151
|
Jiang Y, Zhou X, Wong HY, Ouyang L, Ip FCF, Chau VMN, Lau SF, Wu W, Wong DYK, Seo H, Fu WY, Lai NCH, Chen Y, Chen Y, Tong EPS, Mok VCT, Kwok TCY, Mok KY, Shoai M, Lehallier B, Losada PM, O'Brien E, Porter T, Laws SM, Hardy J, Wyss-Coray T, Masters CL, Fu AKY, Ip NY. An IL1RL1 genetic variant lowers soluble ST2 levels and the risk effects of APOE-ε4 in female patients with Alzheimer's disease. NATURE AGING 2022; 2:616-634. [PMID: 37117777 PMCID: PMC10154240 DOI: 10.1038/s43587-022-00241-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 05/23/2022] [Indexed: 04/30/2023]
Abstract
Changes in the levels of circulating proteins are associated with Alzheimer's disease (AD), whereas their pathogenic roles in AD are unclear. Here, we identified soluble ST2 (sST2), a decoy receptor of interleukin-33-ST2 signaling, as a new disease-causing factor in AD. Increased circulating sST2 level is associated with more severe pathological changes in female individuals with AD. Genome-wide association analysis and CRISPR-Cas9 genome editing identified rs1921622 , a genetic variant in an enhancer element of IL1RL1, which downregulates gene and protein levels of sST2. Mendelian randomization analysis using genetic variants, including rs1921622 , demonstrated that decreased sST2 levels lower AD risk and related endophenotypes in females carrying the Apolipoprotein E (APOE)-ε4 genotype; the association is stronger in Chinese than in European-descent populations. Human and mouse transcriptome and immunohistochemical studies showed that rs1921622 /sST2 regulates amyloid-beta (Aβ) pathology through the modulation of microglial activation and Aβ clearance. These findings demonstrate how sST2 level is modulated by a genetic variation and plays a disease-causing role in females with AD.
Collapse
Affiliation(s)
- Yuanbing Jiang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Xiaopu Zhou
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development; Shenzhen-Hong Kong Institute of Brain Science, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Hiu Yi Wong
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Li Ouyang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Fanny C F Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development; Shenzhen-Hong Kong Institute of Brain Science, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Vicky M N Chau
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Shun-Fat Lau
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Wei Wu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Daniel Y K Wong
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Heukjin Seo
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Wing-Yu Fu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Nicole C H Lai
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Yuewen Chen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development; Shenzhen-Hong Kong Institute of Brain Science, HKUST Shenzhen Research Institute, Shenzhen, China
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Yu Chen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development; Shenzhen-Hong Kong Institute of Brain Science, HKUST Shenzhen Research Institute, Shenzhen, China
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Estella P S Tong
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Vincent C T Mok
- Gerald Choa Neuroscience Centre, Lui Che Woo Institute of Innovative Medicine, Therese Pei Fong Chow Research Centre for Prevention of Dementia, Division of Neurology, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Timothy C Y Kwok
- Therese Pei Fong Chow Research Centre for Prevention of Dementia, Division of Geriatrics, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Kin Y Mok
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute, University College London, London, UK
| | - Maryam Shoai
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute, University College London, London, UK
| | - Benoit Lehallier
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Alkahest Inc, San Carlos, California, USA
| | - Patricia Morán Losada
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
| | - Eleanor O'Brien
- Centre for Precision Health, Edith Cowan University, Joondalup, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - Tenielle Porter
- Centre for Precision Health, Edith Cowan University, Joondalup, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Australia
| | - Simon M Laws
- Centre for Precision Health, Edith Cowan University, Joondalup, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Australia
| | - John Hardy
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
- UK Dementia Research Institute, University College London, London, UK
- Institute for Advanced Study, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Tony Wyss-Coray
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, USA
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, California, USA
| | - Colin L Masters
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Amy K Y Fu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development; Shenzhen-Hong Kong Institute of Brain Science, HKUST Shenzhen Research Institute, Shenzhen, China
| | - Nancy Y Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China.
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development; Shenzhen-Hong Kong Institute of Brain Science, HKUST Shenzhen Research Institute, Shenzhen, China.
| |
Collapse
|
152
|
Mrug S, Barker-Kamps M, Orihuela CA, Patki A, Tiwari HK. Childhood Neighborhood Disadvantage, Parenting, and Adult Health. Am J Prev Med 2022; 63:S28-S36. [PMID: 35725138 PMCID: PMC9219037 DOI: 10.1016/j.amepre.2022.01.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/04/2022] [Accepted: 01/20/2022] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Growing up in disadvantaged neighborhoods is associated with poor adult health indicators. Consistent and supportive parenting plays a key role in life-long health, but it is not known whether positive parenting can mitigate the relationship between neighborhood adversity and poor health. This study examines parenting as a moderator of the links between childhood neighborhood characteristics and adult health indicators. METHODS A sample of 305 individuals (61% female; 82% African American, 18% Caucasian) were assessed in childhood (T1; age 11 years; 2003‒2004) and adulthood (T2; age 27 years; 2018‒2021). At T1, neighborhood poverty was derived from census data; neighborhood disorder was reported by parents. Children reported on parental harsh discipline, inconsistent discipline, and parental nurturance. At T2, health outcomes included BMI, serum cortisol and C-reactive protein (CRP), and salivary DNA methylation index related to CRP. Regression models predicted T2 health outcomes from T1 neighborhood and parenting variables and their interactions, adjusting for clustering and confounders. Data were analyzed in 2021. RESULTS Neighborhood poverty was associated with lower cortisol, whereas neighborhood disorder was linked with CRP‒related DNA methylation. Multiple interactions between neighborhood and parenting variables emerged, indicating that adverse neighborhood conditions were only related to poor adult health when combined with inconsistent discipline and low parental nurturance. By contrast, warm and supportive parenting, consistent discipline, and to a lesser extent harsh discipline buffered children from poor health outcomes associated with neighborhood disadvantage. CONCLUSIONS Interventions enhancing consistent and nurturing parenting may help to reduce the long-term associations of neighborhood disadvantage with poor health.
Collapse
Affiliation(s)
- Sylvie Mrug
- Department of Psychology, College of Arts and Sciences, The University of Alabama at Birmingham, Birmingham, Alabama.
| | - Malcolm Barker-Kamps
- Department of Psychology, College of Arts and Sciences, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Catheryn A Orihuela
- Department of Psychology, College of Arts and Sciences, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Amit Patki
- Department of Biostatistics, School of Public Health, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Hemant K Tiwari
- Department of Biostatistics, School of Public Health, The University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
153
|
Neumann A, Jolicoeur‐Martineau A, Szekely E, Sallis HM, O’Donnel K, Greenwood CM, Levitan R, Meaney MJ, Wazana A, Evans J, Tiemeier H. Combined polygenic risk scores of different psychiatric traits predict general and specific psychopathology in childhood. J Child Psychol Psychiatry 2022; 63:636-645. [PMID: 34389974 PMCID: PMC9291767 DOI: 10.1111/jcpp.13501] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/14/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Polygenic risk scores (PRSs) operationalize genetic propensity toward a particular mental disorder and hold promise as early predictors of psychopathology, but before a PRS can be used clinically, explanatory power must be increased and the specificity for a psychiatric domain established. To enable early detection, it is crucial to study these psychometric properties in childhood. We examined whether PRSs associate more with general or with specific psychopathology in school-aged children. Additionally, we tested whether psychiatric PRSs can be combined into a multi-PRS score for improved performance. METHODS We computed 16 PRSs based on GWASs of psychiatric phenotypes, but also neuroticism and cognitive ability, in mostly adult populations. Study participants were 9,247 school-aged children from three population-based cohorts of the DREAM-BIG consortium: ALSPAC (UK), The Generation R Study (Netherlands), and MAVAN (Canada). We associated each PRS with general and specific psychopathology factors, derived from a bifactor model based on self-report and parental, teacher, and observer reports. After fitting each PRS in separate models, we also tested a multi-PRS model, in which all PRSs are entered simultaneously as predictors of the general psychopathology factor. RESULTS Seven PRSs were associated with the general psychopathology factor after multiple testing adjustment, two with specific externalizing and five with specific internalizing psychopathology. PRSs predicted general psychopathology independently of each other, with the exception of depression and depressive symptom PRSs. Most PRSs associated with a specific psychopathology domain, were also associated with general child psychopathology. CONCLUSIONS The results suggest that PRSs based on current GWASs of psychiatric phenotypes tend to be associated with general psychopathology, or both general and specific psychiatric domains, but not with one specific psychopathology domain only. Furthermore, PRSs can be combined to improve predictive ability. PRS users should therefore be conscious of nonspecificity and consider using multiple PRSs simultaneously, when predicting psychiatric disorders.
Collapse
Affiliation(s)
- Alexander Neumann
- Department of Child and Adolescent PsychiatryErasmus University Medical CenterRotterdamThe Netherlands
- Lady Davis Institute for Medical ResearchJewish General HospitalMontrealQCCanada
- VIB Center for Molecular NeurologyVIBAntwerpBelgium
- Department of Biomedical SciencesUniversity of AntwerpAntwerpBelgium
| | | | - Eszter Szekely
- Lady Davis Institute for Medical ResearchJewish General HospitalMontrealQCCanada
- Department of PsychiatryMcGill University Faculty of MedicineMontrealQCCanada
| | - Hannah M. Sallis
- MRC Integrative Epidemiology UnitUniversity of BristolBristolUK
- Centre for Academic Mental Health, Population Health SciencesBristol Medical SchoolUniversity of BristolBristolUK
- School of Psychological ScienceUniversity of BristolBristolUK
| | - Kieran O’Donnel
- Department of Psychiatry and Sackler Program for Epigenetics and PsychobiologyMcGill UniversityMontrealQCCanada
- Ludmer Centre for Neuroinformatics and Mental HealthMcGill UniversityMontrealQCCanada
| | - Celia M.T. Greenwood
- Lady Davis Institute for Medical ResearchJewish General HospitalMontrealQCCanada
- Departments of Oncology, Human Genetics, and Epidemiology, Biostatistics and Occupational HealthMcGill UniversityMontrealQCCanada
| | - Robert Levitan
- Centre for Addiction and Mental HealthTorontoONCanada
- Department of PsychiatryUniversity of TorontoTorontoONCanada
| | - Michael J. Meaney
- Department of PsychiatryMcGill University Faculty of MedicineMontrealQCCanada
- Douglas Mental Health InstituteMontrealQCCanada
- Singapore Institute for Clinical SciencesSingapore CitySingapore
| | - Ashley Wazana
- Lady Davis Institute for Medical ResearchJewish General HospitalMontrealQCCanada
- Department of PsychiatryMcGill University Faculty of MedicineMontrealQCCanada
- Centre for Child Development and Mental HealthJewish General HospitalMontrealQCCanada
| | - Jonathan Evans
- Centre for Academic Mental Health, Population Health SciencesBristol Medical SchoolUniversity of BristolBristolUK
| | - Henning Tiemeier
- Department of Child and Adolescent PsychiatryErasmus University Medical CenterRotterdamThe Netherlands
- Department of Social and Behavioral SciencesHarvard T. H. Chan School of Public HealthBostonMAUSA
| |
Collapse
|
154
|
Herrera‐Luis E, Ortega VE, Ampleford EJ, Sio YY, Granell R, de Roos E, Terzikhan N, Vergara E, Hernandez‐Pacheco N, Perez‐Garcia J, Martin‐Gonzalez E, Lorenzo‐Diaz F, Hashimoto S, Brinkman P, U‐BIOPRED Study Group, Jorgensen AL, Yan Q, Forno E, Vijverberg SJ, Lethem R, Espuela‐Ortiz A, Gorenjak M, Eng C, González‐Pérez R, Hernández‐Pérez JM, Poza‐Guedes P, Sardón O, Corcuera P, Hawkins G, Marsico A, Bahmer T, Rabe KF, Hansen G, Kopp MV, Rios R, Cruz M, González‐Barcala F, Olaguibel JM, Plaza V, Quirce S, Canino G, Cloutier M, del Pozo V, Rodriguez‐Santana JR, Korta‐Murua J, Villar J, Potočnik U, Figueiredo C, Kabesch M, Mukhopadhyay S, Pirmohamed M, Hawcutt D, Melén E, Palmer CN, Turner S, Maitland‐van der Zee AH, von Mutius E, Celedón JC, Brusselle G, Chew FT, Bleecker E, Meyers D, Burchard EG, Pino‐Yanes M. Multi-ancestry genome-wide association study of asthma exacerbations. Pediatr Allergy Immunol 2022; 33:e13802. [PMID: 35754128 PMCID: PMC9671132 DOI: 10.1111/pai.13802] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/01/2022] [Accepted: 05/04/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND Asthma exacerbations are a serious public health concern due to high healthcare resource utilization, work/school productivity loss, impact on quality of life, and risk of mortality. The genetic basis of asthma exacerbations has been studied in several populations, but no prior study has performed a multi-ancestry meta-analysis of genome-wide association studies (meta-GWAS) for this trait. We aimed to identify common genetic loci associated with asthma exacerbations across diverse populations and to assess their functional role in regulating DNA methylation and gene expression. METHODS A meta-GWAS of asthma exacerbations in 4989 Europeans, 2181 Hispanics/Latinos, 1250 Singaporean Chinese, and 972 African Americans analyzed 9.6 million genetic variants. Suggestively associated variants (p ≤ 5 × 10-5 ) were assessed for replication in 36,477 European and 1078 non-European asthma patients. Functional effects on DNA methylation were assessed in 595 Hispanic/Latino and African American asthma patients and in publicly available databases. The effect on gene expression was evaluated in silico. RESULTS One hundred and twenty-six independent variants were suggestively associated with asthma exacerbations in the discovery phase. Two variants independently replicated: rs12091010 located at vascular cell adhesion molecule-1/exostosin like glycosyltransferase-2 (VCAM1/EXTL2) (discovery: odds ratio (ORT allele ) = 0.82, p = 9.05 × 10-6 and replication: ORT allele = 0.89, p = 5.35 × 10-3 ) and rs943126 from pantothenate kinase 1 (PANK1) (discovery: ORC allele = 0.85, p = 3.10 × 10-5 and replication: ORC allele = 0.89, p = 1.30 × 10-2 ). Both variants regulate gene expression of genes where they locate and DNA methylation levels of nearby genes in whole blood. CONCLUSIONS This multi-ancestry study revealed novel suggestive regulatory loci for asthma exacerbations located in genomic regions participating in inflammation and host defense.
Collapse
Affiliation(s)
- Esther Herrera‐Luis
- Genomics and Health GroupDepartment of Biochemistry, Microbiology, Cell Biology and GeneticsUniversidad de La Laguna (ULL)San Cristóbal de La Laguna, TenerifeSpain
| | - Victor E. Ortega
- Division of Respiratory MedicineDepartment of Internal MedicineMayo ClinicScottsdaleArizonaUSA
| | - Elizabeth J. Ampleford
- Department of Internal MedicineCenter for Precision MedicineWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Yang Yie Sio
- Department of Biological SciencesNational University of SingaporeSingapore CitySingapore
| | - Raquel Granell
- MRC Integrative Epidemiology Unit (IEU)Population Health SciencesBristol Medical SchoolUniversity of BristolBristolUK
| | - Emmely de Roos
- Department of EpidemiologyErasmus University Medical CenterRotterdamThe Netherlands
- Department of Respiratory MedicineGhent University HospitalGhentBelgium
| | - Natalie Terzikhan
- Department of EpidemiologyErasmus University Medical CenterRotterdamThe Netherlands
- Department of Respiratory MedicineGhent University HospitalGhentBelgium
| | - Ernesto Elorduy Vergara
- Institute of Computation BiologyHelmholtz Zentrum MünchenGerman Research Center for Environmental HealthMunichGermany
| | - Natalia Hernandez‐Pacheco
- Department of Clinical Sciences and EducationSödersjukhusetKarolinska InstitutetStockholmSweden
- CIBER de Enfermedades Respiratorias (CIBERES)MadridSpain
| | - Javier Perez‐Garcia
- Genomics and Health GroupDepartment of Biochemistry, Microbiology, Cell Biology and GeneticsUniversidad de La Laguna (ULL)San Cristóbal de La Laguna, TenerifeSpain
| | - Elena Martin‐Gonzalez
- Genomics and Health GroupDepartment of Biochemistry, Microbiology, Cell Biology and GeneticsUniversidad de La Laguna (ULL)San Cristóbal de La Laguna, TenerifeSpain
| | - Fabian Lorenzo‐Diaz
- Genomics and Health GroupDepartment of Biochemistry, Microbiology, Cell Biology and GeneticsUniversidad de La Laguna (ULL)San Cristóbal de La Laguna, TenerifeSpain
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC)Universidad de La Laguna (ULL)San Cristóbal de La Laguna, TenerifeSpain
| | - Simone Hashimoto
- Department of Respiratory MedicineAmsterdam University Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | - Paul Brinkman
- Department of Respiratory MedicineAmsterdam University Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
| | | | - Andrea L. Jorgensen
- Department of Health Data ScienceInstitute of Population HealthUniversity of LiverpoolLiverpoolUK
| | - Qi Yan
- Department of Obstetrics and GynecologyColumbia University Irving Medical CenterNew YorkNew YorkUSA
| | - Erick Forno
- Division of Pediatric Pulmonary MedicineUPMC Children's Hospital of PittsburghUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Susanne J. Vijverberg
- Department of Respiratory MedicineAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Division of Pharmacoepidemiology and Clinical PharmacologyFaculty of ScienceUtrecht UniversityUtrechtThe Netherlands
- Department of Paediatric Respiratory Medicine and AllergyEmma's Children HospitalAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Ryan Lethem
- MRC Integrative Epidemiology Unit (IEU)Population Health SciencesBristol Medical SchoolUniversity of BristolBristolUK
| | - Antonio Espuela‐Ortiz
- Genomics and Health GroupDepartment of Biochemistry, Microbiology, Cell Biology and GeneticsUniversidad de La Laguna (ULL)San Cristóbal de La Laguna, TenerifeSpain
| | - Mario Gorenjak
- Center for Human Molecular Genetics and PharmacogenomicsFaculty of MedicineUniversity of MariborMariborSlovenia
| | - Celeste Eng
- Department of MedicineUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Ruperto González‐Pérez
- Allergy DepartmentHospital Universitario de CanariasSanta Cruz de TenerifeTenerifeSpain
- Severe Asthma Unit, Allergy DepartmentHospital Universitario de CanariasSanta Cruz de TenerifeTenerifeSpain
| | - José M. Hernández‐Pérez
- Pulmonary MedicineHospital Universitario de N.S de CandelariaSanta Cruz de TenerifeSpain
- Pulmonary MedicineHospital General de La PalmaLa Palma, Santa Cruz de TenerifeSpain
| | - Paloma Poza‐Guedes
- Allergy DepartmentHospital Universitario de CanariasSanta Cruz de TenerifeTenerifeSpain
- Severe Asthma Unit, Allergy DepartmentHospital Universitario de CanariasSanta Cruz de TenerifeTenerifeSpain
| | - Olaia Sardón
- Division of Pediatric Respiratory MedicineHospital Universitario DonostiaSan SebastiánSpain
- Department of PediatricsUniversity of the Basque Country (UPV/EHU)San SebastiánSpain
| | - Paula Corcuera
- Division of Pediatric Respiratory MedicineHospital Universitario DonostiaSan SebastiánSpain
| | - Greg A. Hawkins
- Department of BiochemistryWake Forest School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Annalisa Marsico
- Computational Health CenterHelmholtz Zentrum MünchenGerman Research Center for Environmental HealthMunichGermany
| | - Thomas Bahmer
- LungenClinic Grosshansdorf, PneumologyGrosshansdorfGermany
- Airway Research Center North (ARCN)Members of the Germany Center for Lung Research (DZL)GrosshansdorfGermany
| | - Klaus F. Rabe
- LungenClinic Grosshansdorf, PneumologyGrosshansdorfGermany
- Airway Research Center North (ARCN)Members of the Germany Center for Lung Research (DZL)GrosshansdorfGermany
| | - Gesine Hansen
- Department of Pediatric Pneumology, Allergology and NeonatologyHannover Medical SchoolHannoverGermany
| | - Matthias Volkmar Kopp
- Division of Pediatric Pneumology & AllergologyUniversity Medical Center Schleswig‐HolsteinLübeckGermany
- Airway Research Center North (ARCN)Members of the Germany Center for Lung Research (DZL)LübeckGermany
- Department of Paediatric Respiratory MedicineInselspitalUniversity Children's Hospital of BernUniversity of BernBernSwitzerland
| | - Raimon Rios
- Programa de Pós Graduação em Imunologia (PPGIm)Instituto de Ciências da SaúdeUniversidade Federal da Bahia (UFBA)SalvadorBrazil
| | - Maria Jesus Cruz
- CIBER de Enfermedades Respiratorias (CIBERES)MadridSpain
- Servicio de NeumologíaHospital Vall d’HebronBarcelonaSpain
| | | | - José María Olaguibel
- CIBER de Enfermedades Respiratorias (CIBERES)MadridSpain
- Servicio de AlergologíaComplejo Hospitalario de NavarraPamplonaNavarraSpain
| | - Vicente Plaza
- CIBER de Enfermedades Respiratorias (CIBERES)MadridSpain
- Departamento de Medicina RespiratoriaHospital de la Santa Creu i Sant PauInstituto de Investigación Biomédica Sant Pau (IIB Sant Pau)BarcelonaSpain
| | - Santiago Quirce
- CIBER de Enfermedades Respiratorias (CIBERES)MadridSpain
- Department of AllergyLa Paz University HospitalIdiPAZMadridSpain
| | - Glorisa Canino
- Behavioral Sciences Research InstituteUniversity of Puerto RicoSan JuanPuerto Rico
| | - Michelle Cloutier
- Department of PediatricsUniversity of ConnecticutFarmingtonConnecticutUSA
| | - Victoria del Pozo
- CIBER de Enfermedades Respiratorias (CIBERES)MadridSpain
- Immunology DepartmentInstituto de Investigación Sanitaria Hospital Universitario Fundación Jiménez DíazMadridSpain
| | | | - Javier Korta‐Murua
- Department of PediatricsUniversity of the Basque Country (UPV/EHU)San SebastiánSpain
| | - Jesús Villar
- CIBER de Enfermedades Respiratorias (CIBERES)MadridSpain
- Multidisciplinary Organ Dysfunction Evaluation Research NetworkResearch UnitHospital Universitario Dr. NegrínLas Palmas de Gran CanariaSpain
| | - Uroš Potočnik
- Laboratory for Biochemistry, Molecular Biology and GenomicsFaculty for Chemistry and Chemical EngineeringUniversity of MariborMariborSlovenia
| | - Camila Figueiredo
- Instituto de Ciências da SaúdeUniversidade Federal da BahiaSalvadorBrazil
| | - Michael Kabesch
- Department of Paediatric Pneumology and AllergyUniversity Children's Hospital Regensburg (KUNO)RegensburgGermany
| | - Somnath Mukhopadhyay
- Academic Department of PaediatricsBrighton and Sussex Medical School, Royal Alexandra Children's HospitalBrightonUK
- Population Pharmacogenetics GroupBiomedical Research InstituteNinewells Hospital and Medical SchoolUniversity of DundeeDundeeUK
| | - Munir Pirmohamed
- Department of Pharmacology and TherapeuticsInstitute of Systems, Molecular and Integrative BiologyUniversity of LiverpoolLiverpoolUK
| | - Daniel B. Hawcutt
- Department of Women's and Children's HealthUniversity of LiverpoolLiverpoolUK
- Alder Hey Children's HospitalLiverpoolUK
- NIHR Alder Hey Clinical Research FacilityAlder Hey Children's HospitalLiverpoolUK
| | - Erik Melén
- Department of Clinical Sciences and EducationSödersjukhusetKarolinska InstitutetStockholmSweden
- Sachs’ Children’s HospitalSouth General HospitalStockholmSweden
| | - Colin N. Palmer
- Population Pharmacogenetics GroupBiomedical Research InstituteNinewells Hospital and Medical SchoolUniversity of DundeeDundeeUK
| | | | - Anke H. Maitland‐van der Zee
- Department of Respiratory MedicineAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Division of Pharmacoepidemiology and Clinical PharmacologyFaculty of ScienceUtrecht UniversityUtrechtThe Netherlands
- Department of Paediatric Respiratory Medicine and AllergyEmma's Children HospitalAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Erika von Mutius
- Institute for Asthma and Allergy PreventionHelmholtz Zentrum MünchenGerman Research Center for Environmental HealthMunichGermany
- Dr von Hauner Children's HospitalLudwig‐Maximilians‐UniversitätMunichGermany
- Comprehensive Pneumology Center Munich (CPC‐M)Member of the German Center for Lung ResearchMunichGermany
| | - Juan C. Celedón
- Division of Pediatric Pulmonary MedicineUPMC Children's Hospital of PittsburghUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Guy Brusselle
- Department of EpidemiologyErasmus University Medical CenterRotterdamThe Netherlands
- Department of Respiratory MedicineGhent University HospitalGhentBelgium
- Department of Respiratory MedicineErasmus University Medical CenterRotterdamThe Netherlands
| | - Fook Tim Chew
- Department of Biological SciencesNational University of SingaporeSingapore CitySingapore
| | - Eugene Bleecker
- Division of Genetics, Genomics, and Precision MedicineDepartment of Internal MedicineUniversity of Arizona College of MedicineTucsonArizonaUSA
| | - Deborah Meyers
- Division of Genetics, Genomics, and Precision MedicineDepartment of Internal MedicineUniversity of Arizona College of MedicineTucsonArizonaUSA
| | - Esteban G. Burchard
- Severe Asthma Unit, Allergy DepartmentHospital Universitario de CanariasSanta Cruz de TenerifeTenerifeSpain
- Department of Bioengineering and Therapeutic SciencesUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Maria Pino‐Yanes
- Genomics and Health GroupDepartment of Biochemistry, Microbiology, Cell Biology and GeneticsUniversidad de La Laguna (ULL)San Cristóbal de La Laguna, TenerifeSpain
- CIBER de Enfermedades Respiratorias (CIBERES)MadridSpain
- Instituto de Tecnologías Biomédicas (ITB)Universidad de La Laguna (ULL)San Cristóbal de La Laguna, TenerifeSpain
| |
Collapse
|
155
|
Ament Z, Patki A, Chaudhary N, Bhave VM, Garcia Guarniz AL, Gao Y, Gerszten RE, Correa A, Judd SE, Cushman M, Long DL, Irvin MR, Kimberly WT. Nucleosides Associated With Incident Ischemic Stroke in the REGARDS and JHS Cohorts. Neurology 2022; 98:e2097-e2107. [PMID: 35264422 PMCID: PMC9169945 DOI: 10.1212/wnl.0000000000200262] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 02/04/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Both genetic and environmental factors contribute to stroke risk. We sought to identify novel metabolites associated with incident stroke in the Reasons for Geographic and Racial Differences in Stroke (REGARDS) cohort and determine whether they reflected genetic or environmental variation. METHODS This was a stroke case-cohort observational study nested in REGARDS. Cases were defined as incident stroke and metabolomic profiles were compared to a randomly selected control cohort. In baseline plasma samples, 162 metabolites were measured using liquid chromatography-tandem mass spectrometry. Cox proportional hazards models were adjusted for age, sex, race, and age by race in the base model. Fully adjusted models included traditional stroke risk factors. Mediation analyses conducted for these stroke risk factors used the metabolite as mediator. Genome-wide associations with the leading candidate metabolites were calculated using array data. Replication analyses in the Jackson Heart Study (JHS) were conducted using random effects meta-analysis. RESULTS There were 2,043 participants who were followed over an average period of 7.1 years, including 1,075 stroke cases and 968 random controls. Nine metabolites were associated with stroke in the base model, 8 of which were measured and remained significant in meta-analysis with JHS. In the fully adjusted model in REGARDS, guanosine (hazard ratio [HR] 1.34, 95% CI 1.18-1.53; p = 7.26 × 10-6) and pseudouridine (HR 1.28, 95% CI 1.13-1.45; p = 1.03 × 10-4) were associated with incident ischemic stroke following Bonferroni adjustment. Guanosine also partially mediated the relationship between hypertension and stroke (17.6%) and pseudouridine did not mediate any risk factor. Genome-wide association analysis identified loci rs34631560 and rs34631560 associated with pseudouridine, but these did not explain the association of pseudouridine with stroke. DISCUSSION Guanosine and pseudouridine are nucleosides associated with incident ischemic stroke independently of other risk factors. Genetic and mediation analyses suggest that environmental exposures rather than genetic variation link nucleoside levels to stroke risk. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that guanosine and pseudouridine are associated with incident stroke.
