
| | - Chaim Huyser
- Levvel, Academic Center for Child and Adolescent Psychiatry, Amsterdam, The Netherlands
- Amsterdam UMC, Department of Child and Adolescent Psychiatry, Amsterdam, The Netherlands
| | - Pablo Mir
- Unidad de Trastornos del Movimiento. Instituto de Biomedicina de Sevilla (IBiS). Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla. Seville, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Astrid Morer
- Department of Child and Adolescent Psychiatry and Psychology, Institute of Neurosciences, Hospital Clinic Universitari, Barcelona, Spain
- Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigacion en Red de Salud Mental (CIBERSAM), Instituto Carlos III, Spain
| | - Norbert Mueller
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Alexander Munchau
- Institute of Systems Motor Science, University of Lübeck, Lübeck, Germany
| | - Kerstin J Plessen
- Child and Adolescent Mental Health Centre, Mental Health Services, Capital Region of Denmark and University of Copenhagen, Copenhagen, Denmark
- Division of Child and Adolescent Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Cesare Porcelli
- ASL BA, Maternal and Childood Department; Adolescence and Childhood Neuropsychiatry Unit; Bari, Italy
| | - Veit Roessner
- Department of Child and Adolescent Psychiatry, Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Susanne Walitza
- Department of Child and Adolescent Psychiatry and Psychotherapy, University of Zurich, Zurich, Switzerland
| | - Anette Schrag
- Department of Clinical Neuroscience, UCL Institute of Neurology, University College London, London, UK
| | - Davide Martino
- Department of Clinical Neurosciences, Cumming School of Medicine & Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | | | | | | | | | | | | | - Jay A. Tischfield
- Department of Genetics and the Human Genetics Institute of New Jersey, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Gary A. Heiman
- Department of Genetics and the Human Genetics Institute of New Jersey, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - A. Jeremy Willsey
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Andrea Dietrich
- University of Groningen, University Medical Centre Groningen, Department of Child and Adolescent Psychiatry, Groningen, the Netherlands
| | - Lea K. Davis
- Division of Genetic Medicine, Department of Medicine Vanderbilt University Medical Center Nashville, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James J. Crowley
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Carol A. Mathews
- Department of Psychiatry and Genetics Institute, University of Florida College of Medicine, USA
| | - Jeremiah M. Scharf
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Neurology, Brigham and Women’s Hospital, and the Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Marianthi Georgitsi
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
- 1st Laboratory of Medical Biology-Genetics, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Pieter J. Hoekstra
- University of Groningen, University Medical Centre Groningen, Department of Child and Adolescent Psychiatry, Groningen, the Netherlands
| | - Peristera Paschou
- Department of Biological Sciences, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
50
|
Xiu Z, Sun L, Liu K, Cao H, Qu HQ, Glessner JT, Ding Z, Zheng G, Wang N, Xia Q, Li J, Li MJ, Hakonarson H, Liu W, Li J. Shared molecular mechanisms and transdiagnostic potential of neurodevelopmental disorders and immune disorders. Brain Behav Immun 2024; 119:767-780. [PMID: 38677625 DOI: 10.1016/j.bbi.2024.04.026] [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: 10/21/2023] [Revised: 02/27/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024] Open
Abstract
The co-occurrence and familial clustering of neurodevelopmental disorders and immune disorders suggest shared genetic risk factors. Based on genome-wide association summary statistics from five neurodevelopmental disorders and four immune disorders, we conducted genome-wide, local genetic correlation and polygenic overlap analysis. We further performed a cross-trait GWAS meta-analysis. Pleotropic loci shared between the two categories of diseases were mapped to candidate genes using multiple algorithms and approaches. Significant genetic correlations were observed between neurodevelopmental disorders and immune disorders, including both positive and negative correlations. Neurodevelopmental disorders exhibited higher polygenicity compared to immune disorders. Around 50%-90% of genetic variants of the immune disorders were shared with neurodevelopmental disorders. The cross-trait meta-analysis revealed 154 genome-wide significant loci, including 8 novel pleiotropic loci. Significant associations were observed for 30 loci with both types of diseases. Pathway analysis on the candidate genes at these loci revealed common pathways shared by the two types of diseases, including neural signaling, inflammatory response, and PI3K-Akt signaling pathway. In addition, 26 of the 30 lead SNPs were associated with blood cell traits. Neurodevelopmental disorders exhibit complex polygenic architecture, with a subset of individuals being at a heightened genetic risk for both neurodevelopmental and immune disorders. The identification of pleiotropic loci has important implications for exploring opportunities for drug repurposing, enabling more accurate patient stratification, and advancing genomics-informed precision in the medical field of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Zhanjie Xiu
- Department of Cell Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Medical Epigenetics, Tianjin Institute of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China; Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ling Sun
- Department of Child and Adolescent Psychology, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Kunlun Liu
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Haiyan Cao
- Department of Child and Adolescent Psychology, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Hui-Qi Qu
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Joseph T Glessner
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States; Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, United States; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Zhiyong Ding
- Mills Institute for Personalized Cancer Care, Fynn Biotechnologies Ltd., Jinan, China
| | - Gang Zheng
- National Supercomputer Center in Tianjin (NSCC-TJ), Tianjin, China
| | - Nan Wang
- Mills Institute for Personalized Cancer Care, Fynn Biotechnologies Ltd., Jinan, China
| | - Qianghua Xia
- Department of Cell Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Medical Epigenetics, Tianjin Institute of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China; Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jie Li
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Tianjin Medical University, Tianjin, China
| | - Mulin Jun Li
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States; Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, United States; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| | - Wei Liu
- Tianjin Children's Hospital (Tianjin University Children's Hospital), Tianjin, China; Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin, China.
| | - Jin Li
- Department of Cell Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Medical Epigenetics, Tianjin Institute of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China; Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|