Collapse
Affiliation(s)
- Zsuzsanna Ament
- From the Center for Genomic Medicine, Harvard Medical School (Z.A., W.T.K.), and Department of Neurology (Z.A., A.-L.G.G., W.T.K.), Massachusetts General Hospital, Boston; Departments of Epidemiology (A.P., N.C., R.M.I.) and Biostatistics (S.E.J., L.L.), School of Public Health, University of Alabama at Birmingham; Harvard Medical School (V.M.B.), Boston, MA; The Jackson Heart Study (Y.G., A.C.), University of Mississippi Medical Center, Jackson; Department of Medicine (R.E.G.), Beth Israel Deaconess Medical Center, Boston, MA; and Department of Medicine (M.C.), Larner College of Medicine at the University of Vermont, Burlington
| | - Amit Patki
- From the Center for Genomic Medicine, Harvard Medical School (Z.A., W.T.K.), and Department of Neurology (Z.A., A.-L.G.G., W.T.K.), Massachusetts General Hospital, Boston; Departments of Epidemiology (A.P., N.C., R.M.I.) and Biostatistics (S.E.J., L.L.), School of Public Health, University of Alabama at Birmingham; Harvard Medical School (V.M.B.), Boston, MA; The Jackson Heart Study (Y.G., A.C.), University of Mississippi Medical Center, Jackson; Department of Medicine (R.E.G.), Beth Israel Deaconess Medical Center, Boston, MA; and Department of Medicine (M.C.), Larner College of Medicine at the University of Vermont, Burlington
| | - Ninad Chaudhary
- From the Center for Genomic Medicine, Harvard Medical School (Z.A., W.T.K.), and Department of Neurology (Z.A., A.-L.G.G., W.T.K.), Massachusetts General Hospital, Boston; Departments of Epidemiology (A.P., N.C., R.M.I.) and Biostatistics (S.E.J., L.L.), School of Public Health, University of Alabama at Birmingham; Harvard Medical School (V.M.B.), Boston, MA; The Jackson Heart Study (Y.G., A.C.), University of Mississippi Medical Center, Jackson; Department of Medicine (R.E.G.), Beth Israel Deaconess Medical Center, Boston, MA; and Department of Medicine (M.C.), Larner College of Medicine at the University of Vermont, Burlington
| | - Varun M Bhave
- From the Center for Genomic Medicine, Harvard Medical School (Z.A., W.T.K.), and Department of Neurology (Z.A., A.-L.G.G., W.T.K.), Massachusetts General Hospital, Boston; Departments of Epidemiology (A.P., N.C., R.M.I.) and Biostatistics (S.E.J., L.L.), School of Public Health, University of Alabama at Birmingham; Harvard Medical School (V.M.B.), Boston, MA; The Jackson Heart Study (Y.G., A.C.), University of Mississippi Medical Center, Jackson; Department of Medicine (R.E.G.), Beth Israel Deaconess Medical Center, Boston, MA; and Department of Medicine (M.C.), Larner College of Medicine at the University of Vermont, Burlington
| | - Ana-Lucia Garcia Guarniz
- From the Center for Genomic Medicine, Harvard Medical School (Z.A., W.T.K.), and Department of Neurology (Z.A., A.-L.G.G., W.T.K.), Massachusetts General Hospital, Boston; Departments of Epidemiology (A.P., N.C., R.M.I.) and Biostatistics (S.E.J., L.L.), School of Public Health, University of Alabama at Birmingham; Harvard Medical School (V.M.B.), Boston, MA; The Jackson Heart Study (Y.G., A.C.), University of Mississippi Medical Center, Jackson; Department of Medicine (R.E.G.), Beth Israel Deaconess Medical Center, Boston, MA; and Department of Medicine (M.C.), Larner College of Medicine at the University of Vermont, Burlington
| | - Yan Gao
- From the Center for Genomic Medicine, Harvard Medical School (Z.A., W.T.K.), and Department of Neurology (Z.A., A.-L.G.G., W.T.K.), Massachusetts General Hospital, Boston; Departments of Epidemiology (A.P., N.C., R.M.I.) and Biostatistics (S.E.J., L.L.), School of Public Health, University of Alabama at Birmingham; Harvard Medical School (V.M.B.), Boston, MA; The Jackson Heart Study (Y.G., A.C.), University of Mississippi Medical Center, Jackson; Department of Medicine (R.E.G.), Beth Israel Deaconess Medical Center, Boston, MA; and Department of Medicine (M.C.), Larner College of Medicine at the University of Vermont, Burlington
| | - Robert E Gerszten
- From the Center for Genomic Medicine, Harvard Medical School (Z.A., W.T.K.), and Department of Neurology (Z.A., A.-L.G.G., W.T.K.), Massachusetts General Hospital, Boston; Departments of Epidemiology (A.P., N.C., R.M.I.) and Biostatistics (S.E.J., L.L.), School of Public Health, University of Alabama at Birmingham; Harvard Medical School (V.M.B.), Boston, MA; The Jackson Heart Study (Y.G., A.C.), University of Mississippi Medical Center, Jackson; Department of Medicine (R.E.G.), Beth Israel Deaconess Medical Center, Boston, MA; and Department of Medicine (M.C.), Larner College of Medicine at the University of Vermont, Burlington
| | - Adolfo Correa
- From the Center for Genomic Medicine, Harvard Medical School (Z.A., W.T.K.), and Department of Neurology (Z.A., A.-L.G.G., W.T.K.), Massachusetts General Hospital, Boston; Departments of Epidemiology (A.P., N.C., R.M.I.) and Biostatistics (S.E.J., L.L.), School of Public Health, University of Alabama at Birmingham; Harvard Medical School (V.M.B.), Boston, MA; The Jackson Heart Study (Y.G., A.C.), University of Mississippi Medical Center, Jackson; Department of Medicine (R.E.G.), Beth Israel Deaconess Medical Center, Boston, MA; and Department of Medicine (M.C.), Larner College of Medicine at the University of Vermont, Burlington
| | - Suzanne E Judd
- From the Center for Genomic Medicine, Harvard Medical School (Z.A., W.T.K.), and Department of Neurology (Z.A., A.-L.G.G., W.T.K.), Massachusetts General Hospital, Boston; Departments of Epidemiology (A.P., N.C., R.M.I.) and Biostatistics (S.E.J., L.L.), School of Public Health, University of Alabama at Birmingham; Harvard Medical School (V.M.B.), Boston, MA; The Jackson Heart Study (Y.G., A.C.), University of Mississippi Medical Center, Jackson; Department of Medicine (R.E.G.), Beth Israel Deaconess Medical Center, Boston, MA; and Department of Medicine (M.C.), Larner College of Medicine at the University of Vermont, Burlington
| | - Mary Cushman
- From the Center for Genomic Medicine, Harvard Medical School (Z.A., W.T.K.), and Department of Neurology (Z.A., A.-L.G.G., W.T.K.), Massachusetts General Hospital, Boston; Departments of Epidemiology (A.P., N.C., R.M.I.) and Biostatistics (S.E.J., L.L.), School of Public Health, University of Alabama at Birmingham; Harvard Medical School (V.M.B.), Boston, MA; The Jackson Heart Study (Y.G., A.C.), University of Mississippi Medical Center, Jackson; Department of Medicine (R.E.G.), Beth Israel Deaconess Medical Center, Boston, MA; and Department of Medicine (M.C.), Larner College of Medicine at the University of Vermont, Burlington
| | - D Leann Long
- From the Center for Genomic Medicine, Harvard Medical School (Z.A., W.T.K.), and Department of Neurology (Z.A., A.-L.G.G., W.T.K.), Massachusetts General Hospital, Boston; Departments of Epidemiology (A.P., N.C., R.M.I.) and Biostatistics (S.E.J., L.L.), School of Public Health, University of Alabama at Birmingham; Harvard Medical School (V.M.B.), Boston, MA; The Jackson Heart Study (Y.G., A.C.), University of Mississippi Medical Center, Jackson; Department of Medicine (R.E.G.), Beth Israel Deaconess Medical Center, Boston, MA; and Department of Medicine (M.C.), Larner College of Medicine at the University of Vermont, Burlington
| | - M Ryan Irvin
- From the Center for Genomic Medicine, Harvard Medical School (Z.A., W.T.K.), and Department of Neurology (Z.A., A.-L.G.G., W.T.K.), Massachusetts General Hospital, Boston; Departments of Epidemiology (A.P., N.C., R.M.I.) and Biostatistics (S.E.J., L.L.), School of Public Health, University of Alabama at Birmingham; Harvard Medical School (V.M.B.), Boston, MA; The Jackson Heart Study (Y.G., A.C.), University of Mississippi Medical Center, Jackson; Department of Medicine (R.E.G.), Beth Israel Deaconess Medical Center, Boston, MA; and Department of Medicine (M.C.), Larner College of Medicine at the University of Vermont, Burlington
| | - W Taylor Kimberly
- From the Center for Genomic Medicine, Harvard Medical School (Z.A., W.T.K.), and Department of Neurology (Z.A., A.-L.G.G., W.T.K.), Massachusetts General Hospital, Boston; Departments of Epidemiology (A.P., N.C., R.M.I.) and Biostatistics (S.E.J., L.L.), School of Public Health, University of Alabama at Birmingham; Harvard Medical School (V.M.B.), Boston, MA; The Jackson Heart Study (Y.G., A.C.), University of Mississippi Medical Center, Jackson; Department of Medicine (R.E.G.), Beth Israel Deaconess Medical Center, Boston, MA; and Department of Medicine (M.C.), Larner College of Medicine at the University of Vermont, Burlington
| |
Collapse
|
156
|
Chimusa ER, Defo J. Dissecting Meta-Analysis in GWAS Era: Bayesian Framework for Gene/Subnetwork-Specific Meta-Analysis. Front Genet 2022; 13:838518. [PMID: 35664319 PMCID: PMC9159898 DOI: 10.3389/fgene.2022.838518] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
Over the past decades, advanced high-throughput technologies have continuously contributed to genome-wide association studies (GWASs). GWAS meta-analysis has been increasingly adopted, has cross-ancestry replicability, and has power to illuminate the genetic architecture of complex traits, informing about the reliability of estimation effects and their variability across human ancestries. However, detecting genetic variants that have low disease risk still poses a challenge. Designing a meta-analysis approach that combines the effect of various SNPs within genes or genes within pathways from multiple independent population GWASs may be helpful in identifying associations with small effect sizes and increasing the association power. Here, we proposed ancMETA, a Bayesian graph-based framework, to perform the gene/pathway-specific meta-analysis by combining the effect size of multiple SNPs within genes, and genes within subnetwork/pathways across multiple independent population GWASs to deconvolute the interactions between genes underlying the pathogenesis of complex diseases across human populations. We assessed the proposed framework on simulated datasets, and the results show that the proposed model holds promise for increasing statistical power for meta-analysis of genetic variants underlying the pathogenesis of complex diseases. To illustrate the proposed meta-analysis framework, we leverage seven different European bipolar disorder (BD) cohorts, and we identify variants in the angiotensinogen (AGT) gene to be significantly associated with BD across all 7 studies. We detect a commonly significant BD-specific subnetwork with the ESR1 gene as the main hub of a subnetwork, associated with neurotrophin signaling (p = 4e−14) and myometrial relaxation and contraction (p = 3e−08) pathways. ancMETA provides a new contribution to post-GWAS methodologies and holds promise for comprehensively examining interactions between genes underlying the pathogenesis of genetic diseases and also underlying ethnic differences.
Collapse
|
157
|
Choudhury A, Brandenburg JT, Chikowore T, Sengupta D, Boua PR, Crowther NJ, Agongo G, Asiki G, Gómez-Olivé FX, Kisiangani I, Maimela E, Masemola-Maphutha M, Micklesfield LK, Nonterah EA, Norris SA, Sorgho H, Tinto H, Tollman S, Graham SE, Willer CJ, Hazelhurst S, Ramsay M. Meta-analysis of sub-Saharan African studies provides insights into genetic architecture of lipid traits. Nat Commun 2022; 13:2578. [PMID: 35546142 PMCID: PMC9095599 DOI: 10.1038/s41467-022-30098-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 04/18/2022] [Indexed: 12/30/2022] Open
Abstract
Genetic associations for lipid traits have identified hundreds of variants with clear differences across European, Asian and African studies. Based on a sub-Saharan-African GWAS for lipid traits in the population cross-sectional AWI-Gen cohort (N = 10,603) we report a novel LDL-C association in the GATB region (P-value=1.56 × 10-8). Meta-analysis with four other African cohorts (N = 23,718) provides supporting evidence for the LDL-C association with the GATB/FHIP1A region and identifies a novel triglyceride association signal close to the FHIT gene (P-value =2.66 × 10-8). Our data enable fine-mapping of several well-known lipid-trait loci including LDLR, PMFBP1 and LPA. The transferability of signals detected in two large global studies (GLGC and PAGE) consistently improves with an increase in the size of the African replication cohort. Polygenic risk score analysis shows increased predictive accuracy for LDL-C levels with the narrowing of genetic distance between the discovery dataset and our cohort. Novel discovery is enhanced with the inclusion of African data.
Collapse
Affiliation(s)
- Ananyo Choudhury
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - Jean-Tristan Brandenburg
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Tinashe Chikowore
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- South African Medical Research Council/University of the Witwatersrand Developmental Pathways for Health Research Unit, Department of Paediatrics, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Dhriti Sengupta
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Palwende Romuald Boua
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Clinical Research Unit of Nanoro, Institut de Recherche en Sciences de la Santè, Nanoro, Burkina Faso
| | - Nigel J Crowther
- Department of Chemical Pathology, National Health Laboratory Service, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Godfred Agongo
- Navrongo Health Research Centre, Ghana Health Service, Navrongo, Ghana
- C.K. Tedam University of Technology and Applied Sciences, Navrongo, Ghana
| | - Gershim Asiki
- African Population and Health Research Center, Nairobi, Kenya
| | - F Xavier Gómez-Olivé
- MRC/Wits Rural Public Health and Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Eric Maimela
- Department of Public Health, School of Health Care Sciences, Faculty of Health Sciences, University of Limpopo, Polokwane, South Africa
| | - Matshane Masemola-Maphutha
- Department of Pathology and Medical Sciences, School of Health Care Sciences, Faculty of Health Sciences, University of Limpopo, Polokwane, South Africa
| | - Lisa K Micklesfield
- South African Medical Research Council/University of the Witwatersrand Developmental Pathways for Health Research Unit, Department of Paediatrics, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Shane A Norris
- South African Medical Research Council/University of the Witwatersrand Developmental Pathways for Health Research Unit, Department of Paediatrics, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Hermann Sorgho
- Clinical Research Unit of Nanoro, Institut de Recherche en Sciences de la Santè, Nanoro, Burkina Faso
| | - Halidou Tinto
- Clinical Research Unit of Nanoro, Institut de Recherche en Sciences de la Santè, Nanoro, Burkina Faso
| | - Stephen Tollman
- MRC/Wits Rural Public Health and Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sarah E Graham
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Cristen J Willer
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, 48019, USA
| | - Scott Hazelhurst
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- School of Electrical and Information Engineering, 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.
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
158
|
Mahajan A, Spracklen CN, Zhang W, Ng MCY, Petty LE, Kitajima H, Yu GZ, Rüeger S, Speidel L, Kim YJ, Horikoshi M, Mercader JM, Taliun D, Moon S, Kwak SH, Robertson NR, Rayner NW, Loh M, Kim BJ, Chiou J, Miguel-Escalada I, Della Briotta Parolo P, Lin K, Bragg F, Preuss MH, Takeuchi F, Nano J, Guo X, Lamri A, Nakatochi M, Scott RA, Lee JJ, Huerta-Chagoya A, Graff M, Chai JF, Parra EJ, Yao J, Bielak LF, Tabara Y, Hai Y, Steinthorsdottir V, Cook JP, Kals M, Grarup N, Schmidt EM, Pan I, Sofer T, Wuttke M, Sarnowski C, Gieger C, Nousome D, Trompet S, Long J, Sun M, Tong L, Chen WM, Ahmad M, Noordam R, Lim VJY, Tam CHT, Joo YY, Chen CH, Raffield LM, Lecoeur C, Prins BP, Nicolas A, Yanek LR, Chen G, Jensen RA, Tajuddin S, Kabagambe EK, An P, Xiang AH, Choi HS, Cade BE, Tan J, Flanagan J, Abaitua F, Adair LS, Adeyemo A, Aguilar-Salinas CA, Akiyama M, Anand SS, Bertoni A, Bian Z, Bork-Jensen J, Brandslund I, Brody JA, Brummett CM, Buchanan TA, Canouil M, Chan JCN, Chang LC, Chee ML, Chen J, Chen SH, Chen YT, Chen Z, Chuang LM, Cushman M, et alMahajan A, Spracklen CN, Zhang W, Ng MCY, Petty LE, Kitajima H, Yu GZ, Rüeger S, Speidel L, Kim YJ, Horikoshi M, Mercader JM, Taliun D, Moon S, Kwak SH, Robertson NR, Rayner NW, Loh M, Kim BJ, Chiou J, Miguel-Escalada I, Della Briotta Parolo P, Lin K, Bragg F, Preuss MH, Takeuchi F, Nano J, Guo X, Lamri A, Nakatochi M, Scott RA, Lee JJ, Huerta-Chagoya A, Graff M, Chai JF, Parra EJ, Yao J, Bielak LF, Tabara Y, Hai Y, Steinthorsdottir V, Cook JP, Kals M, Grarup N, Schmidt EM, Pan I, Sofer T, Wuttke M, Sarnowski C, Gieger C, Nousome D, Trompet S, Long J, Sun M, Tong L, Chen WM, Ahmad M, Noordam R, Lim VJY, Tam CHT, Joo YY, Chen CH, Raffield LM, Lecoeur C, Prins BP, Nicolas A, Yanek LR, Chen G, Jensen RA, Tajuddin S, Kabagambe EK, An P, Xiang AH, Choi HS, Cade BE, Tan J, Flanagan J, Abaitua F, Adair LS, Adeyemo A, Aguilar-Salinas CA, Akiyama M, Anand SS, Bertoni A, Bian Z, Bork-Jensen J, Brandslund I, Brody JA, Brummett CM, Buchanan TA, Canouil M, Chan JCN, Chang LC, Chee ML, Chen J, Chen SH, Chen YT, Chen Z, Chuang LM, Cushman M, Das SK, de Silva HJ, Dedoussis G, Dimitrov L, Doumatey AP, Du S, Duan Q, Eckardt KU, Emery LS, Evans DS, Evans MK, Fischer K, Floyd JS, Ford I, Fornage M, Franco OH, Frayling TM, Freedman BI, Fuchsberger C, Genter P, Gerstein HC, Giedraitis V, González-Villalpando C, González-Villalpando ME, Goodarzi MO, Gordon-Larsen P, Gorkin D, Gross M, Guo Y, Hackinger S, Han S, Hattersley AT, Herder C, Howard AG, Hsueh W, Huang M, Huang W, Hung YJ, Hwang MY, Hwu CM, Ichihara S, Ikram MA, Ingelsson M, Islam MT, Isono M, Jang HM, Jasmine F, Jiang G, Jonas JB, Jørgensen ME, Jørgensen T, Kamatani Y, Kandeel FR, Kasturiratne A, Katsuya T, Kaur V, Kawaguchi T, Keaton JM, Kho AN, Khor CC, Kibriya MG, Kim DH, Kohara K, Kriebel J, Kronenberg F, Kuusisto J, Läll K, Lange LA, Lee MS, Lee NR, Leong A, Li L, Li Y, Li-Gao R, Ligthart S, Lindgren CM, Linneberg A, Liu CT, Liu J, Locke AE, Louie T, Luan J, Luk AO, Luo X, Lv J, Lyssenko V, Mamakou V, Mani KR, Meitinger T, Metspalu A, Morris AD, Nadkarni GN, Nadler JL, Nalls MA, Nayak U, Nongmaithem SS, Ntalla I, Okada Y, Orozco L, Patel SR, Pereira MA, Peters A, Pirie FJ, Porneala B, Prasad G, Preissl S, Rasmussen-Torvik LJ, Reiner AP, Roden M, Rohde R, Roll K, Sabanayagam C, Sander M, Sandow K, Sattar N, Schönherr S, Schurmann C, Shahriar M, Shi J, Shin DM, Shriner D, Smith JA, So WY, Stančáková A, Stilp AM, Strauch K, Suzuki K, Takahashi A, Taylor KD, Thorand B, Thorleifsson G, Thorsteinsdottir U, Tomlinson B, Torres JM, Tsai FJ, Tuomilehto J, Tusie-Luna T, Udler MS, Valladares-Salgado A, van Dam RM, van Klinken JB, Varma R, Vujkovic M, Wacher-Rodarte N, Wheeler E, Whitsel EA, Wickremasinghe AR, van Dijk KW, Witte DR, Yajnik CS, Yamamoto K, Yamauchi T, Yengo L, Yoon K, Yu C, Yuan JM, Yusuf S, Zhang L, Zheng W, Raffel LJ, Igase M, Ipp E, Redline S, Cho YS, Lind L, Province MA, Hanis CL, Peyser PA, Ingelsson E, Zonderman AB, Psaty BM, Wang YX, Rotimi CN, Becker DM, Matsuda F, Liu Y, Zeggini E, Yokota M, Rich SS, Kooperberg C, Pankow JS, Engert JC, Chen YDI, Froguel P, Wilson JG, Sheu WHH, Kardia SLR, Wu JY, Hayes MG, Ma RCW, Wong TY, Groop L, Mook-Kanamori DO, Chandak GR, Collins FS, Bharadwaj D, Paré G, Sale MM, Ahsan H, Motala AA, Shu XO, Park KS, Jukema JW, Cruz M, McKean-Cowdin R, Grallert H, Cheng CY, Bottinger EP, Dehghan A, Tai ES, Dupuis J, Kato N, Laakso M, Köttgen A, Koh WP, Palmer CNA, Liu S, Abecasis G, Kooner JS, Loos RJF, North KE, Haiman CA, Florez JC, Saleheen D, Hansen T, Pedersen O, Mägi R, Langenberg C, Wareham NJ, Maeda S, Kadowaki T, Lee J, Millwood IY, Walters RG, Stefansson K, Myers SR, Ferrer J, Gaulton KJ, Meigs JB, Mohlke KL, Gloyn AL, Bowden DW, Below JE, Chambers JC, Sim X, Boehnke M, Rotter JI, McCarthy MI, Morris AP. Multi-ancestry genetic study of type 2 diabetes highlights the power of diverse populations for discovery and translation. Nat Genet 2022; 54:560-572. [PMID: 35551307 PMCID: PMC9179018 DOI: 10.1038/s41588-022-01058-3] [Show More Authors] [Citation(s) in RCA: 388] [Impact Index Per Article: 129.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/23/2022] [Indexed: 02/02/2023]
Abstract
We assembled an ancestrally diverse collection of genome-wide association studies (GWAS) of type 2 diabetes (T2D) in 180,834 affected individuals and 1,159,055 controls (48.9% non-European descent) through the Diabetes Meta-Analysis of Trans-Ethnic association studies (DIAMANTE) Consortium. Multi-ancestry GWAS meta-analysis identified 237 loci attaining stringent genome-wide significance (P < 5 × 10-9), which were delineated to 338 distinct association signals. Fine-mapping of these signals was enhanced by the increased sample size and expanded population diversity of the multi-ancestry meta-analysis, which localized 54.4% of T2D associations to a single variant with >50% posterior probability. This improved fine-mapping enabled systematic assessment of candidate causal genes and molecular mechanisms through which T2D associations are mediated, laying the foundations for functional investigations. Multi-ancestry genetic risk scores enhanced transferability of T2D prediction across diverse populations. Our study provides a step toward more effective clinical translation of T2D GWAS to improve global health for all, irrespective of genetic background.
Collapse
Affiliation(s)
- Anubha Mahajan
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Genentech, South San Francisco, CA, USA.
| | - Cassandra N Spracklen
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Epidemiology and Biostatistics, University of Massachusetts Amherst, Amherst, MA, USA
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, London North West Healthcare NHS Trust, London, UK
| | - Maggie C Y Ng
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Lauren E Petty
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hidetoshi Kitajima
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- The Advanced Research Center for Innovations in Next-Generation Medicine (INGEM), Tohoku University, Sendai, Japan
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
- Cancer Center, Tohoku University Hospital, Tohoku University, Sendai, Japan
| | - Grace Z Yu
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sina Rüeger
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Leo Speidel
- Genetics Institute, University College London, London, UK
- Francis Crick Institute, London, UK
| | - Young Jin Kim
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju-si, Republic of Korea
| | - Momoko Horikoshi
- Laboratory for Genomics of Diabetes and Metabolism, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Josep M Mercader
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Daniel Taliun
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Sanghoon Moon
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju-si, Republic of Korea
| | - Soo-Heon Kwak
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Neil R Robertson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Nigel W Rayner
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Human Genetics, Wellcome Sanger Institute, Hinxton, UK
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Marie Loh
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Translational Laboratory in Genetic Medicine (TLGM), Agency for Science, Technology and Research (A*STAR) and National University of Singapore (NUS), Singapore, Singapore
| | - Bong-Jo Kim
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju-si, Republic of Korea
| | - Joshua Chiou
- Biomedical Sciences Graduate Studies Program, University of California San Diego, La Jolla, CA, USA
- Internal Medicine Research Unit, Pfizer Worldwide Research, Cambridge, MA, USA
| | - Irene Miguel-Escalada
- Regulatory Genomics and Diabetes, Centre for Genomic Regulation, the Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM), Madrid, Spain
| | | | - Kuang Lin
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Fiona Bragg
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, UK
| | - Michael H Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fumihiko Takeuchi
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Jana Nano
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, the Lundquist Institute for Biomedical Innovation (formerly Los Angeles Biomedical Research Institute) at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Amel Lamri
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Robert A Scott
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Jung-Jin Lee
- Division of Translational Medicine and Human Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Alicia Huerta-Chagoya
- Consejo Nacional de Ciencia y Tecnología (CONACYT), Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Departamento de Medicina Genómica y Toxicologí, a AmbientalInstituto de Investigaciones Biomédicas, UNAM, Ciudad de Mexico, Mexico, Mexico
| | - Mariaelisa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jin-Fang Chai
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Esteban J Parra
- Department of Anthropology, University of Toronto at Mississauga, Mississauga, Ontario, Canada
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, the Lundquist Institute for Biomedical Innovation (formerly Los Angeles Biomedical Research Institute) at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Lawrence F Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Yasuharu Tabara
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yang Hai
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, the Lundquist Institute for Biomedical Innovation (formerly Los Angeles Biomedical Research Institute) at Harbor-UCLA Medical Center, Torrance, CA, USA
| | | | - James P Cook
- Department of Health Data Science, University of Liverpool, Liverpool, UK
| | - Mart Kals
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ellen M Schmidt
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Ian Pan
- Department of Epidemiology, Brown University School of Public Health, Providence, RI, USA
| | - Tamar Sofer
- Department of Biostatistics, Harvard University, Boston, MA, USA
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard University, Boston, MA, USA
| | - Matthias Wuttke
- Institute of Genetic Epidemiology, Department of Data Driven Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Chloe Sarnowski
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Department of Epidemiology, Human Genetics, and Environmental Sciences, the University of Texas Health Science Center at Houston, School of Public Health, Houston, TX, USA
| | - Christian Gieger
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Darryl Nousome
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Jirong Long
- Division of Epidemiology, Department of Medicine, Institute for Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Meng Sun
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Lin Tong
- Institute for Population and Precision Health, the University of Chicago, Chicago, IL, USA
| | - Wei-Min Chen
- Department of Public Health Sciences and Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Meraj Ahmad
- Genomic Research on Complex Diseases (GRC-Group), CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad, India
| | - Raymond Noordam
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Victor J Y Lim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Claudia H T Tam
- Department of Medicine and Therapeutics, the Chinese University of Hong Kong, Hong Kong, China
- Chinese University of Hong Kong-Shanghai Jiao Tong University Joint Research Centre in Diabetes Genomics and Precision Medicine, the Chinese University of Hong Kong, Hong Kong, China
| | - Yoonjung Yoonie Joo
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Health and Biomedical Informatics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Institute of Data Science, Korea University, Seoul, South Korea
| | - Chien-Hsiun Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Cécile Lecoeur
- Inserm U1283, CNRS UMR 8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille, France
- University of Lille, Lille University Hospital, Lille, France
| | - Bram Peter Prins
- Department of Human Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Aude Nicolas
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Lisa R Yanek
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Guanjie Chen
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Richard A Jensen
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Salman Tajuddin
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Edmond K Kabagambe
- Division of Epidemiology, Department of Medicine, Institute for Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Academics, Ochsner Health, New Orleans, LA, USA
| | - Ping An
- Division of Statistical Genomics, Washington University School of Medicine, St Louis, MO, USA
| | - Anny H Xiang
- Department of Research and Evaluation, Division of Biostatistics Research, Kaiser Permanente of Southern California, Pasadena, CA, USA
| | - Hyeok Sun Choi
- Department of Biomedical Science, Hallym University, Chuncheon, South Korea
| | - Brian E Cade
- Harvard Medical School, Boston, MA, USA
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
| | - Jingyi Tan
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, the Lundquist Institute for Biomedical Innovation (formerly Los Angeles Biomedical Research Institute) at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jack Flanagan
- Laboratory for Genomics of Diabetes and Metabolism, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Health Data Science, University of Liverpool, Liverpool, UK
| | - Fernando Abaitua
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Vertex Pharmaceuticals Ltd, Oxford, UK
| | - Linda S Adair
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adebowale Adeyemo
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Carlos A Aguilar-Salinas
- Unidad de Investigación en Enfermedades Metabólicas and Departamento de Endocrinología y Metabolismo, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Masato Akiyama
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Ocular Pathology and Imaging Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Sonia S Anand
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Alain Bertoni
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Zheng Bian
- Chinese Academy of Medical Sciences, Beijing, China
| | - Jette Bork-Jensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ivan Brandslund
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
- Department of Clinical Biochemistry, Vejle Hospital, Vejle, Denmark
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Chad M Brummett
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Thomas A Buchanan
- Department of Medicine, Division of Endocrinology and Diabetes, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Mickaël Canouil
- Inserm U1283, CNRS UMR 8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille, France
- University of Lille, Lille University Hospital, Lille, France
| | - Juliana C N Chan
- Department of Medicine and Therapeutics, the Chinese University of Hong Kong, Hong Kong, China
- Chinese University of Hong Kong-Shanghai Jiao Tong University Joint Research Centre in Diabetes Genomics and Precision Medicine, the Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, the Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, the Chinese University of Hong Kong, Hong Kong, China
| | - Li-Ching Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Miao-Li Chee
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Ji Chen
- Wellcome Sanger Institute, Hinxton, UK
- Exeter Centre of Excellence in Diabetes (ExCEeD), Exeter Medical School, University of Exeter, Exeter, UK
| | - Shyh-Huei Chen
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Yuan-Tsong Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Zhengming Chen
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, UK
| | - Lee-Ming Chuang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan
| | - Mary Cushman
- Department of Medicine, University of Vermont, Colchester, VT, USA
| | - Swapan K Das
- Section on Endocrinology and Metabolism, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - H Janaka de Silva
- Department of Medicine, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| | - George Dedoussis
- Department of Nutrition and Dietetics, Harokopio University of Athens, Athens, Greece
| | - Latchezar Dimitrov
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Ayo P Doumatey
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shufa Du
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Qing Duan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Leslie S Emery
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Daniel S Evans
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Krista Fischer
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - James S Floyd
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Ian Ford
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| | - Myriam Fornage
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Oscar H Franco
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Timothy M Frayling
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Barry I Freedman
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Christian Fuchsberger
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
- Institute for Biomedicine, Eurac Research, Affiliated Institute of the University of Lübeck, Bolzano, Italy
| | - Pauline Genter
- Department of Medicine, Division of Endocrinology and Metabolism, Lundquist Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Hertzel C Gerstein
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Vilmantas Giedraitis
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Clicerio González-Villalpando
- Centro de Estudios en Diabetes, Unidad de Investigacion en Diabetes y Riesgo Cardiovascular, Centro de Investigacion en Salud Poblacional, Instituto Nacional de Salud Publica, Mexico City, Mexico
| | - Maria Elena González-Villalpando
- Centro de Estudios en Diabetes, Unidad de Investigacion en Diabetes y Riesgo Cardiovascular, Centro de Investigacion en Salud Poblacional, Instituto Nacional de Salud Publica, Mexico City, Mexico
| | - Mark O Goodarzi
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Penny Gordon-Larsen
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David Gorkin
- Center for Epigenomics, University of California San Diego, La Jolla, CA, USA
| | - Myron Gross
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Yu Guo
- Chinese Academy of Medical Sciences, Beijing, China
| | - Sophie Hackinger
- Department of Human Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Sohee Han
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju-si, Republic of Korea
| | | | - Christian Herder
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Annie-Green Howard
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Willa Hsueh
- Department of Internal Medicine, Diabetes and Metabolism Research Center, the Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Mengna Huang
- Department of Epidemiology, Brown University School of Public Health, Providence, RI, USA
- Center for Global Cardiometabolic Health, Brown University, Providence, RI, USA
| | - Wei Huang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai (CHGC) and Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Shanghai, China
| | - Yi-Jen Hung
- Division of Endocrine and Metabolism, Tri-Service General Hospital Songshan Branch, Taipei, Taiwan
- School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Mi Yeong Hwang
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju-si, Republic of Korea
| | - Chii-Min Hwu
- Section of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Sahoko Ichihara
- Department of Environmental and Preventive Medicine, Jichi Medical University School of Medicine, Shimotsuke, Japan
| | - Mohammad Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Martin Ingelsson
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | | | - Masato Isono
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hye-Mi Jang
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju-si, Republic of Korea
| | - Farzana Jasmine
- Institute for Population and Precision Health, the University of Chicago, Chicago, IL, USA
| | - Guozhi Jiang
- Department of Medicine and Therapeutics, the Chinese University of Hong Kong, Hong Kong, China
- Chinese University of Hong Kong-Shanghai Jiao Tong University Joint Research Centre in Diabetes Genomics and Precision Medicine, the Chinese University of Hong Kong, Hong Kong, China
| | - Jost B Jonas
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
| | - Marit E Jørgensen
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- National Institute of Public Health, Southern Denmark University, Copenhagen, Denmark
| | - Torben Jørgensen
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - Yoichiro Kamatani
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, the University of Tokyo, Tokyo, Japan
| | - Fouad R Kandeel
- Department of Clinical Diabetes, Endocrinology & Metabolism, Department of Translational Research and Cellular Therapeutics, City of Hope, Duarte, CA, USA
| | | | - Tomohiro Katsuya
- Department of Clinical Gene Therapy, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Geriatric and General Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Varinderpal Kaur
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Takahisa Kawaguchi
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Jacob M Keaton
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Division of Epidemiology, Department of Medicine, Institute for Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Abel N Kho
- Division of General Internal Medicine and Geriatrics, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Health Information Partnerships, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Chiea-Chuen Khor
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Muhammad G Kibriya
- Institute for Population and Precision Health, the University of Chicago, Chicago, IL, USA
| | - Duk-Hwan Kim
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Katsuhiko Kohara
- Department of Regional Resource Management, Ehime University Faculty of Collaborative Regional Innovation, Ehime, Japan
- Ibusuki Kozenkai Hospital, Ibusuki, Japan
| | - Jennifer Kriebel
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Kristi Läll
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Mathematics and Statistics, University of Tartu, Tartu, Estonia
| | - Leslie A Lange
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Myung-Shik Lee
- Severance Biomedical Science Institute and Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Nanette R Lee
- USC-Office of Population Studies Foundation, Inc., University of San Carlos, Cebu City, Philippines
| | - Aaron Leong
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Liming Li
- Department of Epidemiology and Biostatistics, Peking University Health Science Centre, Peking University, Beijing, China
| | - Yun Li
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Symen Ligthart
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Cecilia M Lindgren
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Program in Medical & Population Genetics, Broad Institute, Cambridge, MA, USA
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Allan Linneberg
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Jianjun Liu
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
| | - Adam E Locke
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, MO, USA
- Department of Medicine, Division of Genomics and Bioinformatics, Washington University School of Medicine, St Louis, MO, USA
- Regeneron Genetics Center, Tarrytown, NY, USA
| | - Tin Louie
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Jian'an Luan
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Andrea O Luk
- Department of Medicine and Therapeutics, the Chinese University of Hong Kong, Hong Kong, China
- Chinese University of Hong Kong-Shanghai Jiao Tong University Joint Research Centre in Diabetes Genomics and Precision Medicine, the Chinese University of Hong Kong, Hong Kong, China
| | - Xi Luo
- Department of Biostatistics and Data Science, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jun Lv
- Department of Epidemiology and Biostatistics, Peking University Health Science Centre, Peking University, Beijing, China
| | - Valeriya Lyssenko
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University Diabetes Centre, Malmö, Sweden
- Department of Clinical Science, Center for Diabetes Research, University of Bergen, Bergen, Norway
| | - Vasiliki Mamakou
- Dromokaiteio Psychiatric Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - K Radha Mani
- Genomic Research on Complex Diseases (GRC-Group), CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad, India
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Human Genetics, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Andres Metspalu
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Andrew D Morris
- The Usher Institute to the Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Girish N Nadkarni
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Digital Health Center, Digital Engineering Faculty of Hasso Plattner Institue and University Potsdam, Potsdam, Germany
- The Division of Data Driven and Digital Medicine (D3M), Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jerry L Nadler
- Department of Medicine and Pharmacology, New York Medical College, Valhalla, NY, USA
| | - Michael A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International LLC, Glen Echo, MD, USA
- Center for Alzheimer's and Related Dementias, National Institutes of Health, Baltimore, MD, USA
| | - Uma Nayak
- Department of Public Health Sciences and Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Suraj S Nongmaithem
- Genomic Research on Complex Diseases (GRC-Group), CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad, India
| | - Ioanna Ntalla
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Osaka, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Lorena Orozco
- Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Sanjay R Patel
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark A Pereira
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Annette Peters
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Fraser J Pirie
- Department of Diabetes and Endocrinology, Nelson R. Mandela School of Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Bianca Porneala
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Gauri Prasad
- Academy of Scientific and Innovative Research, CSIR-Human Resource Development Centre Campus, Ghaziabad, India
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Sebastian Preissl
- Center for Epigenomics, University of California San Diego, La Jolla, CA, USA
| | - Laura J Rasmussen-Torvik
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Michael Roden
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Rebecca Rohde
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kathryn Roll
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, the Lundquist Institute for Biomedical Innovation (formerly Los Angeles Biomedical Research Institute) at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Charumathi Sabanayagam
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
| | - Maike Sander
- Department of Pediatrics, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
| | - Kevin Sandow
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, the Lundquist Institute for Biomedical Innovation (formerly Los Angeles Biomedical Research Institute) at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Sebastian Schönherr
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Claudia Schurmann
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Digital Health Center, Digital Engineering Faculty of Hasso Plattner Institue and University Potsdam, Potsdam, Germany
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mohammad Shahriar
- Institute for Population and Precision Health, the University of Chicago, Chicago, IL, USA
- Institute for Population and Precision Health (IPPH), Biological Sciences Division, the University of Chicago, Chicago, IL, USA
| | - Jinxiu Shi
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center at Shanghai (CHGC) and Shanghai Institute for Biomedical and Pharmaceutical Technologies (SIBPT), Shanghai, China
| | - Dong Mun Shin
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju-si, Republic of Korea
| | - Daniel Shriner
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Wing Yee So
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Medicine and Therapeutics, the Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, the Chinese University of Hong Kong, Hong Kong, China
| | - Alena Stančáková
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Adrienne M Stilp
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Konstantin Strauch
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Chair of Genetic Epidemiology, IBE, Faculty of Medicine, LMU Munich, Munich, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Ken Suzuki
- Laboratory for Genomics of Diabetes and Metabolism, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Atsushi Takahashi
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Genomic Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, the Lundquist Institute for Biomedical Innovation (formerly Los Angeles Biomedical Research Institute) at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Barbara Thorand
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | | | - Unnur Thorsteinsdottir
- deCODE Genetics, Amgen Inc., Reykjavik, Iceland
- Faculty of Medicine, University of Reykjavik, Reykjavik, Iceland
| | - Brian Tomlinson
- Department of Medicine and Therapeutics, the Chinese University of Hong Kong, Hong Kong, China
- Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Jason M Torres
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Fuu-Jen Tsai
- Department of Medical Genetics and Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Jaakko Tuomilehto
- Department of Health, Finnish Institute for Health and Welfare, Helsinki, Finland
- National School of Public Health, Madrid, Spain
- Department of Neuroscience and Preventive Medicine, Danube University Krems, Krems, Austria
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Teresa Tusie-Luna
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Departamento de Medicina Genómica y Toxiología, Ambiental Instituto de Investigaciones Biomédicas, UNAM, Mexico City, Mexico
| | - Miriam S Udler
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Adan Valladares-Salgado
- Unidad de Investigacion Medica en Bioquimica, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, IMSS, Mexico City, Mexico
| | - Rob M van Dam
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
| | - Jan B van Klinken
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
- Department of Clinical Chemistry, Laboratory of Genetic Metabolic Disease, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Rohit Varma
- Southern California Eye Institute, CHA Hollywood Presbyterian Medical Center, Los Angeles, CA, USA
| | - Marijana Vujkovic
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Niels Wacher-Rodarte
- Unidad de Investigación Médica en Epidemiologia Clinica, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, IMSS, Mexico City, Mexico
| | - Eleanor Wheeler
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Eric A Whitsel
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Ko Willems van Dijk
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
| | - Daniel R Witte
- Department of Public Health, Aarhus University, Aarhus, Denmark
- Danish Diabetes Academy, Odense, Denmark
| | - Chittaranjan S Yajnik
- Diabetology Research Centre, King Edward Memorial Hospital and Research Centre, Pune, India
| | - Ken Yamamoto
- Department of Medical Biochemistry, Kurume University School of Medicine, Kurume, Japan
| | - Toshimasa Yamauchi
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Loïc Yengo
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Kyungheon Yoon
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju-si, Republic of Korea
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, Peking University Health Science Centre, Peking University, Beijing, China
| | - Jian-Min Yuan
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Salim Yusuf
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Liang Zhang
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Institute for Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Leslie J Raffel
- Department of Pediatrics, Division of Genetic and Genomic Medicine, UCI Irvine School of Medicine, Irvine, CA, USA
| | - Michiya Igase
- Department of Anti-Aging Medicine, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Eli Ipp
- Department of Medicine, Division of Endocrinology and Metabolism, Lundquist Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Susan Redline
- Harvard Medical School, Boston, MA, USA
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Yoon Shin Cho
- Department of Biomedical Science, Hallym University, Chuncheon, South Korea
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Michael A Province
- Division of Statistical Genomics, Washington University School of Medicine, St Louis, MO, USA
| | - Craig L Hanis
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Patricia A Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Erik Ingelsson
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Services, University of Washington, Seattle, WA, USA
| | - Ya-Xing Wang
- Beijing Institute of Ophthalmology, Ophthalmology and Visual Sciences Key Laboratory, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Charles N Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Diane M Becker
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yongmei Liu
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Medicine, Division of Cardiology, Duke University School of Medicine, Durham, NC, USA
| | - Eleftheria Zeggini
- Department of Human Genetics, Wellcome Sanger Institute, Hinxton, UK
- Institute of Translational Genomics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Technical University of Munich (TUM) and Klinikum Rechts der Isar, TUM School of Medicine, Munich, Germany
| | | | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | | | - James S Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - James C Engert
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, the Lundquist Institute for Biomedical Innovation (formerly Los Angeles Biomedical Research Institute) at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Philippe Froguel
- Inserm U1283, CNRS UMR 8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille, France
- University of Lille, Lille University Hospital, Lille, France
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, London, UK
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Wayne H H Sheu
- School of Medicine, National Defense Medical Center, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Endocrinology and Metabolism, Department of Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Jer-Yuarn Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - M Geoffrey Hayes
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Anthropology, Northwestern University, Evanston, IL, USA
| | - Ronald C W Ma
- Department of Medicine and Therapeutics, the Chinese University of Hong Kong, Hong Kong, China
- Chinese University of Hong Kong-Shanghai Jiao Tong University Joint Research Centre in Diabetes Genomics and Precision Medicine, the Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, the Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, the Chinese University of Hong Kong, Hong Kong, China
| | - Tien-Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
| | - Leif Groop
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University Diabetes Centre, Malmö, Sweden
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Giriraj R Chandak
- Genomic Research on Complex Diseases (GRC-Group), CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad, India
| | - Francis S Collins
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Dwaipayan Bharadwaj
- Academy of Scientific and Innovative Research, CSIR-Human Resource Development Centre Campus, Ghaziabad, India
- Systems Genomics Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Guillaume Paré
- Population Health Research Institute, Hamilton Health Sciences and McMaster University, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Michèle M Sale
- Department of Public Health Sciences and Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Habibul Ahsan
- Institute for Population and Precision Health, the University of Chicago, Chicago, IL, USA
| | - Ayesha A Motala
- Department of Diabetes and Endocrinology, Nelson R. Mandela School of Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Institute for Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kyong-Soo Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Netherlands Heart Institute, Utrecht, the Netherlands
| | - Miguel Cruz
- Unidad de Investigacion Medica en Bioquimica, Hospital de Especialidades, Centro Medico Nacional Siglo XXI, IMSS, Mexico City, Mexico
| | - Roberta McKean-Cowdin
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Harald Grallert
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Ophthalmology and Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
| | - Erwin P Bottinger
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Digital Health Center, Digital Engineering Faculty of Hasso Plattner Institue and University Potsdam, Potsdam, Germany
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - E-Shyong Tai
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Norihiro Kato
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Department of Data Driven Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Woon-Puay Koh
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research (A*STAR), Singapore, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Colin N A Palmer
- Pat Macpherson Centre for Pharmacogenetics and Pharmacogenomics, University of Dundee, Dundee, UK
| | - Simin Liu
- Department of Epidemiology, Brown University School of Public Health, Providence, RI, USA
- Center for Global Cardiometabolic Health, Brown University, Providence, RI, USA
- Department of Medicine, Brown University Alpert School of Medicine, Providence, RI, USA
| | - Goncalo Abecasis
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Jaspal S Kooner
- Department of Cardiology, Ealing Hospital, London North West Healthcare NHS Trust, London, UK
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, Imperial College London, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Mindich Child Health and Development Institute, Ichan School of Medicine at Mount Sinai, New York, NY, USA
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christopher A Haiman
- Department of Population and Public Health Sciences, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Jose C Florez
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Danish Saleheen
- Division of Translational Medicine and Human Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA, USA
- Center for Non-Communicable Diseases, Karachi, Pakistan
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Reedik Mägi
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Computational Medicine, Berlin Institute of Health at Charité Universitätsmedizin, Berlin, Germany
| | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Shiro Maeda
- Laboratory for Genomics of Diabetes and Metabolism, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
- Division of Clinical Laboratory and Blood Transfusion, University of the Ryukyus Hospital, Okinawa, Japan
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
- Toranomon Hospital, Tokyo, Japan
| | - Juyoung Lee
- Division of Genome Science, Department of Precision Medicine, National Institute of Health, Cheongju-si, Republic of Korea
| | - Iona Y Millwood
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, UK
| | - Robin G Walters
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, UK
| | - Kari Stefansson
- deCODE Genetics, Amgen Inc., Reykjavik, Iceland
- Faculty of Medicine, University of Reykjavik, Reykjavik, Iceland
| | - Simon R Myers
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Statistics, University of Oxford, Oxford, UK
| | - Jorge Ferrer
- Regulatory Genomics and Diabetes, Centre for Genomic Regulation, the Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas asociadas (CIBERDEM), Madrid, Spain
- Section of Genetics and Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Kyle J Gaulton
- Department of Pediatrics, Pediatric Diabetes Research Center, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - James B Meigs
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anna L Gloyn
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Division of Endocrinology, Department of Pediatrics, Stanford School of Medicine, Stanford University, Stanford, CA, USA
| | - Donald W Bowden
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jennifer E Below
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John C Chambers
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, London North West Healthcare NHS Trust, London, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, Imperial College London, London, UK
| | - Xueling Sim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, the Lundquist Institute for Biomedical Innovation (formerly Los Angeles Biomedical Research Institute) at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Mark I McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
- Genentech, South San Francisco, CA, USA.
| | - Andrew P Morris
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Department of Health Data Science, University of Liverpool, Liverpool, UK.
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia.
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, University of Manchester, Manchester, UK.
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, UK.
| |
Collapse
|
159
|
Investigating the genetic architecture of eye colour in a Canadian cohort. iScience 2022; 25:104485. [PMID: 35712076 PMCID: PMC9194134 DOI: 10.1016/j.isci.2022.104485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 04/18/2022] [Accepted: 05/24/2022] [Indexed: 11/24/2022] Open
Abstract
Eye color is highly variable in populations with European ancestry, ranging from low to high quantities of melanin in the iris. Polymorphisms in the HERC2/OCA2 locus have the largest effect on eye color in these populations, although other genomic regions also influence eye color. We performed genome-wide association studies of eye color in a Canadian cohort of European ancestry (N = 5,641) and investigated candidate causal variants. We uncovered several candidate causal signals in the HERC2/OCA2 region, whereas other loci likely harbor a single causal signal. We observed colocalization of eye color signals with the expression or methylation profiles of cultured primary melanocytes. Genetic correlations of eye and hair color suggest high genome-wide pleiotropy, but locus-level differences in the genetic architecture of both traits. Overall, we provide a better picture of the polymorphisms underpinning eye color variation, which may be a consequence of specific molecular processes in the iris melanocytes. Genome-wide association studies of eye color in 5,641 participants Multiple independent candidate causal variants were identified across HERC2/OCA2 Single candidate causal variants observed on or near IRF4, SLC24A4, TYR, and TYRP1 Colocalization of eye color signals with expression and methylation profiles
Collapse
|
160
|
Perez RK, Gordon MG, Subramaniam M, Kim MC, Hartoularos GC, Targ S, Sun Y, Ogorodnikov A, Bueno R, Lu A, Thompson M, Rappoport N, Dahl A, Lanata CM, Matloubian M, Maliskova L, Kwek SS, Li T, Slyper M, Waldman J, Dionne D, Rozenblatt-Rosen O, Fong L, Dall’Era M, Balliu B, Regev A, Yazdany J, Criswell LA, Zaitlen N, Ye CJ. Single-cell RNA-seq reveals cell type-specific molecular and genetic associations to lupus. Science 2022; 376:eabf1970. [PMID: 35389781 PMCID: PMC9297655 DOI: 10.1126/science.abf1970] [Citation(s) in RCA: 234] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune disease. Knowledge of circulating immune cell types and states associated with SLE remains incomplete. We profiled more than 1.2 million peripheral blood mononuclear cells (162 cases, 99 controls) with multiplexed single-cell RNA sequencing (mux-seq). Cases exhibited elevated expression of type 1 interferon-stimulated genes (ISGs) in monocytes, reduction of naïve CD4+ T cells that correlated with monocyte ISG expression, and expansion of repertoire-restricted cytotoxic GZMH+ CD8+ T cells. Cell type-specific expression features predicted case-control status and stratified patients into two molecular subtypes. We integrated dense genotyping data to map cell type-specific cis-expression quantitative trait loci and to link SLE-associated variants to cell type-specific expression. These results demonstrate mux-seq as a systematic approach to characterize cellular composition, identify transcriptional signatures, and annotate genetic variants associated with SLE.
Collapse
Affiliation(s)
- Richard K. Perez
- School of Medicine, University of California, San Francisco, CA, USA
| | - M. Grace Gordon
- Biological and Medical Informatics Graduate Program, University of California, San Francisco, CA, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
| | - Meena Subramaniam
- Biological and Medical Informatics Graduate Program, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Min Cheol Kim
- School of Medicine, University of California, San Francisco, CA, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
- Medical Scientist Training Program, University of California, San Francisco, CA, USA
- UC Berkeley–UCSF Graduate Program in Bioengineering, San Francisco, CA, USA
| | - George C. Hartoularos
- Biological and Medical Informatics Graduate Program, University of California, San Francisco, CA, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Sasha Targ
- School of Medicine, University of California, San Francisco, CA, USA
- Biological and Medical Informatics Graduate Program, University of California, San Francisco, CA, USA
- Medical Scientist Training Program, University of California, San Francisco, CA, USA
| | - Yang Sun
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Anton Ogorodnikov
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Raymund Bueno
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Andrew Lu
- UCLA-Caltech Medical Scientist Training Program, Los Angeles, CA, USA
| | - Mike Thompson
- Department of Computer Science, University of California, Los Angeles, CA, USA
| | - Nadav Rappoport
- Department of Software and Information Systems Engineering, Ben-Gurion University of the Negev, Be’er Sheva, Israel
| | - Andrew Dahl
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Cristina M. Lanata
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Rosalind Russell/Ephraim P. Engleman Rheumatology Research Center, University of California, San Francisco, CA, USA
| | - Mehrdad Matloubian
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Rosalind Russell/Ephraim P. Engleman Rheumatology Research Center, University of California, San Francisco, CA, USA
| | - Lenka Maliskova
- Institute for Human Genetics, University of California, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Serena S. Kwek
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Tony Li
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Michal Slyper
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA
| | - Julia Waldman
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA
| | - Danielle Dionne
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA
| | | | - Lawrence Fong
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Maria Dall’Era
- School of Medicine, University of California, San Francisco, CA, USA
| | - Brunilda Balliu
- Department of Computational Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jinoos Yazdany
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Lindsey A. Criswell
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
- Rosalind Russell/Ephraim P. Engleman Rheumatology Research Center, University of California, San Francisco, CA, USA
| | - Noah Zaitlen
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Chun Jimmie Ye
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
- Rosalind Russell/Ephraim P. Engleman Rheumatology Research Center, University of California, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, USA
| |
Collapse
|
161
|
Sarnowski C, Ghanbari M, Bis JC, Logue M, Fornage M, Mishra A, Ahmad S, Beiser AS, Boerwinkle E, Bouteloup V, Chouraki V, Cupples LA, Damotte V, DeCarli CS, DeStefano AL, Djoussé L, Fohner AE, Franz CE, Kautz TF, Lambert JC, Lyons MJ, Mosley TH, Mukamal KJ, Pase MP, Portilla Fernandez EC, Rissman RA, Satizabal CL, Vasan RS, Yaqub A, Debette S, Dufouil C, Launer LJ, Kremen WS, Longstreth WT, Ikram MA, Seshadri S. Meta-analysis of genome-wide association studies identifies ancestry-specific associations underlying circulating total tau levels. Commun Biol 2022; 5:336. [PMID: 35396452 PMCID: PMC8993877 DOI: 10.1038/s42003-022-03287-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 03/17/2022] [Indexed: 01/27/2023] Open
Abstract
Circulating total-tau levels can be used as an endophenotype to identify genetic risk factors for tauopathies and related neurological disorders. Here, we confirmed and better characterized the association of the 17q21 MAPT locus with circulating total-tau in 14,721 European participants and identified three novel loci in 953 African American participants (4q31, 5p13, and 6q25) at P < 5 × 10-8. We additionally detected 14 novel loci at P < 5 × 10-7, specific to either Europeans or African Americans. Using whole-exome sequence data in 2,279 European participants, we identified ten genes associated with circulating total-tau when aggregating rare variants. Our genetic study sheds light on genes reported to be associated with neurological diseases including stroke, Alzheimer's, and Parkinson's (F5, MAP1B, and BCAS3), with Alzheimer's pathological hallmarks (ADAMTS12, IL15, and FHIT), or with an important function in the brain (PARD3, ELFN2, UBASH3B, SLIT3, and NSD3), and suggests that the genetic architecture of circulating total-tau may differ according to ancestry.
Collapse
Affiliation(s)
- Chloé Sarnowski
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Mark Logue
- National Center for PTSD, Behavioral Sciences Division, VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry and Biomedical Genetics, Boston University School of Medicine, Boston, MA, USA
| | - Myriam Fornage
- University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Aniket Mishra
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, F-33000, Bordeaux, France
| | - Shahzad Ahmad
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Alexa S Beiser
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Eric Boerwinkle
- University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Vincent Bouteloup
- Centre Inserm U1219 Bordeaux Population Health, CIC1401-EC, Institut de Santé Publique, d'Epidémiologie et de Développement, Université de Bordeaux, CHU de Bordeaux, Pôle Santé Publique, Bordeaux, France
| | - Vincent Chouraki
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE- LabEx DISTALZ - Risk factors and molecular determinants of aging diseases, F-59000, Lille, France
| | - L Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, USA
| | - Vincent Damotte
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE- LabEx DISTALZ - Risk factors and molecular determinants of aging diseases, F-59000, Lille, France
| | - Charles S DeCarli
- Department of Neurology and Center for Neuroscience, University of California at Davis, Davis, CA, USA
| | - Anita L DeStefano
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Luc Djoussé
- Department of Medicine, Division of Aging, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alison E Fohner
- Institute of Public Health Genetics and Department of Epidemiology and Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
| | - Carol E Franz
- Department of Psychiatry and Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, USA
| | - Tiffany F Kautz
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Jean-Charles Lambert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE- LabEx DISTALZ - Risk factors and molecular determinants of aging diseases, F-59000, Lille, France
| | - Michael J Lyons
- Department of Psychology and Brain Sciences, Boston University, Boston, MA, USA
| | | | - Kenneth J Mukamal
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Matthew P Pase
- Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | | | - Robert A Rissman
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Claudia L Satizabal
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Ramachandran S Vasan
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, USA
- Preventive Medicine & Epidemiology, Boston University School of Medicine, Boston, MA, USA
| | - Amber Yaqub
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Stephanie Debette
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, F-33000, Bordeaux, France
- Bordeaux University Hospital, Department of Neurology, Institute for Neurodegenerative Diseases, Bordeaux, France
| | - Carole Dufouil
- Bordeaux University Hospital, Department of Neurology, Institute for Neurodegenerative Diseases, Bordeaux, France
| | | | - William S Kremen
- Department of Psychiatry and Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, USA
| | - William T Longstreth
- Departments of Neurology and Epidemiology, University of Washington, Seattle, WA, USA
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sudha Seshadri
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| |
Collapse
|
162
|
Caramaschi D, Neumann A, Cardenas A, Tindula G, Alemany S, Zillich L, Pesce G, Lahti JMT, Havdahl A, Mulder R, Felix JF, Tiemeier H, Sirignano L, Frank J, Witt SH, Rietschel M, Deuschle M, Huen K, Eskenazi B, Send TS, Ferrer M, Gilles M, de Agostini M, Baïz N, Rifas-Shiman SL, Kvist T, Czamara D, Tuominen ST, Relton CL, Rai D, London SJ, Räikkönen K, Holland N, Annesi-Maesano I, Streit F, Hivert MF, Oken E, Sunyer J, Cecil CAM, Sharp G. Meta-analysis of epigenome-wide associations between DNA methylation at birth and childhood cognitive skills. Mol Psychiatry 2022; 27:2126-2135. [PMID: 35145228 PMCID: PMC9126809 DOI: 10.1038/s41380-022-01441-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 01/11/2022] [Indexed: 12/21/2022]
Abstract
Cognitive skills are a strong predictor of a wide range of later life outcomes. Genetic and epigenetic associations across the genome explain some of the variation in general cognitive abilities in the general population and it is plausible that epigenetic associations might arise from prenatal environmental exposures and/or genetic variation early in life. We investigated the association between cord blood DNA methylation at birth and cognitive skills assessed in children from eight pregnancy cohorts within the Pregnancy And Childhood Epigenetics (PACE) Consortium across overall (total N = 2196), verbal (total N = 2206) and non-verbal cognitive scores (total N = 3300). The associations at single CpG sites were weak for all of the cognitive domains investigated. One region near DUSP22 on chromosome 6 was associated with non-verbal cognition in a model adjusted for maternal IQ. We conclude that there is little evidence to support the idea that variation in cord blood DNA methylation at single CpG sites is associated with cognitive skills and further studies are needed to confirm the association at DUSP22.
Collapse
Affiliation(s)
- Doretta Caramaschi
- Medical Research Council Integrative Epidemiology Unit (MRC IEU), Bristol Medical School, Population Health Science, University of Bristol, Bristol, UK.
- Department of Psychology, College of Life and Environmental Sciences, University of Exeter, Exeter, UK.
| | - Alexander Neumann
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
| | - Andres Cardenas
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Gwen Tindula
- Children's Environmental Health Laboratory, Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, USA
| | - Silvia Alemany
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Lea Zillich
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Giancarlo Pesce
- Epidemiology of Allergic and Respiratory Diseases Team (EPAR), Institute Pierre Louis of Epidemiology and Public Health, UMR-S 1136 INSERM and Sorbonne Université, Paris, France
| | - Jari M T Lahti
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Alexandra Havdahl
- Medical Research Council Integrative Epidemiology Unit (MRC IEU), Bristol Medical School, Population Health Science, University of Bristol, Bristol, UK
- Department of Mental Disorders, Norwegian Institute of Public Health and Nic Waals Institute of Lovisenberg Diaconal Hospital, Oslo, Norway
| | - Rosa Mulder
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Janine F Felix
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Henning Tiemeier
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Lea Sirignano
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Josef Frank
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Stephanie H Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Michael Deuschle
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Karen Huen
- Children's Environmental Health Laboratory, Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, USA
| | - Brenda Eskenazi
- Center for Environmental Research and Children's Health (CERCH), School of Public Health, University of California, Berkeley, CA, USA
| | - Tabea Sarah Send
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Muriel Ferrer
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain
| | - Maria Gilles
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Maria de Agostini
- Inserm, Centre for Research in Epidemiology and StatisticS (CRESS), Research Team on Early Life Origins of Health (EAROH), Villejuif, France
| | - Nour Baïz
- Epidemiology of Allergic and Respiratory Diseases Team (EPAR), Institute Pierre Louis of Epidemiology and Public Health, UMR-S 1136 INSERM and Sorbonne Université, Paris, France
| | - Sheryl L Rifas-Shiman
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Tuomas Kvist
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Darina Czamara
- Department of Translational Research in Psychiatry, Max-Planck-Institute of Psychiatry, Munich, Germany
| | - Samuli T Tuominen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Caroline L Relton
- Medical Research Council Integrative Epidemiology Unit (MRC IEU), Bristol Medical School, Population Health Science, University of Bristol, Bristol, UK
| | - Dheeraj Rai
- Medical Research Council Integrative Epidemiology Unit (MRC IEU), Bristol Medical School, Population Health Science, University of Bristol, Bristol, UK
| | - Stephanie J London
- National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Katri Räikkönen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Nina Holland
- Children's Environmental Health Laboratory, Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, USA
| | - Isabella Annesi-Maesano
- Epidemiology of Allergic and Respiratory Diseases Team (EPAR), Institute Pierre Louis of Epidemiology and Public Health, UMR-S 1136 INSERM and Sorbonne Université, Paris, France
| | - Fabian Streit
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Marie-France Hivert
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Emily Oken
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Jordi Sunyer
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Charlotte A M Cecil
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Gemma Sharp
- Medical Research Council Integrative Epidemiology Unit (MRC IEU), Bristol Medical School, Population Health Science, University of Bristol, Bristol, UK
| |
Collapse
|
163
|
Greenbaum J, Su KJ, Zhang X, Liu Y, Liu A, Zhao LJ, Luo Z, Tian Q, Shen H, Deng HW. A multiethnic whole genome sequencing study to identify novel loci for bone mineral density. Hum Mol Genet 2022; 31:1067-1081. [PMID: 34673960 PMCID: PMC8976433 DOI: 10.1093/hmg/ddab305] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 11/13/2022] Open
Abstract
At present, there have only been a few DNA sequencing-based studies to explore the genetic determinants of bone mineral density (BMD). We carried out the largest whole genome sequencing analysis to date for femoral neck and spine BMD (n = 4981), with one of the highest average sequencing depths implemented thus far at 22×, in a multiethnic sample (58% Caucasian and 42% African American) from the Louisiana Osteoporosis Study (LOS). The LOS samples were combined with summary statistics from the GEFOS consortium and several independent samples of various ethnicities to perform GWAS meta-analysis (n = 44 506). We identified 31 and 30 genomic risk loci for femoral neck and spine BMD, respectively. The findings substantiate many previously reported susceptibility loci (e.g. WNT16 and ESR1) and reveal several others that are either novel or have not been widely replicated in GWAS for BMD, including two for femoral neck (IGF2 and ZNF423) and one for spine (SIPA1). Although we were not able to uncover ethnicity specific differences in the genetic determinants of BMD, we did identify several loci which demonstrated sex-specific associations, including two for women (PDE4D and PIGN) and three for men (TRAF3IP2, NFIB and LYSMD4). Gene-based rare variant association testing detected MAML2, a regulator of the Notch signaling pathway, which has not previously been suggested, for association with spine BMD. The findings provide novel insights into the pathophysiological mechanisms of osteoporosis.
Collapse
Affiliation(s)
- Jonathan Greenbaum
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Kuan-Jui Su
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Xiao Zhang
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Yong Liu
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA 70112, USA
- School of Basic Medical Science, Central South University, Changsha 410013, Hunan Province, PR China
| | - Anqi Liu
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Lan-Juan Zhao
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Zhe Luo
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Qing Tian
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Hui Shen
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Hong-Wen Deng
- Tulane Center of Biomedical Informatics and Genomics, Deming Department of Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
164
|
Zhu Q, Schultz E, Long J, Roh JM, Valice E, Laurent CA, Radimer KH, Yan L, Ergas IJ, Davis W, Ranatunga D, Gandhi S, Kwan ML, Bao PP, Zheng W, Shu XO, Ambrosone C, Yao S, Kushi LH. UACA locus is associated with breast cancer chemoresistance and survival. NPJ Breast Cancer 2022; 8:39. [PMID: 35322040 PMCID: PMC8943134 DOI: 10.1038/s41523-022-00401-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 02/16/2022] [Indexed: 12/13/2022] Open
Abstract
Few germline genetic variants have been robustly linked with breast cancer outcomes. We conducted trans-ethnic meta genome-wide association study (GWAS) of overall survival (OS) in 3973 breast cancer patients from the Pathways Study, one of the largest prospective breast cancer survivor cohorts. A locus spanning the UACA gene, a key regulator of tumor suppressor Par-4, was associated with OS in patients taking Par-4 dependent chemotherapies, including anthracyclines and anti-HER2 therapy, at a genome-wide significance level (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$P = 1.27 \times 10^{ - 9}$$\end{document}P=1.27×10−9). This association was confirmed in meta-analysis across four independent prospective breast cancer cohorts (combined hazard ratio = 1.84, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$P = 1.28 \times 10^{ - 11}$$\end{document}P=1.28×10−11). Transcriptome-wide association study revealed higher UACA gene expression was significantly associated with worse OS (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$P = 4.68 \times 10^{ - 7}$$\end{document}P=4.68×10−7). Our study identified the UACA locus as a genetic predictor of patient outcome following treatment with anthracyclines and/or anti-HER2 therapy, which may have clinical utility in formulating appropriate treatment strategies for breast cancer patients based on their genetic makeup.
Collapse
Affiliation(s)
- Qianqian Zhu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| | - Emily Schultz
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jirong Long
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Janise M Roh
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Emily Valice
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Cecile A Laurent
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Kelly H Radimer
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Li Yan
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Isaac J Ergas
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Warren Davis
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Dilrini Ranatunga
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Shipra Gandhi
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Marilyn L Kwan
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Ping-Ping Bao
- Shanghai Municipal Center for Disease Prevention and Control, Shanghai, China
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christine Ambrosone
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Song Yao
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| | - Lawrence H Kushi
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA.
| |
Collapse
|
165
|
Mortlock S, Corona RI, Kho PF, Pharoah P, Seo JH, Freedman ML, Gayther SA, Siedhoff MT, Rogers PA, Leuchter R, Walsh CS, Cass I, Karlan BY, Rimel B, Ovarian Cancer Association Consortium, International Endometriosis Genetics Consortium, Montgomery GW, Lawrenson K, Kar SP. A multi-level investigation of the genetic relationship between endometriosis and ovarian cancer histotypes. Cell Rep Med 2022; 3:100542. [PMID: 35492879 PMCID: PMC9040176 DOI: 10.1016/j.xcrm.2022.100542] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 12/13/2021] [Accepted: 01/29/2022] [Indexed: 11/27/2022]
Abstract
Endometriosis is associated with increased risk of epithelial ovarian cancers (EOCs). Using data from large endometriosis and EOC genome-wide association meta-analyses, we estimate the genetic correlation and evaluate the causal relationship between genetic liability to endometriosis and EOC histotypes, and identify shared susceptibility loci. We estimate a significant genetic correlation (rg) between endometriosis and clear cell (rg = 0.71), endometrioid (rg = 0.48), and high-grade serous (rg = 0.19) ovarian cancer, associations supported by Mendelian randomization analyses. Bivariate meta-analysis identified 28 loci associated with both endometriosis and EOC, including 19 with evidence for a shared underlying association signal. Differences in the shared risk suggest different underlying pathways may contribute to the relationship between endometriosis and the different histotypes. Functional annotation using transcriptomic and epigenomic profiles of relevant tissues/cells highlights several target genes. This comprehensive analysis reveals profound genetic overlap between endometriosis and EOC histotypes with valuable genomic targets for understanding the biological mechanisms linking the diseases.
Collapse
Affiliation(s)
- Sally Mortlock
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Rosario I. Corona
- Women’s Cancer Research Program at Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Pik Fang Kho
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Paul Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, CB1 8RN Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, CB1 8RN Cambridge, UK
| | - Ji-Heui Seo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Matthew L. Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Simon A. Gayther
- Center for Bioinformatics and Functional Genomics, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Matthew T. Siedhoff
- Division of Minimally Invasive Gynecologic Surgery, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Peter A.W. Rogers
- University of Melbourne Department of Obstetrics and Gynaecology, and Gynaecology Research Centre, Royal Women’s Hospital, Parkville, VIC 3052, Australia
| | - Ronald Leuchter
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Christine S. Walsh
- Department of Obstetrics and Gynecology, University of Colorado, Aurora, CO, USA
| | - Ilana Cass
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Beth Y. Karlan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - B.J. Rimel
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ovarian Cancer Association Consortium, International Endometriosis Genetics Consortium
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
- Women’s Cancer Research Program at Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, CB1 8RN Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, CB1 8RN Cambridge, UK
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
- Center for Bioinformatics and Functional Genomics, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Division of Minimally Invasive Gynecologic Surgery, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- University of Melbourne Department of Obstetrics and Gynaecology, and Gynaecology Research Centre, Royal Women’s Hospital, Parkville, VIC 3052, Australia
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Obstetrics and Gynecology, University of Colorado, Aurora, CO, USA
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, BS8 2BN Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, BS8 2BN Bristol, UK
| | - Grant W. Montgomery
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Kate Lawrenson
- Women’s Cancer Research Program at Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Center for Bioinformatics and Functional Genomics, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Siddhartha P. Kar
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, BS8 2BN Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, BS8 2BN Bristol, UK
| |
Collapse
|
166
|
Sun J, Lyu R, Deng L, Li Q, Zhao Y, Zhang Y. SMetABF: A rapid algorithm for Bayesian GWAS meta-analysis with a large number of studies included. PLoS Comput Biol 2022; 18:e1009948. [PMID: 35286307 PMCID: PMC8947622 DOI: 10.1371/journal.pcbi.1009948] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 03/24/2022] [Accepted: 02/21/2022] [Indexed: 12/15/2022] Open
Abstract
Bayesian methods are widely used in the GWAS meta-analysis. But the considerable consumption in both computing time and memory space poses great challenges for large-scale meta-analyses. In this research, we propose an algorithm named SMetABF to rapidly obtain the optimal ABF in the GWAS meta-analysis, where shotgun stochastic search (SSS) is introduced to improve the Bayesian GWAS meta-analysis framework, MetABF. Simulation studies confirm that SMetABF performs well in both speed and accuracy, compared to exhaustive methods and MCMC. SMetABF is applied to real GWAS datasets to find several essential loci related to Parkinson's disease (PD) and the results support the underlying relationship between PD and other autoimmune disorders. Developed as an R package and a web tool, SMetABF will become a useful tool to integrate different studies and identify more variants associated with complex traits.
Collapse
Affiliation(s)
- Jianle Sun
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ruiqi Lyu
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Luojia Deng
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Qianwen Li
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yang Zhao
- Department of Biostatistics, Nanjing Medical University School of Public Health, Nanjing, Jiangsu, China
- * E-mail: (YAZ); (YUZ)
| | - Yue Zhang
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- * E-mail: (YAZ); (YUZ)
| |
Collapse
|
167
|
Cao H, Zhou X, Chen Y, Ip FC, Chen Y, Lai NC, Lo RM, Tong EP, Mok VC, Kwok TC, Fu AK, Ip NY. Association of SPI1 Haplotypes with Altered SPI1 Gene Expression and Alzheimer’s Disease Risk. J Alzheimers Dis 2022; 86:1861-1873. [PMID: 35253752 PMCID: PMC9108557 DOI: 10.3233/jad-215311] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background: Genetic studies reveal that single-nucleotide polymorphisms (SNPs) of SPI1 are associated with Alzheimer’s disease (AD), while their effects in the Chinese population remain unclear. Objective: We aimed to examine the AD-association of SPI1 SNPs in the Chinese population and investigate the underlying mechanisms of these SNPs in modulating AD risk. Methods: We conducted a genetic analysis of three SPI1 SNPs (i.e., rs1057233, rs3740688, and rs78245530) in a Chinese cohort (n = 333 patients with AD, n = 721 normal controls). We also probed public European-descent AD cohorts and gene expression datasets to investigate the putative functions of those SNPs. Results: We showed that SPI1 SNP rs3740688 is significantly associated with AD in the Chinese population (odds ratio [OR] = 0.72 [0.58–0.89]) and identified AD-protective SPI1 haplotypes β (tagged by rs1057233 and rs3740688) and γ (tagged by rs3740688 and rs78245530). Specifically, haplotypes β and γ are associated with decreased SPI1 gene expression level in the blood and brain tissues, respectively. The regulatory roles of these haplotypes are potentially mediated by changes in miRNA binding and the epigenetic landscape. Our results suggest that the AD-protective SPI1 haplotypes regulate pathways involved in immune and neuronal functions. Conclusion: This study is the first to report a significant association of SPI1 with AD in the Chinese population. It also identifies SPI1 haplotypes that are associated with SPI1 gene expression and decreased AD risk.
Collapse
Affiliation(s)
- Han Cao
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
| | - Xiaopu Zhou
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen, Guangdong, China
| | - Yu Chen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen, Guangdong, China
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; Shenzhen–Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong, China
| | - Fanny C.F. Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen, Guangdong, China
| | - Yuewen Chen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen, Guangdong, China
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Translational Research for Brain Diseases, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; Shenzhen–Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong, China
| | - Nicole C.H. Lai
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Ronnie M.N. Lo
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Estella P.S. Tong
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Vincent C.T. Mok
- Gerald Choa Neuroscience Centre, Lui Che Woo Institute of Innovative Medicine, Therese Pei Fong Chow Research Centre for Prevention of Dementia, Division of Neurology, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Timothy C.Y. Kwok
- Therese Pei Fong Chow Research Centre for Prevention of Dementia, Division of Geriatrics, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Amy K.Y. Fu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen, Guangdong, China
| | - Nancy Y. Ip
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen–Hong Kong Institute of Brain Science, Shenzhen, Guangdong, China
| | | |
Collapse
|
168
|
Sikdar S. Robust meta-analysis for large-scale genomic experiments based on an empirical approach. BMC Med Res Methodol 2022; 22:43. [PMID: 35144554 PMCID: PMC8832678 DOI: 10.1186/s12874-022-01530-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 01/18/2022] [Indexed: 11/19/2022] Open
Abstract
Background Recent high-throughput technologies have opened avenues for simultaneous analyses of thousands of genes. With the availability of a multitude of public databases, one can easily access multiple genomic study results where each study comprises of significance testing results of thousands of genes. Researchers currently tend to combine this genomic information from these multiple studies in the form of a meta-analysis. As the number of genes involved is very large, the classical meta-analysis approaches need to be updated to acknowledge this large-scale aspect of the data. Methods In this article, we discuss how application of standard theoretical null distributional assumptions of the classical meta-analysis methods, such as Fisher’s p-value combination and Stouffer’s Z, can lead to incorrect significant testing results, and we propose a robust meta-analysis method that empirically modifies the individual test statistics and p-values before combining them. Results Our proposed meta-analysis method performs best in significance testing among several meta-analysis approaches, especially in presence of hidden confounders, as shown through a wide variety of simulation studies and real genomic data analysis. Conclusion The proposed meta-analysis method produces superior meta-analysis results compared to the standard p-value combination approaches for large-scale simultaneous testing in genomic experiments. This is particularly useful in studies with large number of genes where the standard meta-analysis approaches can result in gross false discoveries due to the presence of unobserved confounding variables. Supplementary Information The online version contains supplementary material available at 10.1186/s12874-022-01530-y.
Collapse
Affiliation(s)
- Sinjini Sikdar
- Department of Mathematics and Statistics, Old Dominion University, Norfolk, VA, USA.
| |
Collapse
|
169
|
Dang R, Qu B, Guo K, Zhou S, Sun H, Wang W, Han J, Feng K, Lin J, Hu Y. Weighted Co-Expression Network Analysis Identifies RNF181 as a Causal Gene of Coronary Artery Disease. Front Genet 2022; 12:818813. [PMID: 35222523 PMCID: PMC8867041 DOI: 10.3389/fgene.2021.818813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 12/24/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Coronary artery disease (CAD) exerts a global challenge to public health. Genetic heritability is one of the most vital contributing factors in the pathophysiology of CAD. Co-expression network analysis is an applicable and robust method for the interpretation of biological interaction from microarray data. Previous CAD studies have focused on peripheral blood samples since the processes of CAD may vary from tissue to blood. It is therefore necessary to find biomarkers for CAD in heart tissues; their association also requires further illustration. Materials and Methods: To filter for causal genes, an analysis of microarray expression profiles, GSE12504 and GSE22253, was performed with weighted gene co-expression network analysis (WGCNA). Co-expression modules were constructed after batch effect removal and data normalization. The results showed that 7 co-expression modules with 8,525 genes and 1,210 differentially expressed genes (DEGs) were identified. Furthermore, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted. Four major pathways in CAD tissue and hub genes were addressed in the Hybrid Mouse Diversity Panel (HMDP) and Human Protein Atlas (HPA), and isoproterenol (ISO)/doxycycline (DOX)-induced heart toxicity models were used to validate the hub genes. Lastly, the hub genes and risk variants were verified in the CAD cohort and in genome-wide association studies (GWAS). Results: The results showed that RNF181 and eight other hub genes are perturbed during CAD in heart tissues. Additionally, the expression of RNF181 was validated using RT-PCR and immunohistochemistry (IHC) staining in two cardiotoxicity mouse models. The association was further verified in the CAD patient cohort and in GWAS. Conclusion: Our findings illustrated for the first time that the E3 ubiquitination ligase protein RNF181 may serve as a potential biomarker in CAD, but further in vivo validation is warranted.
Collapse
Affiliation(s)
- Ruoyu Dang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Bojian Qu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
- Pharmaceutical Intelligence Platform, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Kaimin Guo
- GeneNet Pharmaceuticals Co. Ltd., Tianjin, China
| | - Shuiping Zhou
- The State Key Laboratory of Core Technology in Innovative Chinese Medicine, Tasly Academy, Tasly Holding Group Co., Ltd, Tianjin, China
| | - He Sun
- GeneNet Pharmaceuticals Co. Ltd., Tianjin, China
| | - Wenjia Wang
- GeneNet Pharmaceuticals Co. Ltd., Tianjin, China
| | - Jihong Han
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Ke Feng
- College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China
| | - Jianping Lin
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
- Pharmaceutical Intelligence Platform, Tianjin International Joint Academy of Biomedicine, Tianjin, China
- *Correspondence: Jianping Lin, ; Yunhui Hu,
| | - Yunhui Hu
- GeneNet Pharmaceuticals Co. Ltd., Tianjin, China
- *Correspondence: Jianping Lin, ; Yunhui Hu,
| |
Collapse
|
170
|
Wang F, Panjwani N, Wang C, Sun L, Strug LJ. A flexible summary statistics-based colocalization method with application to the mucin cystic fibrosis lung disease modifier locus. Am J Hum Genet 2022; 109:253-269. [PMID: 35065708 PMCID: PMC8874229 DOI: 10.1016/j.ajhg.2021.12.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 12/15/2021] [Indexed: 12/18/2022] Open
Abstract
Mucus obstruction is a central feature in the cystic fibrosis (CF) airways. A genome-wide association study (GWAS) of lung disease by the CF Gene Modifier Consortium (CFGMC) identified a significant locus containing two mucin genes, MUC20 and MUC4. Expression quantitative trait locus (eQTL) analysis using human nasal epithelia (HNE) from 94 CF-affected Canadians in the CFGMC demonstrated MUC4 eQTLs that mirrored the lung association pattern in the region, suggesting that MUC4 expression may mediate CF lung disease. Complications arose, however, with colocalization testing using existing methods: the locus is complex and the associated SNPs span a 0.2 Mb region with high linkage disequilibrium (LD) and evidence of allelic heterogeneity. We previously developed the Simple Sum (SS), a powerful colocalization test in regions with allelic heterogeneity, but SS assumed eQTLs to be present to achieve type I error control. Here we propose a two-stage SS (SS2) colocalization test that avoids a priori eQTL assumptions, accounts for multiple hypothesis testing and the composite null hypothesis, and enables meta-analysis. We compare SS2 to published approaches through simulation and demonstrate type I error control for all settings with the greatest power in the presence of high LD and allelic heterogeneity. Applying SS2 to the MUC20/MUC4 CF lung disease locus with eQTLs from CF HNE revealed significant colocalization with MUC4 (p = 1.31 × 10-5) rather than with MUC20. The SS2 is a powerful method to inform the responsible gene(s) at a locus and guide future functional studies. SS2 has been implemented in the application LocusFocus.
Collapse
Affiliation(s)
- Fan Wang
- Department of Statistical Sciences, University of Toronto, Toronto, ON M5G 1Z5, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Naim Panjwani
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Cheng Wang
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Lei Sun
- Department of Statistical Sciences, University of Toronto, Toronto, ON M5G 1Z5, Canada; Biostatistics Division, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada.
| | - Lisa J Strug
- Department of Statistical Sciences, University of Toronto, Toronto, ON M5G 1Z5, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Biostatistics Division, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada; Department of Computer Science, University of Toronto, Toronto, ON M5S 2E4, Canada; The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.
| |
Collapse
|
171
|
Zeng B, Bendl J, Kosoy R, Fullard JF, Hoffman GE, Roussos P. Multi-ancestry eQTL meta-analysis of human brain identifies candidate causal variants for brain-related traits. Nat Genet 2022; 54:161-169. [PMID: 35058635 PMCID: PMC8852232 DOI: 10.1038/s41588-021-00987-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 11/17/2021] [Indexed: 12/11/2022]
Abstract
While large-scale, genome-wide association studies (GWAS) have identified hundreds of loci associated with brain-related traits, identification of the variants, genes and molecular mechanisms underlying these traits remains challenging. Integration of GWAS with expression quantitative trait loci (eQTLs) and identification of shared genetic architecture have been widely adopted to nominate genes and candidate causal variants. However, this approach is limited by sample size, statistical power and linkage disequilibrium. We developed the multivariate multiple QTL approach and performed a large-scale, multi-ancestry eQTL meta-analysis to increase power and fine-mapping resolution. Analysis of 3,983 RNA-sequenced samples from 2,119 donors, including 474 non-European individuals, yielded an effective sample size of 3,154. Joint statistical fine-mapping of eQTL and GWAS identified 329 variant-trait pairs for 24 brain-related traits driven by 204 unique candidate causal variants for 189 unique genes. This integrative analysis identifies candidate causal variants and elucidates potential regulatory mechanisms for genes underlying schizophrenia, bipolar disorder and Alzheimer's disease.
Collapse
Affiliation(s)
- Biao Zeng
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jaroslav Bendl
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roman Kosoy
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John F Fullard
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gabriel E Hoffman
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Panos Roussos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mental Illness Research, Education and Clinical Centers, James J. Peters VA Medical Center, Bronx, NY, USA.
- Center for Dementia Research, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, USA.
| |
Collapse
|
172
|
Adewuyi EO, Mehta D. Genetic overlap analysis of endometriosis and asthma identifies shared loci implicating sex hormones and thyroid signalling pathways. Hum Reprod 2022; 37:366-383. [PMID: 35472084 PMCID: PMC8804329 DOI: 10.1093/humrep/deab254] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/13/2021] [Indexed: 08/27/2023] Open
Abstract
STUDY QUESTION Is there a shared genetic or causal association of endometriosis with asthma or what biological mechanisms may underlie their potential relationships? SUMMARY ANSWER Our results confirm a significant but non-causal association of endometriosis with asthma implicating shared genetic susceptibility and biological pathways in the mechanisms of the disorders, and potentially, their co-occurrence. WHAT IS KNOWN ALREADY Some observational studies have reported a pattern of co-occurring relationship between endometriosis and asthma; however, there is conflicting evidence and the aetiology, as well as the underlying mechanisms of the relationship, remain unclear. STUDY DESIGN, SIZE, DURATION We applied multiple statistical genetic approaches in the analysis of well-powered, genome-wide association study (GWAS) summary data to comprehensively assess the relationship of endometriosis with asthma. Endometriosis GWAS from the International Endogene Consortium (IEC, 17 054 cases and 191 858 controls) and asthma GWAS from the United Kingdom Biobank (UKB, 26 332 cases and 375 505 controls) were analysed. Additional asthma data from the Trans-National Asthma Genetic Consortium (TAGC, 19 954 cases and 107 715 controls) were utilized for replication testing. PARTICIPANTS/MATERIALS, SETTING, METHODS We assessed single-nucleotide polymorphism (SNP)-level genetic overlap and correlation between endometriosis and asthma using SNP effect concordance analysis (SECA) and linkage disequilibrium score regression analysis (LDSC) methods, respectively. GWAS meta-analysis, colocalization (GWAS-PW), gene-based and pathway-based functional enrichment analysis methods were applied, respectively, to identify SNP loci, genomic regions, genes and biological pathways shared by endometriosis and asthma. Potential causal associations between endometriosis and asthma were assessed using Mendelian randomization (MR) methods. MAIN RESULTS AND THE ROLE OF CHANCE SECA revealed significant concordance of SNP risk effects across the IEC endometriosis and the UKB asthma GWAS. Also, LDSC analysis found a positive and significant genetic correlation (rG = 0.16, P = 2.01 × 10-6) between the two traits. GWAS meta-analysis of the IEC endometriosis and UKB asthma GWAS identified 14 genome-wide significant (Pmeta-analysis < 5.0 × 10-8) independent loci, five of which are putatively novel. Three of these loci were consistently replicated using TAGC asthma GWAS and reinforced in colocalization and gene-based analyses. Additional shared genomic regions were identified in the colocalization analysis. MR found no evidence of a significant causal association between endometriosis and asthma. However, combining gene-based association results across the GWAS for endometriosis and asthma, we identified 17 shared genes with a genome-wide significant Fisher's combined P-value (FCPgene) <2.73 × 10-6. Additional analyses (independent gene-based analysis) replicated evidence of gene-level genetic overlap between endometriosis and asthma. Biological mechanisms including 'thyroid hormone signalling', 'abnormality of immune system physiology', 'androgen biosynthetic process' and 'brain-derived neurotrophic factor signalling pathway', among others, were significantly enriched for endometriosis and asthma in a pathway-based analysis. LARGE SCALE DATA The GWAS for endometriosis data were sourced from the International Endogen Consortium (IEC) and can be accessed by contacting the consortium. The GWAS data for asthma are freely available online at Lee Lab (https://www.leelabsg.org/resources) and from the Trans-National Asthma Genetic Consortium (TAGC). LIMITATIONS, REASONS FOR CAUTION Given we analysed GWAS datasets from mainly European populations, our results may not be generalizable to other ancestries. WIDER IMPLICATIONS OF THE FINDINGS This study provides novel insights into mechanisms underpinning endometriosis and asthma, and potentially their observed relationship. Findings support a co-occurring relationship of endometriosis with asthma largely due to shared genetic components. Agents targeting 'selective androgen receptor modulators' may be therapeutically relevant in both disorders. Moreover, SNPs, loci, genes and biological pathways identified in our study provide potential targets for further investigation in endometriosis and asthma. STUDY FUNDING/COMPETING INTEREST(S) National Health and Medical Research Council (NHMRC) of Australia (241,944, 339,462, 389,927, 389,875, 389,891, 389,892, 389,938, 443,036, 442,915, 442,981, 496,610, 496,739, 552,485, 552,498, 1,026,033 and 1,050,208), Wellcome Trust (awards 076113 and 085475) and the Lundbeck Foundation (R102-A9118 and R155-2014-1724). All researchers had full independence from the funders. Authors do not have any conflict of interest.
Collapse
Affiliation(s)
- E O Adewuyi
- Queensland University of Technology, Faculty of Health, School of Biomedical Sciences, Centre for Genomics and Personalised Health, Brisbane, Queensland, Australia
- Centre for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Perth, Australia
| | - D Mehta
- Queensland University of Technology, Faculty of Health, School of Biomedical Sciences, Centre for Genomics and Personalised Health, Brisbane, Queensland, Australia
| |
Collapse
|
173
|
Herrera-Luis E, Li A, Mak ACY, Perez-Garcia J, Elhawary JR, Oh SS, Hu D, Eng C, Keys KL, Huntsman S, Beckman KB, Borrell LN, Rodriguez-Santana J, Burchard EG, Pino-Yanes M. Epigenome-wide association study of lung function in Latino children and youth with asthma. Clin Epigenetics 2022; 14:9. [PMID: 35033200 PMCID: PMC8760660 DOI: 10.1186/s13148-022-01227-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 01/03/2022] [Indexed: 11/10/2022] Open
Abstract
INTRODUCTION DNA methylation studies have associated methylation levels at different CpG sites or genomic regions with lung function. Moreover, genetic ancestry has been associated with lung function in Latinos. However, no epigenome-wide association study (EWAS) of lung function has been performed in this population. Here, we aimed to identify DNA methylation patterns associated with lung function in pediatric asthma among Latinos. RESULTS We conducted an EWAS in whole blood from 250 Puerto Rican and 148 Mexican American children and young adults with asthma. A total of five CpGs exceeded the genome-wide significance threshold of p = 1.17 × 10-7 in the combined analyses from Puerto Ricans and Mexican Americans: cg06035600 (MAP3K6, p = 6.13 × 10-8) showed significant association with pre-bronchodilator Tiffeneau-Pinelli index, the probes cg00914963 (TBC1D16, p = 1.04 × 10-7), cg16405908 (MRGPRE, p = 2.05 × 10-8), and cg07428101 (MUC2, p = 5.02 × 10-9) were associated with post-bronchodilator forced vital capacity (FVC), and cg20515679 (KCNJ6) with post-bronchodilator Tiffeneau-Pinelli index (p = 1.13 × 10-8). However, these markers did not show significant associations in publicly available data from Europeans (p > 0.05). A methylation quantitative trait loci analysis revealed that methylation levels at these CpG sites were regulated by genetic variation in Latinos and the Biobank-based Integrative Omics Studies (BIOS) consortium. Additionally, two differentially methylated regions in REXOC and AURKC were associated with pre-bronchodilator Tiffeneau-Pinelli index (adjusted p < 0.05) in Puerto Ricans and Mexican Americans. Moreover, we replicated some of the previous differentially methylated signals associated with lung function in non-Latino populations. CONCLUSIONS We replicated previous associations of epigenetic markers with lung function in whole blood and identified novel population-specific associations shared among Latino subgroups.
Collapse
Affiliation(s)
- Esther Herrera-Luis
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, Apartado 456, 38200, San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - Annie Li
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Angel C Y Mak
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Javier Perez-Garcia
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, Apartado 456, 38200, San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - Jennifer R Elhawary
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Sam S Oh
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Donglei Hu
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Celeste Eng
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Kevin L Keys
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Berkeley Institute for Data Science, University of California Berkeley, Berkeley, CA, USA
| | - Scott Huntsman
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | | | - Luisa N Borrell
- Department of Epidemiology and Biostatistics, Graduate School of Public Health and Health Policy, City University of New York, New York, NY, USA
| | | | - Esteban G Burchard
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Maria Pino-Yanes
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, Apartado 456, 38200, San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain.
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.
- Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, San Cristóbal de La Laguna, Tenerife, Spain.
| |
Collapse
|
174
|
Li J, Glover JD, Zhang H, Peng M, Tan J, Mallick CB, Hou D, Yang Y, Wu S, Liu Y, Peng Q, Zheng SC, Crosse EI, Medvinsky A, Anderson RA, Brown H, Yuan Z, Zhou S, Xu Y, Kemp JP, Ho YYW, Loesch DZ, Wang L, Li Y, Tang S, Wu X, Walters RG, Lin K, Meng R, Lv J, Chernus JM, Neiswanger K, Feingold E, Evans DM, Medland SE, Martin NG, Weinberg SM, Marazita ML, Chen G, Chen Z, Zhou Y, Cheeseman M, Wang L, Jin L, Headon DJ, Wang S. Limb development genes underlie variation in human fingerprint patterns. Cell 2022; 185:95-112.e18. [PMID: 34995520 PMCID: PMC8740935 DOI: 10.1016/j.cell.2021.12.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 10/20/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022]
Abstract
Fingerprints are of long-standing practical and cultural interest, but little is known about the mechanisms that underlie their variation. Using genome-wide scans in Han Chinese cohorts, we identified 18 loci associated with fingerprint type across the digits, including a genetic basis for the long-recognized "pattern-block" correlations among the middle three digits. In particular, we identified a variant near EVI1 that alters regulatory activity and established a role for EVI1 in dermatoglyph patterning in mice. Dynamic EVI1 expression during human development supports its role in shaping the limbs and digits, rather than influencing skin patterning directly. Trans-ethnic meta-analysis identified 43 fingerprint-associated loci, with nearby genes being strongly enriched for general limb development pathways. We also found that fingerprint patterns were genetically correlated with hand proportions. Taken together, these findings support the key role of limb development genes in influencing the outcome of fingerprint patterning.
Collapse
Affiliation(s)
- Jinxi Li
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai 200438, PRC; CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC
| | - James D Glover
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Haiguo Zhang
- Ministry of Education Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai 200438, PRC
| | - Meifang Peng
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC; Ministry of Education Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai 200438, PRC
| | - Jingze Tan
- Ministry of Education Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai 200438, PRC
| | - Chandana Basu Mallick
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK; Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Dan Hou
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC
| | - Yajun Yang
- Ministry of Education Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai 200438, PRC
| | - Sijie Wu
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai 200438, PRC; CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC
| | - Yu Liu
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC
| | - Qianqian Peng
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC
| | - Shijie C Zheng
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC
| | - Edie I Crosse
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | | | - Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Helen Brown
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Ziyu Yuan
- Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu 225326, PRC
| | - Shen Zhou
- Shanghai Foreign Language School, Shanghai 200083, PRC
| | - Yanqing Xu
- Forest Ridge School of the Sacred Heart, Bellevue, WA 98006, USA
| | - John P Kemp
- University of Queensland Diamantina Institute, University of Queensland, Brisbane, QLD, Australia
| | - Yvonne Y W Ho
- QIMR Berghofer Medical Rese Institute, Brisbane, QLD, Australia
| | - Danuta Z Loesch
- Psychology Department, La Trobe University, Melbourne, VIC, Australia
| | | | | | | | - Xiaoli Wu
- WeGene, Shenzhen, Guangdong 518040, PRC
| | - Robin G Walters
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Medical Research Council Population Health Research Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Kuang Lin
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Ruogu Meng
- Center for Data Science in Health and Medicine, Peking University, Beijing 100191, PRC
| | - Jun Lv
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, PRC
| | - Jonathan M Chernus
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Katherine Neiswanger
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Eleanor Feingold
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - David M Evans
- University of Queensland Diamantina Institute, University of Queensland, Brisbane, QLD, Australia; Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia; MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Sarah E Medland
- QIMR Berghofer Medical Rese Institute, Brisbane, QLD, Australia
| | | | - Seth M Weinberg
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15261, USA; Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Anthropology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Mary L Marazita
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15261, USA; Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA 15219, USA; Clinical and Translational Science, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Gang Chen
- WeGene, Shenzhen, Guangdong 518040, PRC
| | - Zhengming Chen
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Medical Research Council Population Health Research Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Yong Zhou
- Clinical Research Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PRC
| | - Michael Cheeseman
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Lan Wang
- Chinese Academy of Sciences Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, and Human Phenome Institute, Fudan University, Shanghai 200438, PRC; CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC; Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases (2019RU058), Chinese Academy of Medical Sciences, Shanghai 200438, PRC.
| | - Denis J Headon
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK.
| | - Sijia Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, PRC; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, PRC.
| |
Collapse
|
175
|
Albert E, Sauvage C. Identification and Validation of Candidate Genes from Genome-Wide Association Studies. Methods Mol Biol 2022; 2481:249-272. [PMID: 35641769 DOI: 10.1007/978-1-0716-2237-7_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Exploiting the statistical associations coming out from a GWAS experiment to identify and validate candidate genes may be potentially difficult and time consuming. To fill the gap between the identification of candidate genes toward their functional validation onto the trait performance, the prioritization of variants underlying the GWAS-associated regions is necessary. In parallel, recent developments in genomics and statistical methods have been achieved notably in human genetic and they are accordingly being adopted in plant breeding toward the study of the genetic architecture of traits to sustain genetic gains. In this chapter, we aim at providing both theoretical and practical aspects underlying three main options including (1) the MetaGWAS analysis, (2) the statistical fine mapping and (3) the integration of functional data toward the identification and validation of candidate genes from a GWAS experiment.
Collapse
|
176
|
Longchamps RJ, Yang SY, Castellani CA, Shi W, Lane J, Grove ML, Bartz TM, Sarnowski C, Liu C, Burrows K, Guyatt AL, Gaunt TR, Kacprowski T, Yang J, De Jager PL, Yu L, Bergman A, Xia R, Fornage M, Feitosa MF, Wojczynski MK, Kraja AT, Province MA, Amin N, Rivadeneira F, Tiemeier H, Uitterlinden AG, Broer L, Van Meurs JBJ, Van Duijn CM, Raffield LM, Lange L, Rich SS, Lemaitre RN, Goodarzi MO, Sitlani CM, Mak ACY, Bennett DA, Rodriguez S, Murabito JM, Lunetta KL, Sotoodehnia N, Atzmon G, Ye K, Barzilai N, Brody JA, Psaty BM, Taylor KD, Rotter JI, Boerwinkle E, Pankratz N, Arking DE. Genome-wide analysis of mitochondrial DNA copy number reveals loci implicated in nucleotide metabolism, platelet activation, and megakaryocyte proliferation. Hum Genet 2022; 141:127-146. [PMID: 34859289 PMCID: PMC8758627 DOI: 10.1007/s00439-021-02394-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 10/22/2021] [Indexed: 12/18/2022]
Abstract
Mitochondrial DNA copy number (mtDNA-CN) measured from blood specimens is a minimally invasive marker of mitochondrial function that exhibits both inter-individual and intercellular variation. To identify genes involved in regulating mitochondrial function, we performed a genome-wide association study (GWAS) in 465,809 White individuals from the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) consortium and the UK Biobank (UKB). We identified 133 SNPs with statistically significant, independent effects associated with mtDNA-CN across 100 loci. A combination of fine-mapping, variant annotation, and co-localization analyses was used to prioritize genes within each of the 133 independent sites. Putative causal genes were enriched for known mitochondrial DNA depletion syndromes (p = 3.09 × 10-15) and the gene ontology (GO) terms for mtDNA metabolism (p = 1.43 × 10-8) and mtDNA replication (p = 1.2 × 10-7). A clustering approach leveraged pleiotropy between mtDNA-CN associated SNPs and 41 mtDNA-CN associated phenotypes to identify functional domains, revealing three distinct groups, including platelet activation, megakaryocyte proliferation, and mtDNA metabolism. Finally, using mitochondrial SNPs, we establish causal relationships between mitochondrial function and a variety of blood cell-related traits, kidney function, liver function and overall (p = 0.044) and non-cancer mortality (p = 6.56 × 10-4).
Collapse
Affiliation(s)
- R J Longchamps
- Department of Genetic Medicine, McKusick-Nathans Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - S Y Yang
- Department of Genetic Medicine, McKusick-Nathans Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - C A Castellani
- Department of Genetic Medicine, McKusick-Nathans Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - W Shi
- Department of Genetic Medicine, McKusick-Nathans Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - J Lane
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - M L Grove
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, Human Genetics Center, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - T M Bartz
- Cardiovascular Health Research Unit, Departments of Medicine and Biostatistics, University of Washington, Seattle, WA, USA
| | - C Sarnowski
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - C Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - K Burrows
- MRC Integrative Epidemiology Unit at the University of Bristol, University of Bristol, Oakfield House, Oakfield Grove, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, UK
| | - A L Guyatt
- Department of Health Sciences, University of Leicester, University Road, Leicester, UK
| | - T R Gaunt
- MRC Integrative Epidemiology Unit at the University of Bristol, University of Bristol, Oakfield House, Oakfield Grove, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, UK
| | - T Kacprowski
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
- Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics, TU Braunschweig and Hannover Medical School, Brunswick, Germany
| | - J Yang
- Rush Alzheimer's Disease Center and Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - P L De Jager
- Center for Translational and Systems Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - L Yu
- Rush Alzheimer's Disease Center and Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - A Bergman
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - R Xia
- Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - M Fornage
- Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genetics Center, The University of Texas Health Science Center at Houston, Houston, USA
| | - M F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, USA
| | - M K Wojczynski
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, USA
| | - A T Kraja
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, USA
| | - M A Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, USA
| | - N Amin
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - F Rivadeneira
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - H Tiemeier
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Social and Behavioral Science, Harvard T.H. School of Public Health, Boston, USA
| | - A G Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - L Broer
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - J B J Van Meurs
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - C M Van Duijn
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - L M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - L Lange
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - S S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - R N Lemaitre
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - M O Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - C M Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - A C Y Mak
- Cardiovascular Research Institute and Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - D A Bennett
- Rush Alzheimer's Disease Center and Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | - S Rodriguez
- MRC Integrative Epidemiology Unit at the University of Bristol, University of Bristol, Oakfield House, Oakfield Grove, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, UK
| | - J M Murabito
- Boston University School of Medicine, Boston University, Boston, MA, USA
| | - K L Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - N Sotoodehnia
- Cardiovascular Health Research Unit, Division of Cardiology, University of Washington, Seattle, WA, USA
| | - G Atzmon
- Department of Natural Science, University of Haifa, Haifa, Israel
- Departments of Medicine and Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - K Ye
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - N Barzilai
- Departments of Medicine and Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - J A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - B M Psaty
- Cardiovascular Health Research Unit, Departments of Epidemiology, Medicine and Health Services, University of Washington, Seattle, WA, USA
| | - K D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - J I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - E Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, Human Genetics Center, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Baylor College of Medicine, Human Genome Sequencing Center, Houston, TX, USA
| | - N Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - D E Arking
- Department of Genetic Medicine, McKusick-Nathans Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
177
|
Lopes KDP, Snijders GJL, Humphrey J, Allan A, Sneeboer MAM, Navarro E, Schilder BM, Vialle RA, Parks M, Missall R, van Zuiden W, Gigase FAJ, Kübler R, van Berlekom AB, Hicks EM, Bӧttcher C, Priller J, Kahn RS, de Witte LD, Raj T. Genetic analysis of the human microglial transcriptome across brain regions, aging and disease pathologies. Nat Genet 2022; 54:4-17. [PMID: 34992268 PMCID: PMC9245609 DOI: 10.1038/s41588-021-00976-y] [Citation(s) in RCA: 132] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 10/19/2021] [Indexed: 12/12/2022]
Abstract
Microglia have emerged as important players in brain aging and pathology. To understand how genetic risk for neurological and psychiatric disorders is related to microglial function, large transcriptome studies are essential. Here we describe the transcriptome analysis of 255 primary human microglial samples isolated at autopsy from multiple brain regions of 100 individuals. We performed systematic analyses to investigate various aspects of microglial heterogeneities, including brain region and aging. We mapped expression and splicing quantitative trait loci and showed that many neurological disease susceptibility loci are mediated through gene expression or splicing in microglia. Fine-mapping of these loci nominated candidate causal variants that are within microglia-specific enhancers, finding associations with microglial expression of USP6NL for Alzheimer's disease and P2RY12 for Parkinson's disease. We have built the most comprehensive catalog to date of genetic effects on the microglial transcriptome and propose candidate functional variants in neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Katia de Paiva Lopes
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gijsje J L Snijders
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, New York, NY, USA
| | - Jack Humphrey
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amanda Allan
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marjolein A M Sneeboer
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Elisa Navarro
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian M Schilder
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ricardo A Vialle
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Madison Parks
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roy Missall
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Welmoed van Zuiden
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Frederieke A J Gigase
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, New York, NY, USA
| | - Raphael Kübler
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amber Berdenis van Berlekom
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Emily M Hicks
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chotima Bӧttcher
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Josef Priller
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - René S Kahn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, New York, NY, USA
| | - Lot D de Witte
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, New York, NY, USA.
| | - Towfique Raj
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences & Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
178
|
Ferwerda B, Abdellaoui A, Nieuwdorp M, Zwinderman K. A Genetic Map of the Modern Urban Society of Amsterdam. Front Genet 2021; 12:727269. [PMID: 34917125 PMCID: PMC8670378 DOI: 10.3389/fgene.2021.727269] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 11/11/2021] [Indexed: 11/13/2022] Open
Abstract
Genetic differences between individuals underlie susceptibility to many diseases. Genome-wide association studies (GWAS) have discovered many susceptibility genes but were often limited to cohorts of predominantly European ancestry. Genetic diversity between individuals due to different ancestries and evolutionary histories shows that this approach has limitations. In order to gain a better understanding of the associated genetic variation, we need a more global genomics approach including a greater diversity. Here, we introduce the Healthy Life in an Urban Setting (HELIUS) cohort. The HELIUS cohort consists of participants living in Amsterdam, with a level of diversity that reflects the Dutch colonial and recent migration past. The current study includes 10,283 participants with genetic data available from seven groups of inhabitants, namely, Dutch, African Surinamese, South-Asian Surinamese, Turkish, Moroccan, Ghanaian, and Javanese Surinamese. First, we describe the genetic variation and admixture within the HELIUS cohort. Second, we show the challenges during imputation when having a genetically diverse cohort. Third, we conduct a body mass index (BMI) and height GWAS where we investigate the effects of a joint analysis of the entire cohort and a meta-analysis approach for the different subgroups. Finally, we construct polygenic scores for BMI and height and compare their predictive power across the different ethnic groups. Overall, we give a comprehensive overview of a genetically diverse cohort from Amsterdam. Our study emphasizes the importance of a less biased and more realistic representation of urban populations for mapping genetic associations with complex traits and disease risk for all.
Collapse
Affiliation(s)
- Bart Ferwerda
- Department of Clinical Epidemiology and Biostatistics, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Abdel Abdellaoui
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Max Nieuwdorp
- Department of Vascular Medicine, Amsterdam University Medical Center, Amsterdam, Netherlands.,Internal Medicine, Amsterdam University Medical Center, Amsterdam, Netherlands.,Institute for Cardiovascular Research, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Koos Zwinderman
- Department of Clinical Epidemiology and Biostatistics, Amsterdam University Medical Centers, Amsterdam, Netherlands
| |
Collapse
|
179
|
Suarez-Pajes E, Díaz-García C, Rodríguez-Pérez H, Lorenzo-Salazar JM, Marcelino-Rodríguez I, Corrales A, Zheng X, Callero A, Perez-Rodriguez E, Garcia-Robaina JC, González-Montelongo R, Flores C, Guillen-Guio B. Targeted analysis of genomic regions enriched in African ancestry reveals novel classical HLA alleles associated with asthma in Southwestern Europeans. Sci Rep 2021; 11:23686. [PMID: 34880287 PMCID: PMC8654850 DOI: 10.1038/s41598-021-02893-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/24/2021] [Indexed: 12/30/2022] Open
Abstract
Despite asthma has a considerable genetic component, an important proportion of genetic risks remain unknown, especially for non-European populations. Canary Islanders have the largest African genetic ancestry observed among Southwestern Europeans and the highest asthma prevalence in Spain. Here we examined broad chromosomal regions previously associated with an excess of African genetic ancestry in Canary Islanders, with the aim of identifying novel risk variants associated with asthma susceptibility. In a two-stage cases-control study, we revealed a variant within HLA-DQB1 significantly associated with asthma risk (rs1049213, meta-analysis p = 1.30 × 10-7, OR [95% CI] = 1.74 [1.41-2.13]) previously associated with asthma and broad allergic phenotype. Subsequent fine-mapping analyses of classical HLA alleles revealed a novel allele significantly associated with asthma protection (HLA-DQA1*01:02, meta-analysis p = 3.98 × 10-4, OR [95% CI] = 0.64 [0.50-0.82]) that had been linked to infectious and autoimmune diseases, and peanut allergy. HLA haplotype analyses revealed a novel haplotype DQA1*01:02-DQB1*06:04 conferring asthma protection (meta-analysis p = 4.71 × 10-4, OR [95% CI] = 0.47 [0.29- 0.73]).
Collapse
Affiliation(s)
- Eva Suarez-Pajes
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - Claudio Díaz-García
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - Héctor Rodríguez-Pérez
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - Jose M Lorenzo-Salazar
- Genomics Division, Instituto Tecnológico Y de Energías Renovables (ITER), Santa Cruz de Tenerife, Spain
| | - Itahisa Marcelino-Rodríguez
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - Almudena Corrales
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Xiuwen Zheng
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Ariel Callero
- Allergy Unit, Hospital Universitario N.S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Eva Perez-Rodriguez
- Allergy Unit, Hospital Universitario N.S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Jose C Garcia-Robaina
- Allergy Unit, Hospital Universitario N.S. de Candelaria, Santa Cruz de Tenerife, Spain
| | | | - Carlos Flores
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain.
- Genomics Division, Instituto Tecnológico Y de Energías Renovables (ITER), Santa Cruz de Tenerife, Spain.
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.
| | - Beatriz Guillen-Guio
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain.
- Department of Health Sciences, University of Leicester, Leicester, UK.
| |
Collapse
|
180
|
Scott AJ, Chiang C, Hall IM. Structural variants are a major source of gene expression differences in humans and often affect multiple nearby genes. Genome Res 2021; 31:2249-2257. [PMID: 34544830 PMCID: PMC8647827 DOI: 10.1101/gr.275488.121] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 09/14/2021] [Indexed: 11/29/2022]
Abstract
Structural variants (SVs) are an important source of human genome diversity, but their functional effects are poorly understood. We mapped 61,668 SVs in 613 individuals from the GTEx project and measured their effects on gene expression. We estimate that common SVs are causal at 2.66% of eQTLs, a 10.5-fold enrichment relative to their abundance in the genome. Duplications and deletions were the most impactful variant types, whereas the contribution of mobile element insertions was small (0.12% of eQTLs, 1.9-fold enriched). Multitissue analysis of eQTLs revealed that gene-altering SVs show more constitutive effects than other variant types, with 62.09% of coding SV-eQTLs active in all tissues with eQTL activity compared with 23.08% of coding SNV- and indel-eQTLs. Noncoding SVs, SNVs and indels show broadly similar patterns. We also identified 539 rare SVs associated with nearby gene expression outliers. Of these, 62.34% are noncoding SVs that affect gene expression but have modest enrichment at regulatory elements, showing that rare noncoding SVs are a major source of gene expression differences but remain difficult to predict from current annotations. Both common and rare SVs often affect the expression of multiple genes: SV-eQTLs affect an average of 1.82 nearby genes, whereas SNV- and indel-eQTLs affect an average of 1.09 genes, and 21.34% of rare expression-altering SVs show effects on two to nine different genes. We also observe significant effects on rare gene expression changes extending 1 Mb from the SV. This provides a mechanism by which individual SVs may have strong or pleiotropic effects on phenotypic variation.
Collapse
Affiliation(s)
- Alexandra J Scott
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri 63108, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Colby Chiang
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri 63108, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Ira M Hall
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri 63108, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut 06510, USA
| |
Collapse
|
181
|
Makinde FL, Tchamga MSS, Jafali J, Fatumo S, Chimusa ER, Mulder N, Mazandu GK. Reviewing and assessing existing meta-analysis models and tools. Brief Bioinform 2021; 22:bbab324. [PMID: 34415019 PMCID: PMC8575034 DOI: 10.1093/bib/bbab324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 07/07/2021] [Accepted: 07/25/2021] [Indexed: 01/03/2023] Open
Abstract
Over the past few years, meta-analysis has become popular among biomedical researchers for detecting biomarkers across multiple cohort studies with increased predictive power. Combining datasets from different sources increases sample size, thus overcoming the issue related to limited sample size from each individual study and boosting the predictive power. This leads to an increased likelihood of more accurately predicting differentially expressed genes/proteins or significant biomarkers underlying the biological condition of interest. Currently, several meta-analysis methods and tools exist, each having its own strengths and limitations. In this paper, we survey existing meta-analysis methods, and assess the performance of different methods based on results from different datasets as well as assessment from prior knowledge of each method. This provides a reference summary of meta-analysis models and tools, which helps to guide end-users on the choice of appropriate models or tools for given types of datasets and enables developers to consider current advances when planning the development of new meta-analysis models and more practical integrative tools.
Collapse
Affiliation(s)
- Funmilayo L Makinde
- Computational Biology Division at University of Cape Town in collaboration with the African Institute for Mathematical Sciences (AIMS), South Africa
| | - Milaine S S Tchamga
- Division of Human Genetics at University of Cape in collaboration with the African Institute for Mathematical Sciences (AIMS), South Africa
| | - James Jafali
- Pathogen Biology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Malawi
| | - Segun Fatumo
- London School of Hygiene and Tropical Medicine, University of London, UK
| | - Emile R Chimusa
- Division of Human Genetics, Department of Pathology, University of Cape Town, South Africa
| | - Nicola Mulder
- Computational Biology Division at University of Cape Town, South Africa
| | - Gaston K Mazandu
- Division of Human Genetics, Department of Pathology at University of Cape Town, and Associate Researcher at the African Institute for Mathematical Sciences (AIMS), South Africa
| |
Collapse
|
182
|
A large Canadian cohort provides insights into the genetic architecture of human hair colour. Commun Biol 2021; 4:1253. [PMID: 34737440 PMCID: PMC8568909 DOI: 10.1038/s42003-021-02764-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 10/08/2021] [Indexed: 12/05/2022] Open
Abstract
Hair colour is a polygenic phenotype that results from differences in the amount and ratio of melanins located in the hair bulb. Genome-wide association studies (GWAS) have identified many loci involved in the pigmentation pathway affecting hair colour. However, most of the associated loci overlap non-protein coding regions and many of the molecular mechanisms underlying pigmentation variation are still not understood. Here, we conduct GWAS meta-analyses of hair colour in a Canadian cohort of 12,741 individuals of European ancestry. By performing fine-mapping analyses we identify candidate causal variants in pigmentation loci associated with blonde, red and brown hair colour. Additionally, we observe colocalization of several GWAS hits with expression and methylation quantitative trait loci (QTLs) of cultured melanocytes. Finally, transcriptome-wide association studies (TWAS) further nominate the expression of EDNRB and CDK10 as significantly associated with hair colour. Our results provide insights on the mechanisms regulating pigmentation biology in humans.
Collapse
|
183
|
Espuela-Ortiz A, Herrera-Luis E, Lorenzo-Díaz F, Hu D, Eng C, Villar J, Rodriguez-Santana JR, Burchard EG, Pino-Yanes M. Role of Sex on the Genetic Susceptibility to Childhood Asthma in Latinos and African Americans. J Pers Med 2021; 11:1140. [PMID: 34834492 PMCID: PMC8625344 DOI: 10.3390/jpm11111140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/28/2021] [Accepted: 10/30/2021] [Indexed: 01/08/2023] Open
Abstract
Asthma is a respiratory disease whose prevalence changes throughout the lifespan and differs by sex, being more prevalent in males during childhood and in females after puberty. In this study, we assessed the influence of sex on the genetic susceptibility to childhood asthma in admixed populations. Sex-interaction and sex-stratified genome-wide association studies (GWAS) were performed in 4291 Latinos and 1730 African Americans separately, and results were meta-analyzed. Genome-wide (p ≤ 9.35 × 10-8) and suggestive (p ≤ 1.87 × 10-6) population-specific significance thresholds were calculated based on 1000 Genomes Project data. Additionally, protein quantitative trait locus (pQTL) information was gathered for the suggestively associated variants, and enrichment analyses of the proteins identified were carried out. Four independent loci showed interaction with sex at a suggestive level. The stratified GWAS highlighted the 17q12-21 asthma locus as a contributor to asthma susceptibility in both sexes but reached genome-wide significance only in females (p-females < 9.2 × 10-8; p-males < 1.25 × 10-2). Conversely, genetic variants upstream of ligand-dependent nuclear receptor corepressor-like gene (LCORL), previously involved in height determination and spermatogenesis, were associated with asthma only in males (minimum p = 5.31 × 10-8 for rs4593128). Enrichment analyses revealed an overrepresentation of processes related to the immune system and highlighted differences between sexes. In conclusion, we identified sex-specific polymorphisms that could contribute to the differences in the prevalence of childhood asthma between males and females.
Collapse
Grants
- SAF2017-83417R European Regional Development Fund from the European Union
- P60MD006902, R01MD010443, and R56MD013312 NIMHD NIH HHS
- SAF2017-83417R State Research Agency
- M-ULL MICIU/ULL
- Amos Medical Faculty Development Program Robert Wood Johnson Foundation
- R01ES015794 NIEHS NIH HHS
- R21ES24844 NIEHS NIH HHS
- R01HL128439, R01HL135156, R01HL141992, and R01HL141845 National Heart Lung and Blood Institute
- RL5 GM118984 NIGMS NIH HHS
- RYC-2015-17205 Spanish Ministry of Science, Innovation, and Universities
- American Asthma Foundation
- R01HL117004 and X01HL134589 National Heart Lung and Blood Institute
- SAF2017-83417R Spanish Ministry of Science, Innovation, and Universities
- Distinguished Professorship in Pharmaceutical Sciences II Harry Wm. and Diana V. Hind
- U01HG009080 NHGRI NIH HHS
- 24RT-0025 and 27IR-0030 Tobacco-Related Disease Research Program
- PRE2018-083837 Spanish Ministry of Science, Innovation, and Universities
- UL1 TR001872 NCATS NIH HHS
- RL5GM118984 NIGMS NIH HHS
- Sandler Foundation
Collapse
Affiliation(s)
- Antonio Espuela-Ortiz
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, 38200 San Cristóbal de La Laguna, Spain; (A.E.-O.); (E.H.-L.); (F.L.-D.)
| | - Esther Herrera-Luis
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, 38200 San Cristóbal de La Laguna, Spain; (A.E.-O.); (E.H.-L.); (F.L.-D.)
| | - Fabián Lorenzo-Díaz
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, 38200 San Cristóbal de La Laguna, Spain; (A.E.-O.); (E.H.-L.); (F.L.-D.)
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38200 San Cristóbal de La Laguna, Spain
| | - Donglei Hu
- Department of Medicine, University of California San Francisco, San Francisco, CA 94158, USA; (D.H.); (C.E.); (E.G.B.)
| | - Celeste Eng
- Department of Medicine, University of California San Francisco, San Francisco, CA 94158, USA; (D.H.); (C.E.); (E.G.B.)
| | - Jesús Villar
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Multidisciplinary Organ Dysfunction Evaluation Research Network (MODERN), Research Unit, Hospital Universitario Dr. Negrín, 35019 Las Palmas de Gran Canaria, Spain
| | | | - Esteban G. Burchard
- Department of Medicine, University of California San Francisco, San Francisco, CA 94158, USA; (D.H.); (C.E.); (E.G.B.)
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158, USA
| | - María Pino-Yanes
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, 38200 San Cristóbal de La Laguna, Spain; (A.E.-O.); (E.H.-L.); (F.L.-D.)
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, 38200 San Cristóbal de La Laguna, Spain
| |
Collapse
|
184
|
Bentley AR, Chen G, Doumatey AP, Shriner D, Meeks KAC, Gouveia MH, Ekoru K, Zhou J, Adeyemo A, Rotimi CN. GWAS in Africans identifies novel lipids loci and demonstrates heterogenous association within Africa. Hum Mol Genet 2021; 30:2205-2214. [PMID: 34196372 PMCID: PMC8561421 DOI: 10.1093/hmg/ddab174] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 01/11/2023] Open
Abstract
Serum lipids are biomarkers of cardiometabolic disease risk, and understanding genomic factors contributing to their distribution is of interest. Studies of lipids in Africans are rare, though it is expected that such studies could identify novel loci. We conducted a GWAS of 4317 Africans enrolled from Nigeria, Ghana and Kenya. We evaluated linear mixed models of high-density lipoprotein cholesterol (HDLC), low-density lipoprotein cholesterol (LDLC), total cholesterol (CHOL), triglycerides (TG) and TG/HDLC. Replication was attempted in 9542 African Americans (AA). In our main analysis, we identified 28 novel associations in Africans. Of the 18 of these that could be tested in AA, three associations replicated (GPNMB-TG, ENPP1-TG and SMARCA4-LDLC). Five additional novel loci were discovered upon meta-analysis with AA (rs138282551-TG, PGBD5-HDLC, CD80-TG/HDLC, SLC44A1-CHOL and TLL2-CHOL). Analyses considering only those with predominantly West African ancestry (Nigeria, Ghana and AA) yielded new insights: ORC5-LDLC and chr20:60973327-CHOL. Among our novel findings are some loci with known connections to lipids pathways. For instance, rs147706369 (TLL2) alters a regulatory motif for sterol regulatory element-binding proteins, a family of transcription factors that control the expression of a range of enzymes involved in cholesterol, fatty acid and TG synthesis, and rs115749422 (SMARCA4), an independent association near the known LDLR locus that is rare or absent in populations without African ancestry. These findings demonstrate the utility of conducting genomic analyses in Africans for discovering novel loci and provide some preliminary evidence for caution against treating 'African ancestry' as a monolithic category.
Collapse
Affiliation(s)
- Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Guanjie Chen
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Ayo P Doumatey
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Daniel Shriner
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Karlijn A C Meeks
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Mateus H Gouveia
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Kenneth Ekoru
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Jie Zhou
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Adebowale Adeyemo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| | - Charles N Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
185
|
Jin Q, Shi G. Meta-Analysis of Joint Test of SNP and SNP-Environment Interaction with Heterogeneity. Hum Hered 2021; 86:1-9. [PMID: 34700323 DOI: 10.1159/000519098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 07/29/2021] [Indexed: 12/13/2022] Open
Abstract
Many complex diseases are caused by single nucleotide polymorphisms (SNPs), environmental factors, and the interaction between SNPs and environment. Joint tests of the SNP and SNP-environment interaction effects (JMA) and meta-regression (MR) are commonly used to evaluate these SNP-environment interactions. However, these two methods do not consider genetic heterogeneity. We previously presented a random-effect MR, which provided higher power than the MR in datasets with high heterogeneity. However, this method requires group-level data, which sometimes are not available. Given this, we designed this study to evaluate the introduction of the random effects of SNP and SNP-environment interaction into the JMA, and then extended this to the random effect model. Likelihood ratio statistic is applied to test the JMA and the new method we proposed in this paper. We evaluated the null distributions of these tests, and the powers for this method. This method was verified by simulation and was shown to provide similar powers to the random effect meta-regression method (RMR). However, this method only requires study-level data which relaxed the condition of the RMR. Our study suggests that this method is more suitable for finding the association between SNP and diseases in the absence of group-level data.
Collapse
Affiliation(s)
- Qinqin Jin
- State Key Laboratory of Integrated Services Networks, Xidian University, Xi'an, China.,Applied Science College, Taiyuan University of Science and Technology, Taiyuan, China
| | - Gang Shi
- State Key Laboratory of Integrated Services Networks, Xidian University, Xi'an, China
| |
Collapse
|
186
|
Li B, Ritchie MD. From GWAS to Gene: Transcriptome-Wide Association Studies and Other Methods to Functionally Understand GWAS Discoveries. Front Genet 2021; 12:713230. [PMID: 34659337 PMCID: PMC8515949 DOI: 10.3389/fgene.2021.713230] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022] Open
Abstract
Since their inception, genome-wide association studies (GWAS) have identified more than a hundred thousand single nucleotide polymorphism (SNP) loci that are associated with various complex human diseases or traits. The majority of GWAS discoveries are located in non-coding regions of the human genome and have unknown functions. The valley between non-coding GWAS discoveries and downstream affected genes hinders the investigation of complex disease mechanism and the utilization of human genetics for the improvement of clinical care. Meanwhile, advances in high-throughput sequencing technologies reveal important genomic regulatory roles that non-coding regions play in the transcriptional activities of genes. In this review, we focus on data integrative bioinformatics methods that combine GWAS with functional genomics knowledge to identify genetically regulated genes. We categorize and describe two types of data integrative methods. First, we describe fine-mapping methods. Fine-mapping is an exploratory approach that calibrates likely causal variants underneath GWAS signals. Fine-mapping methods connect GWAS signals to potentially causal genes through statistical methods and/or functional annotations. Second, we discuss gene-prioritization methods. These are hypothesis generating approaches that evaluate whether genetic variants regulate genes via certain genetic regulatory mechanisms to influence complex traits, including colocalization, mendelian randomization, and the transcriptome-wide association study (TWAS). TWAS is a gene-based association approach that investigates associations between genetically regulated gene expression and complex diseases or traits. TWAS has gained popularity over the years due to its ability to reduce multiple testing burden in comparison to other variant-based analytic approaches. Multiple types of TWAS methods have been developed with varied methodological designs and biological hypotheses over the past 5 years. We dive into discussions of how TWAS methods differ in many aspects and the challenges that different TWAS methods face. Overall, TWAS is a powerful tool for identifying complex trait-associated genes. With the advent of single-cell sequencing, chromosome conformation capture, gene editing technologies, and multiplexing reporter assays, we are expecting a more comprehensive understanding of genomic regulation and genetically regulated genes underlying complex human diseases and traits in the future.
Collapse
Affiliation(s)
- Binglan Li
- Department of Biomedical Data Science, Stanford University, Stanford, CA, United States
| | - Marylyn D Ritchie
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, United States.,Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
187
|
Takayanagi Y, Kulason S, Sasabayashi D, Takahashi T, Katagiri N, Sakuma A, Ohmuro N, Katsura M, Nishiyama S, Kido M, Furuichi A, Noguchi K, Matsumoto K, Mizuno M, Ratnanather JT, Suzuki M. Volume Reduction of the Dorsal Lateral Prefrontal Cortex Prior to the Onset of Frank Psychosis in Individuals with an At-Risk Mental State. Cereb Cortex 2021; 32:2245-2253. [PMID: 34649274 DOI: 10.1093/cercor/bhab353] [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: 04/13/2021] [Revised: 08/11/2021] [Accepted: 08/30/2021] [Indexed: 11/14/2022] Open
Abstract
Although some individuals with at-risk mental states (ARMS) develop overt psychosis, surrogate markers which can reliably predict a future onset of psychosis are not well established. The dorsal lateral prefrontal cortex (DLPFC) is thought to be involved in psychotic disorders such as schizophrenia. In this study, 73 ARMS patients and 74 healthy controls underwent 1.5-T 3D magnetic resonance imaging scans at three sites. Using labeled cortical distance mapping, cortical thickness, gray matter (GM) volume, and surface area of DLPFC were estimated. These measures were compared across the diagnostic groups. We also evaluated cognitive function among 36 ARMS subjects to clarify the relationships between the DLPFC morphology and cognitive performance. The GM volume of the right DLPFC was significantly reduced in ARMS subjects who later developed frank psychosis (ARMS-P) relative to those who did not (P = 0.042). There was a positive relationship between the right DLPFC volume and the duration prior to the onset of frank psychosis in ARMS-P subjects (r = 0.58, P = 0.018). Our data may suggest that GM reduction of the DLPFC might be a potential marker of future onset of psychosis in individuals with ARMS.
Collapse
Affiliation(s)
- Yoichiro Takayanagi
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama 9300194, Japan.,Arisawabashi Hospital, Toyama 9392704, Japan
| | - Sue Kulason
- Center for Imaging Science and Institute for Computational Medicine, Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21211, USA
| | - Daiki Sasabayashi
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama 9300194, Japan.,Research Center for Idling Brain Science, University of Toyama, Toyama 9300194, Japan
| | - Tsutomu Takahashi
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama 9300194, Japan.,Research Center for Idling Brain Science, University of Toyama, Toyama 9300194, Japan
| | - Naoyuki Katagiri
- Department of Neuropsychiatry, Toho University School of Medicine, Tokyo 1438541, Japan
| | - Atsushi Sakuma
- Department of Psychiatry, Tohoku University Hospital, Sendai, Miyagi 9808574, Japan
| | - Noriyuki Ohmuro
- Department of Psychiatry, Tohoku University Hospital, Sendai, Miyagi 9808574, Japan.,Osaki Citizen Hospital, Sendai, Miyagi 9896183, Japan
| | - Masahiro Katsura
- Department of Psychiatry, Tohoku University Hospital, Sendai, Miyagi 9808574, Japan
| | - Shimako Nishiyama
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama 9300194, Japan.,Health Administration Center, University of Toyama, Toyama 9308555, Japan
| | - Mikio Kido
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama 9300194, Japan.,Research Center for Idling Brain Science, University of Toyama, Toyama 9300194, Japan
| | - Atsushi Furuichi
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama 9300194, Japan.,Research Center for Idling Brain Science, University of Toyama, Toyama 9300194, Japan
| | - Kyo Noguchi
- Department of Radiology, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama 9300194, Japan
| | - Kazunori Matsumoto
- Department of Psychiatry, Tohoku University Hospital, Sendai, Miyagi 9808574, Japan.,Kokoro no Clinic OASIS, Sendai, Miyagi 9800802, Japan
| | - Masafumi Mizuno
- Department of Neuropsychiatry, Toho University School of Medicine, Tokyo 1438541, Japan
| | - J Tilak Ratnanather
- Center for Imaging Science and Institute for Computational Medicine, Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21211, USA
| | - Michio Suzuki
- Department of Neuropsychiatry, University of Toyama Graduate School of Medicine and Pharmaceutical Sciences, Toyama 9300194, Japan.,Research Center for Idling Brain Science, University of Toyama, Toyama 9300194, Japan
| |
Collapse
|
188
|
Adam Y, Samtal C, Brandenburg JT, Falola O, Adebiyi E. Performing post-genome-wide association study analysis: overview, challenges and recommendations. F1000Res 2021; 10:1002. [PMID: 35222990 PMCID: PMC8847724 DOI: 10.12688/f1000research.53962.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/22/2021] [Indexed: 12/17/2022] Open
Abstract
Genome-wide association studies (GWAS) provide huge information on statistically significant single-nucleotide polymorphisms (SNPs) associated with various human complex traits and diseases. By performing GWAS studies, scientists have successfully identified the association of hundreds of thousands to millions of SNPs to a single phenotype. Moreover, the association of some SNPs with rare diseases has been intensively tested. However, classic GWAS studies have not yet provided solid, knowledgeable insight into functional and biological mechanisms underlying phenotypes or mechanisms of diseases. Therefore, several post-GWAS (pGWAS) methods have been recommended. Currently, there is no simple scientific document to provide a quick guide for performing pGWAS analysis. pGWAS is a crucial step for a better understanding of the biological machinery beyond the SNPs. Here, we provide an overview to performing pGWAS analysis and demonstrate the challenges behind each method. Furthermore, we direct readers to key articles for each pGWAS method and present the overall issues in pGWAS analysis. Finally, we include a custom pGWAS pipeline to guide new users when performing their research.
Collapse
Affiliation(s)
- Yagoub Adam
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota, Ogun, 112233, Nigeria
| | - Chaimae Samtal
- Laboratory of Biotechnology, Environment, Agri-food and Health, Sidi Mohammed Ben Abdellah University, Fez, Fez-Meknes, 30000, Morocco
| | - Jean-tristan Brandenburg
- Sydney Brenner Institute for Molecular Bioscience (SBIMB), University of the Witwatersrand, Johannesburg, South Africa
| | - Oluwadamilare Falola
- Laboratory of Biotechnology, Environment, Agri-food and Health, Sidi Mohammed Ben Abdellah University, Fez, Fez-Meknes, 30000, Morocco
| | - Ezekiel Adebiyi
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota, Ogun, 112233, Nigeria
- Computer & Information Sciences, Covenant University, Ota, Ogun, 112233, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence, Covenant University, Ota, Ogun, 112233, Nigeria
- Applied Bioinformatics Division, German Cancer Center DKFZ - Heidelberg University, Heidelberg, Baden-Württemberg, 69120, Germany
| |
Collapse
|
189
|
Shorey S, Ng ED, Wong CHJ. Global prevalence of depression and elevated depressive symptoms among adolescents: A systematic review and meta-analysis. BRITISH JOURNAL OF CLINICAL PSYCHOLOGY 2021; 61:287-305. [PMID: 34569066 DOI: 10.1111/bjc.12333] [Citation(s) in RCA: 523] [Impact Index Per Article: 130.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 08/02/2021] [Indexed: 12/01/2022]
Abstract
OBJECTIVE Adolescence is a formative and turbulent phase where physiological, psychosocial, and cognitive changes leave adolescents vulnerable to psychological disorders. Given the lack of reviews that consolidate and compare worldwide prevalence of depression among adolescents, this review aims to examine the global prevalence of major depressive disorders, dysthymia, and elevated depressive symptoms among adolescents. METHODS A systematic review and meta-analysis was conducted. Six databases were searched for studies published from 2001 to December 2020. Seventy-two studies were included. Subgroup analysis were performed for year of publication, geographical region, gender, and assessment tools used. RESULTS The global point prevalence rate of elevated self-reported depressive symptoms from 2001 to 2020 was 34% (95% CI: 0.30-0.38). Point prevalence for major depressive disorder (MDD) and dysthymia was 8% (95% CI: 0.02-0.13) and 4% (95% CI: 0.01-0.07), respectively. The pooled one-year prevalence and lifetime prevalence for MDD were 8% (95% CI: 0.05-0.12) and 19% (95% CI: 0.12-0.26). Point prevalence of elevated depressive symptoms among adolescents increased from 24% (95% CI: 0.19-0.28) between 2001 and 2010 to 37% (95% CI: 0.32-0.42) between 2011 and 2020. The Middle East, Africa, and Asia have the highest prevalence of elevated depressive symptoms, and female adolescents were reported to have a higher prevalence of elevated depressive symptoms than male adolescents. CONCLUSION Besides targeting those with existing clinical depression, research and policies should also focus on educational and supportive mitigation efforts to curb depressive symptoms among adolescents before escalation. The findings encourage future research to develop more gender-specific and culturally relevant intervention programmes. PRACTITIONER POINTS 34% of adolescents globally, aged 10-19 years, are at risk of developing clinical depression, which exceeds the reported estimates of individuals aged 18 to 25 years. Practitioners are highly encouraged to prioritize depression screening and intervention implementation for individuals in this age group. Female adolescents and adolescents from Middle East, Africa, and Asia have the highest risk of developing depression. This urges practitioners and researchers to develop more gender-specific and culturally relevant intervention programmes.
Collapse
Affiliation(s)
- Shefaly Shorey
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Esperanza Debby Ng
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Celine H J Wong
- Department of Psychological Medicine, National University Hospital, Singapore
| |
Collapse
|
190
|
Casares-Marfil D, Strauss M, Bosch-Nicolau P, Lo Presti MS, Molina I, Chevillard C, Cunha-Neto E, Sabino E, Ribeiro ALP, González CI, Martín J, Acosta-Herrera M. A Genome-Wide Association Study Identifies Novel Susceptibility loci in Chronic Chagas Cardiomyopathy. Clin Infect Dis 2021; 73:672-679. [PMID: 33539531 DOI: 10.1093/cid/ciab090] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 02/01/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Chagas disease is an infectious disease caused by the parasite Trypanosoma cruzi and is endemic from Latin American countries. The goal of our study was to identify novel genetic loci associated with chronic Chagas cardiomyopathy development in Chagas disease patients from different Latin American populations. METHODS We performed a cross-sectional, nested case-control study including 3 sample collections from Colombia, Argentina, and Bolivia. Samples were genotyped to conduct a genome-wide association study (GWAS). These results were meta-analyzed with summary statistic data from Brazil, gathering a total of 3413 Chagas disease patients. To identify the functional impact of the associated variant and its proxies, we performed an in silico analysis of this region. RESULTS The meta-analysis revealed a novel genome-wide statistically significant association with chronic Chagas cardiomyopathy development in rs2458298 (OR = 0.90, 95%CI = 0.87-0.94, P-value = 3.27 × 10-08), nearby the SAC3D1 gene. In addition, further in silico analyses displayed functional relationships between the associated variant and the SNX15, BAFT2, and FERMT3 genes, related to cardiovascular traits. CONCLUSIONS Our findings support the role of the host genetic factors in the susceptibility to the development of the chronic cardiac form of this neglected disease.
Collapse
Affiliation(s)
| | - Mariana Strauss
- Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, FCM, INICSA-CONICET-UNC, Córdoba, Argentina
| | - Pau Bosch-Nicolau
- Unidad de Medicina Tropical y Salud Internacional Hospital Universitari Vall d'Hebron, PROSICS, Barcelona, Spain
| | - María Silvina Lo Presti
- Centro de Estudios e Investigación de la Enfermedad de Chagas y Leishmaniasis, FCM, INICSA-CONICET-UNC, Córdoba, Argentina
| | - Israel Molina
- Unidad de Medicina Tropical y Salud Internacional Hospital Universitari Vall d'Hebron, PROSICS, Barcelona, Spain
| | | | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute (InCor)/Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Ester Sabino
- Instituto de Medicina Tropical Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Antonio Luiz P Ribeiro
- Centro de Telessaúde, Hospital das Clínicas, Belo Horizonte, Brazil.,Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Clara Isabel González
- Grupo de Inmunología y Epidemiología Molecular, Escuela de Microbiología, Universidad Industrial de Santander, Bucaramanga, Colombia
| | - Javier Martín
- Instituto de Parasitología y Biomedicina López-Neyra, CSIC, Granada, Spain
| | | |
Collapse
|
191
|
LaPierre N, Taraszka K, Huang H, He R, Hormozdiari F, Eskin E. Identifying causal variants by fine mapping across multiple studies. PLoS Genet 2021; 17:e1009733. [PMID: 34543273 PMCID: PMC8491908 DOI: 10.1371/journal.pgen.1009733] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/05/2021] [Accepted: 07/21/2021] [Indexed: 11/18/2022] Open
Abstract
Increasingly large Genome-Wide Association Studies (GWAS) have yielded numerous variants associated with many complex traits, motivating the development of "fine mapping" methods to identify which of the associated variants are causal. Additionally, GWAS of the same trait for different populations are increasingly available, raising the possibility of refining fine mapping results further by leveraging different linkage disequilibrium (LD) structures across studies. Here, we introduce multiple study causal variants identification in associated regions (MsCAVIAR), a method that extends the popular CAVIAR fine mapping framework to a multiple study setting using a random effects model. MsCAVIAR only requires summary statistics and LD as input, accounts for uncertainty in association statistics using a multivariate normal model, allows for multiple causal variants at a locus, and explicitly models the possibility of different SNP effect sizes in different populations. We demonstrate the efficacy of MsCAVIAR in both a simulation study and a trans-ethnic, trans-biobank fine mapping analysis of High Density Lipoprotein (HDL).
Collapse
Affiliation(s)
- Nathan LaPierre
- Department of Computer Science, University of California, Los Angeles, California, United States
| | - Kodi Taraszka
- Department of Computer Science, University of California, Los Angeles, California, United States
| | - Helen Huang
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, California, United States
| | - Rosemary He
- Department of Mathematics, University of California, Los Angeles, California, United States
| | - Farhad Hormozdiari
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States
| | - Eleazar Eskin
- Department of Computer Science, University of California, Los Angeles, California, United States
- Department of Human Genetics, University of California, Los Angeles, California, United States
- Department of Computational Medicine, University of California, Los Angeles, California, United States
| |
Collapse
|
192
|
Wu P, Ding L, Li X, Liu S, Cheng F, He Q, Xiao M, Wu P, Hou H, Jiang M, Long P, Wang H, Liu L, Qu M, Shi X, Jiang Q, Mo T, Ding W, Fu Y, Han S, Huo X, Zeng Y, Zhou Y, Zhang Q, Ke J, Xu X, Ni W, Shao Z, Wang J, Liu P, Li Z, Jin Y, Zheng F, Wang F, Liu L, Li W, Liu K, Peng R, Xu X, Lin Y, Gao H, Shi L, Geng Z, Mu X, Yan Y, Wang K, Wu D, Hao X, Cheng S, Qiu G, Guo H, Li K, Chen G, Sun Z, Lin X, Jin X, Wang F, Sun C, Wang C. Trans-ethnic genome-wide association study of severe COVID-19. Commun Biol 2021; 4:1034. [PMID: 34465887 PMCID: PMC8408224 DOI: 10.1038/s42003-021-02549-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/12/2021] [Indexed: 01/08/2023] Open
Abstract
COVID-19 has caused numerous infections with diverse clinical symptoms. To identify human genetic variants contributing to the clinical development of COVID-19, we genotyped 1457 (598/859 with severe/mild symptoms) and sequenced 1141 (severe/mild: 474/667) patients of Chinese ancestry. We further incorporated 1401 genotyped and 948 sequenced ancestry-matched population controls, and tested genome-wide association on 1072 severe cases versus 3875 mild or population controls, followed by trans-ethnic meta-analysis with summary statistics of 3199 hospitalized cases and 897,488 population controls from the COVID-19 Host Genetics Initiative. We identified three significant signals outside the well-established 3p21.31 locus: an intronic variant in FOXP4-AS1 (rs1853837, odds ratio OR = 1.28, P = 2.51 × 10-10, allele frequencies in Chinese/European AF = 0.345/0.105), a frameshift insertion in ABO (rs8176719, OR = 1.19, P = 8.98 × 10-9, AF = 0.422/0.395) and a Chinese-specific intronic variant in MEF2B (rs74490654, OR = 8.73, P = 1.22 × 10-8, AF = 0.004/0). These findings highlight an important role of the adaptive immunity and the ABO blood-group system in protection from developing severe COVID-19.
Collapse
Affiliation(s)
- Peng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Medical Center for Major Public Health Events, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Ding
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaodong Li
- Hepatic Disease Institute, Hubei Key Laboratory of Theoretical and Applied Research of Liver and Kidney in Traditional Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Provincial Academy of Traditional Chinese Medicine, Wuhan, China
| | - Siyang Liu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Fanjun Cheng
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing He
- The Third People's Hospital of Shenzhen, National Clinical Research Center for Infectious Disease, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Mingzhong Xiao
- Hepatic Disease Institute, Hubei Key Laboratory of Theoretical and Applied Research of Liver and Kidney in Traditional Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Provincial Academy of Traditional Chinese Medicine, Wuhan, China
| | - Ping Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Medical Center for Major Public Health Events, Huazhong University of Science and Technology, Wuhan, China
| | - Hongyan Hou
- National Medical Center for Major Public Health Events, Huazhong University of Science and Technology, Wuhan, China
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minghui Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pinpin Long
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Wang
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Linlin Liu
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
| | - Minghan Qu
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xian Shi
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Jiang
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Mo
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wencheng Ding
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Medical Center for Major Public Health Events, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Fu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Medical Center for Major Public Health Events, Huazhong University of Science and Technology, Wuhan, China
| | - Shi Han
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
| | - Xixiang Huo
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
| | - Yingchun Zeng
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
| | - Yana Zhou
- Hepatic Disease Institute, Hubei Key Laboratory of Theoretical and Applied Research of Liver and Kidney in Traditional Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Provincial Academy of Traditional Chinese Medicine, Wuhan, China
| | - Qing Zhang
- Hepatic Disease Institute, Hubei Key Laboratory of Theoretical and Applied Research of Liver and Kidney in Traditional Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Provincial Academy of Traditional Chinese Medicine, Wuhan, China
| | - Jia Ke
- Hepatic Disease Institute, Hubei Key Laboratory of Theoretical and Applied Research of Liver and Kidney in Traditional Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Provincial Academy of Traditional Chinese Medicine, Wuhan, China
| | - Xi Xu
- Hepatic Disease Institute, Hubei Key Laboratory of Theoretical and Applied Research of Liver and Kidney in Traditional Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Provincial Academy of Traditional Chinese Medicine, Wuhan, China
| | - Wei Ni
- Hepatic Disease Institute, Hubei Key Laboratory of Theoretical and Applied Research of Liver and Kidney in Traditional Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Provincial Academy of Traditional Chinese Medicine, Wuhan, China
| | - Zuoyu Shao
- Hepatic Disease Institute, Hubei Key Laboratory of Theoretical and Applied Research of Liver and Kidney in Traditional Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Provincial Academy of Traditional Chinese Medicine, Wuhan, China
| | - Jingzhi Wang
- Hepatic Disease Institute, Hubei Key Laboratory of Theoretical and Applied Research of Liver and Kidney in Traditional Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Provincial Academy of Traditional Chinese Medicine, Wuhan, China
| | - Panhong Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Zilong Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yan Jin
- Department of Emergency, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Zheng
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Wang
- The Third People's Hospital of Shenzhen, National Clinical Research Center for Infectious Disease, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Lei Liu
- The Third People's Hospital of Shenzhen, National Clinical Research Center for Infectious Disease, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Wending Li
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kang Liu
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Peng
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuedan Xu
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuhui Lin
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Gao
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Limei Shi
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziyue Geng
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuanwen Mu
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Degang Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shanshan Cheng
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gaokun Qiu
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huan Guo
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kezhen Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Medical Center for Major Public Health Events, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Medical Center for Major Public Health Events, Huazhong University of Science and Technology, Wuhan, China
| | - Ziyong Sun
- National Medical Center for Major Public Health Events, Huazhong University of Science and Technology, Wuhan, China
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xihong Lin
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Statistics, Harvard University, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Xin Jin
- School of Medicine, South China University of Technology, Guangzhou, China.
| | - Feng Wang
- National Medical Center for Major Public Health Events, Huazhong University of Science and Technology, Wuhan, China.
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Chaoyang Sun
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- National Medical Center for Major Public Health Events, Huazhong University of Science and Technology, Wuhan, China.
| | - Chaolong Wang
- National Medical Center for Major Public Health Events, Huazhong University of Science and Technology, Wuhan, China.
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Ministry of Education Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
193
|
Wallen ZD, Stone WJ, Factor SA, Molho E, Zabetian CP, Standaert DG, Payami H. Exploring human-genome gut-microbiome interaction in Parkinson's disease. NPJ PARKINSONS DISEASE 2021; 7:74. [PMID: 34408160 PMCID: PMC8373869 DOI: 10.1038/s41531-021-00218-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 07/23/2021] [Indexed: 12/16/2022]
Abstract
The causes of complex diseases remain an enigma despite decades of epidemiologic research on environmental risks and genome-wide studies that have uncovered tens or hundreds of susceptibility loci for each disease. We hypothesize that the microbiome is the missing link. Genetic studies have shown that overexpression of alpha-synuclein, a key pathological protein in Parkinson’s disease (PD), can cause familial PD and variants at alpha-synuclein locus confer risk of idiopathic PD. Recently, dysbiosis of gut microbiome in PD was identified: altered abundances of three microbial clusters were found, one of which was composed of opportunistic pathogens. Using two large datasets, we found evidence that the overabundance of opportunistic pathogens in PD gut is influenced by the host genotype at the alpha-synuclein locus, and that the variants responsible modulate alpha-synuclein expression. Results put forth testable hypotheses on the role of gut microbiome in the pathogenesis of PD, the incomplete penetrance of PD susceptibility genes, and potential triggers of pathology in the gut.
Collapse
Affiliation(s)
- Zachary D Wallen
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - William J Stone
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stewart A Factor
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Eric Molho
- Department of Neurology, Albany Medical College, Albany, NY, USA
| | - Cyrus P Zabetian
- VA Puget Sound Health Care System and Department of Neurology, University of Washington, Seattle, WA, USA
| | - David G Standaert
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Haydeh Payami
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
194
|
Topaloudi A, Zagoriti Z, Flint AC, Martinez MB, Yang Z, Tsetsos F, Christou YP, Lagoumintzis G, Yannaki E, Zamba-Papanicolaou E, Tzartos J, Tsekmekidou X, Kotsa K, Maltezos E, Papanas N, Papazoglou D, Passadakis P, Roumeliotis A, Roumeliotis S, Theodoridis M, Thodis E, Panagoutsos S, Yovos J, Stamatoyannopoulos J, Poulas K, Kleopa K, Tzartos S, Georgitsi M, Paschou P. Myasthenia gravis genome-wide association study implicates AGRN as a risk locus. J Med Genet 2021; 59:801-809. [PMID: 34400559 DOI: 10.1136/jmedgenet-2021-107953] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 07/20/2021] [Indexed: 11/04/2022]
Abstract
BACKGROUND Myasthenia gravis (MG) is a rare autoimmune disorder affecting the neuromuscular junction (NMJ). Here, we investigate the genetic architecture of MG via a genome-wide association study (GWAS) of the largest MG data set analysed to date. METHODS We performed GWAS meta-analysis integrating three different data sets (total of 1401 cases and 3508 controls). We carried out human leucocyte antigen (HLA) fine-mapping, gene-based and tissue enrichment analyses and investigated genetic correlation with 13 other autoimmune disorders as well as pleiotropy across MG and correlated disorders. RESULTS We confirmed the previously reported MG association with TNFRSF11A (rs4369774; p=1.09×10-13, OR=1.4). Furthermore, gene-based analysis revealed AGRN as a novel MG susceptibility gene. HLA fine-mapping pointed to two independent MG loci: HLA-DRB1 and HLA-B. MG onset-specific analysis reveals differences in the genetic architecture of early-onset MG (EOMG) versus late-onset MG (LOMG). Furthermore, we find MG to be genetically correlated with type 1 diabetes (T1D), rheumatoid arthritis (RA), late-onset vitiligo and autoimmune thyroid disease (ATD). Cross-disorder meta-analysis reveals multiple risk loci that appear pleiotropic across MG and correlated disorders. DISCUSSION Our gene-based analysis identifies AGRN as a novel MG susceptibility gene, implicating for the first time a locus encoding a protein (agrin) that is directly relevant to NMJ activation. Mutations in AGRN have been found to underlie congenital myasthenic syndrome. Our results are also consistent with previous studies highlighting the role of HLA and TNFRSF11A in MG aetiology and the different risk genes in EOMG versus LOMG. Finally, we uncover the genetic correlation of MG with T1D, RA, ATD and late-onset vitiligo, pointing to shared underlying genetic mechanisms.
Collapse
Affiliation(s)
- Apostolia Topaloudi
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Zoi Zagoriti
- Department of Pharmacy, University of Patras, Rio, Greece
| | - Alyssa Camille Flint
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA
| | | | - Zhiyu Yang
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Fotis Tsetsos
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupoli, Greece
| | | | | | - Evangelia Yannaki
- Department of Hematology, George Papanicolaou Hospital, Thessaloniki, Greece
| | - Eleni Zamba-Papanicolaou
- The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus.,Department of Neuroepidemiology and Centre for Neuromuscular Disorders, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | | | - Xanthippi Tsekmekidou
- Division of Endocrinology and Metabolism-Diabetes Center, 1st Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kalliopi Kotsa
- Division of Endocrinology and Metabolism-Diabetes Center, 1st Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Efstratios Maltezos
- Diabetes Center, 2nd Department of Internal Medicine, Alexandroupolis University General Hospital, Democritus University of Thrace, Alexandroupoli, Greece
| | - Nikolaos Papanas
- Diabetes Center, 2nd Department of Internal Medicine, Alexandroupolis University General Hospital, Democritus University of Thrace, Alexandroupoli, Greece
| | - Dimitrios Papazoglou
- Diabetes Center, 2nd Department of Internal Medicine, Alexandroupolis University General Hospital, Democritus University of Thrace, Alexandroupoli, Greece
| | - Ploumis Passadakis
- Department of Nephrology, Alexandroupolis University General Hospital, Democritus University of Thrace, Alexandroupoli, Greece
| | - Athanasios Roumeliotis
- Division of Nephrology and Hypertension, 1st Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Stefanos Roumeliotis
- Division of Nephrology and Hypertension, 1st Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Marios Theodoridis
- Department of Nephrology, Alexandroupolis University General Hospital, Democritus University of Thrace, Alexandroupoli, Greece
| | - Elias Thodis
- Department of Nephrology, Alexandroupolis University General Hospital, Democritus University of Thrace, Alexandroupoli, Greece
| | - Stylianos Panagoutsos
- Department of Nephrology, Alexandroupolis University General Hospital, Democritus University of Thrace, Alexandroupoli, Greece
| | - John Yovos
- Division of Endocrinology and Metabolism-Diabetes Center, 1st Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - John Stamatoyannopoulos
- Departments of Medicine and Genome Sciences, University of Washington, Seattle, Washington, USA
| | | | - Kleopas Kleopa
- The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus.,Department of Neuroscience and Centre for Neuromuscular Disorders, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Socrates Tzartos
- Department of Pharmacy, University of Patras, Rio, Greece.,Hellenic Pasteur Institute, Athens, Greece
| | - Marianthi Georgitsi
- 1st Laboratory of Medical Biology-Genetics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Peristera Paschou
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
195
|
Hale AT, Akinnusotu O, He J, Wang J, Hibshman N, Shannon CN, Naftel RP. Genome-Wide Association Study Identifies Genetic Risk Factors for Spastic Cerebral Palsy. Neurosurgery 2021; 89:435-442. [PMID: 34098570 PMCID: PMC8364821 DOI: 10.1093/neuros/nyab184] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/31/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Although many clinical risk factors of spastic cerebral palsy (CP) have been identified, the genetic basis of spastic CP is largely unknown. Here, using whole-genome genetic information linked to a deidentified electronic health record (BioVU) with replication in the UK Biobank and FinnGen, we perform the first genome-wide association study (GWAS) for spastic CP. OBJECTIVE To define the genetic basis of spastic CP. METHODS Whole-genome data were obtained using the multi-ethnic genotyping array (MEGA) genotyping array capturing single-nucleotide polymorphisms (SNPs), minor allele frequency (MAF) > 0.01, and imputation quality score (r2) > 0.3, imputed based on the 1000 genomes phase 3 reference panel. Threshold for genome-wide significance was defined after Bonferroni correction for the total number of SNPs tested (P < 5.0 × 10-8). Replication analysis (defined as P < .05) was performed in the UK Biobank and FinnGen. RESULTS We identify 1 SNP (rs78686911) reaching genome-wide significance with spastic CP. Expression quantitative trait loci (eQTL) analysis suggests that rs78686911 decreases expression of GRIK4, a gene that encodes a high-affinity kainate glutamatergic receptor of largely unknown function. Replication analysis in the UK Biobank and FinnGen reveals additional SNPs in the GRIK4 loci associated with CP. CONCLUSION To our knowledge, we perform the first GWAS of spastic CP. Our study indicates that genetic variation contributes to CP risk.
Collapse
Affiliation(s)
- Andrew T Hale
- Vanderbilt University School of Medicine, Medical Scientist Training Program, Nashville, Tennessee, USA
- Surgical Outcomes Center for Kids, Monroe Carell Jr Children's Hospital of Vanderbilt University, Nashville, Tennessee, USA
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Oluwatoyin Akinnusotu
- Surgical Outcomes Center for Kids, Monroe Carell Jr Children's Hospital of Vanderbilt University, Nashville, Tennessee, USA
| | - Jing He
- Department of Bioinformatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Janey Wang
- Department of Bioinformatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Natalie Hibshman
- Surgical Outcomes Center for Kids, Monroe Carell Jr Children's Hospital of Vanderbilt University, Nashville, Tennessee, USA
| | - Chevis N Shannon
- Surgical Outcomes Center for Kids, Monroe Carell Jr Children's Hospital of Vanderbilt University, Nashville, Tennessee, USA
- Division of Pediatric Neurosurgery, Monroe Carell Jr Children's Hospital of Vanderbilt University, Nashville, Tennessee, USA
| | - Robert P Naftel
- Surgical Outcomes Center for Kids, Monroe Carell Jr Children's Hospital of Vanderbilt University, Nashville, Tennessee, USA
- Division of Pediatric Neurosurgery, Monroe Carell Jr Children's Hospital of Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
196
|
Casares-Marfil D, Guillen-Guio B, Lorenzo-Salazar JM, Rodríguez-Pérez H, Kerick M, Jaimes-Campos MA, Díaz ML, Estupiñán E, Echeverría LE, González CI, Martin J, Flores C, Acosta-Herrera M. Admixture mapping analysis reveals differential genetic ancestry associated with Chagas disease susceptibility in the Colombian population. Hum Mol Genet 2021; 30:2503-2512. [PMID: 34302177 PMCID: PMC8643504 DOI: 10.1093/hmg/ddab213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 01/02/2023] Open
Abstract
Chagas disease is an infection caused by the parasite Trypanosoma cruzi, endemic in Latino America. Leveraging the three-way admixture between Native American (AMR), European (EUR) and African (AFR) populations in Latin Americans, we aimed to better understand the genetic basis of Chagas disease by performing an admixture mapping study in a Colombian population. A two-stage study was conducted, and subjects were classified as seropositive and seronegative for T. cruzi. In stage 1, global and local ancestries were estimated using reference data from the 1000 Genomes Project (1KGP) and local ancestry associations were performed by logistic regression models. The AMR ancestry showed a protective association with Chagas disease within the Major Histocompatibility Complex region (OR = 0.74, 95%CI = 0.66-0.83, lowest p-value = 4.53x10-8). The fine mapping assessment on imputed genotypes combining data from stage 1 and 2 from an independent Colombian cohort, revealed nominally associated variants in high linkage disequilibrium with the top signal (rs2032134, OR = 0.93, 95%CI = 0.90-0.97, p-value = 3.54x10-4) in the previously associated locus. To assess ancestry-specific adaptive signals, a selective sweep scan in an AMR reference population from 1KGP together with an in silico functional analysis highlighted the Tripartite Motif family and the Human Leukocyte Antigen (HLA) genes, with crucial role in the immune response against pathogens. Furthermore, these analyses emphasized the macrophages, neutrophils, and eosinophils, as key players in the defense against T. cruzi. This first admixture mapping study in Chagas disease provided novel insights underlying the host immune response in the pathogenesis of this neglected disease.
Collapse
Affiliation(s)
| | - Beatriz Guillen-Guio
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Jose M Lorenzo-Salazar
- Genomics Division, Instituto Tecnológico y de Energías Renovables (ITER), Santa Cruz de Tenerife, Spain
| | - Héctor Rodríguez-Pérez
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Martin Kerick
- Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain
| | - Mayra A Jaimes-Campos
- Grupo de Inmunología y Epidemiología Molecular, Escuela de Microbiología, Universidad Industrial de Santander, Bucaramanga, Colombia
| | - Martha L Díaz
- Grupo de Inmunología y Epidemiología Molecular, Escuela de Microbiología, Universidad Industrial de Santander, Bucaramanga, Colombia
| | - Elkyn Estupiñán
- Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain.,Grupo de Inmunología y Epidemiología Molecular, Escuela de Microbiología, Universidad Industrial de Santander, Bucaramanga, Colombia
| | - Luis E Echeverría
- Heart Failure and Heart Transplant Clinic, Fundación Cardiovascular de Colombia, Floridablanca, Colombia
| | - Clara I González
- Grupo de Inmunología y Epidemiología Molecular, Escuela de Microbiología, Universidad Industrial de Santander, Bucaramanga, Colombia
| | - Javier Martin
- Institute of Parasitology and Biomedicine López-Neyra, CSIC, Granada, Spain
| | - Carlos Flores
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain.,Genomics Division, Instituto Tecnológico y de Energías Renovables (ITER), Santa Cruz de Tenerife, Spain.,CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | | |
Collapse
|
197
|
Wang Y, Li T, Fu L, Yang S, Hu YQ. A Novel Method for Mendelian Randomization Analyses With Pleiotropy and Linkage Disequilibrium in Genetic Variants From Individual Data. Front Genet 2021; 12:634394. [PMID: 34322150 PMCID: PMC8312241 DOI: 10.3389/fgene.2021.634394] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 06/15/2021] [Indexed: 12/28/2022] Open
Abstract
Mendelian randomization makes use of genetic variants as instrumental variables to eliminate the influence induced by unknown confounders on causal estimation in epidemiology studies. However, with the soaring genetic variants identified in genome-wide association studies, the pleiotropy, and linkage disequilibrium in genetic variants are unavoidable and may produce severe bias in causal inference. In this study, by modeling the pleiotropic effect as a normally distributed random effect, we propose a novel mixed-effects regression model-based method PLDMR, pleiotropy and linkage disequilibrium adaptive Mendelian randomization, which takes linkage disequilibrium into account and also corrects for the pleiotropic effect in causal effect estimation and statistical inference. We conduct voluminous simulation studies to evaluate the performance of the proposed and existing methods. Simulation results illustrate the validity and advantage of the novel method, especially in the case of linkage disequilibrium and directional pleiotropic effects, compared with other methods. In addition, by applying this novel method to the data on Atherosclerosis Risk in Communications Study, we conclude that body mass index has a significant causal effect on and thus might be a potential risk factor of systolic blood pressure. The novel method is implemented in R and the corresponding R code is provided for free download.
Collapse
Affiliation(s)
- Yuquan Wang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Institute of Biostatistics, School of Life Sciences, Fudan University, Shanghai, China
| | - Tingting Li
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Institute of Biostatistics, School of Life Sciences, Fudan University, Shanghai, China
| | - Liwan Fu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Institute of Biostatistics, School of Life Sciences, Fudan University, Shanghai, China
| | - Siqian Yang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Institute of Biostatistics, School of Life Sciences, Fudan University, Shanghai, China
| | - Yue-Qing Hu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Institute of Biostatistics, School of Life Sciences, Fudan University, Shanghai, China
- Shanghai Center for Mathematical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
198
|
Rowan TN, Durbin HJ, Seabury CM, Schnabel RD, Decker JE. Powerful detection of polygenic selection and evidence of environmental adaptation in US beef cattle. PLoS Genet 2021; 17:e1009652. [PMID: 34292938 PMCID: PMC8297814 DOI: 10.1371/journal.pgen.1009652] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 06/09/2021] [Indexed: 12/19/2022] Open
Abstract
Selection on complex traits can rapidly drive evolution, especially in stressful environments. This polygenic selection does not leave intense sweep signatures on the genome, rather many loci experience small allele frequency shifts, resulting in large cumulative phenotypic changes. Directional selection and local adaptation are changing populations; but, identifying loci underlying polygenic or environmental selection has been difficult. We use genomic data on tens of thousands of cattle from three populations, distributed over time and landscapes, in linear mixed models with novel dependent variables to map signatures of selection on complex traits and local adaptation. We identify 207 genomic loci associated with an animal's birth date, representing ongoing selection for monogenic and polygenic traits. Additionally, hundreds of additional loci are associated with continuous and discrete environments, providing evidence for historical local adaptation. These candidate loci highlight the nervous system's possible role in local adaptation. While advanced technologies have increased the rate of directional selection in cattle, it has likely been at the expense of historically generated local adaptation, which is especially problematic in changing climates. When applied to large, diverse cattle datasets, these selection mapping methods provide an insight into how selection on complex traits continually shapes the genome. Further, understanding the genomic loci implicated in adaptation may help us breed more adapted and efficient cattle, and begin to understand the basis for mammalian adaptation, especially in changing climates. These selection mapping approaches help clarify selective forces and loci in evolutionary, model, and agricultural contexts.
Collapse
Affiliation(s)
- Troy N. Rowan
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, United States of America
- Genetics Area Program, University of Missouri, Columbia, Missouri, United States of America
- Department of Animal Science, University of Tennessee, Knoxville, Tennessee, United States of America
- College of Veterinary Medicine, Large Animal Clinical Science, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Harly J. Durbin
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, United States of America
- Genetics Area Program, University of Missouri, Columbia, Missouri, United States of America
| | - Christopher M. Seabury
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - Robert D. Schnabel
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, United States of America
- Genetics Area Program, University of Missouri, Columbia, Missouri, United States of America
- Institute for Data Science and Informatics, University of Missouri, Columbia, Missouri, United States of America
| | - Jared E. Decker
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, United States of America
- Genetics Area Program, University of Missouri, Columbia, Missouri, United States of America
- Institute for Data Science and Informatics, University of Missouri, Columbia, Missouri, United States of America
| |
Collapse
|
199
|
Jhun MA, Mendelson M, Wilson R, Gondalia R, Joehanes R, Salfati E, Zhao X, Braun KVE, Do AN, Hedman ÅK, Zhang T, Carnero-Montoro E, Shen J, Bartz TM, Brody JA, Montasser ME, O’Connell JR, Yao C, Xia R, Boerwinkle E, Grove M, Guan W, Liliane P, Singmann P, Müller-Nurasyid M, Meitinger T, Gieger C, Peters A, Zhao W, Ware EB, Smith JA, Dhana K, van Meurs J, Uitterlinden A, Ikram MA, Ghanbari M, Zhi D, Gustafsson S, Lind L, Li S, Sun D, Spector TD, Chen YDI, Damcott C, Shuldiner AR, Absher DM, Horvath S, Tsao PS, Kardia S, Psaty BM, Sotoodehnia N, Bell JT, Ingelsson E, Chen W, Dehghan A, Arnett DK, Waldenberger M, Hou L, Whitsel EA, Baccarelli A, Levy D, Fornage M, Irvin MR, Assimes TL. A multi-ethnic epigenome-wide association study of leukocyte DNA methylation and blood lipids. Nat Commun 2021; 12:3987. [PMID: 34183656 PMCID: PMC8238961 DOI: 10.1038/s41467-021-23899-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
Here we examine the association between DNA methylation in circulating leukocytes and blood lipids in a multi-ethnic sample of 16,265 subjects. We identify 148, 35, and 4 novel associations among Europeans, African Americans, and Hispanics, respectively, and an additional 186 novel associations through a trans-ethnic meta-analysis. We observe a high concordance in the direction of effects across racial/ethnic groups, a high correlation of effect sizes between high-density lipoprotein and triglycerides, a modest overlap of associations with epigenome-wide association studies of other cardio-metabolic traits, and a largely non-overlap with lipid loci identified to date through genome-wide association studies. Thirty CpGs reached significance in at least 2 racial/ethnic groups including 7 that showed association with the expression of an annotated gene. CpGs annotated to CPT1A showed evidence of being influenced by triglycerides levels. DNA methylation levels of circulating leukocytes show robust and consistent association with blood lipid levels across multiple racial/ethnic groups.
Collapse
Affiliation(s)
- Min-A Jhun
- grid.214458.e0000000086837370Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI USA ,grid.168010.e0000000419368956Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Michael Mendelson
- grid.94365.3d0000 0001 2297 5165Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD USA ,grid.2515.30000 0004 0378 8438Department of Cardiology, Boston Children’s Hospital, Boston, MA USA
| | - Rory Wilson
- grid.4567.00000 0004 0483 2525Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany ,grid.4567.00000 0004 0483 2525Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Rahul Gondalia
- grid.410711.20000 0001 1034 1720Department of Epidemiology, University of North Carolina, Chapel Hill, NC USA
| | - Roby Joehanes
- grid.38142.3c000000041936754XHebrew SeniorLife, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA USA
| | - Elias Salfati
- grid.168010.e0000000419368956Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Xiaoping Zhao
- grid.267308.80000 0000 9206 2401The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX USA
| | - Kim Valeska Emilie Braun
- grid.5645.2000000040459992XDepartment of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Anh Nguyet Do
- grid.265892.20000000106344187Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL USA
| | - Åsa K. Hedman
- grid.8993.b0000 0004 1936 9457Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Tao Zhang
- grid.265219.b0000 0001 2217 8588Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA USA
| | - Elena Carnero-Montoro
- grid.13097.3c0000 0001 2322 6764Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King’s College London, London, UK ,grid.470860.d0000 0004 4677 7069GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, Granada, Spain
| | - Jincheng Shen
- grid.223827.e0000 0001 2193 0096Department of Population Health Sciences, University of Utah, Salt Lake City, UT USA
| | - Traci M. Bartz
- grid.34477.330000000122986657Cardiovascular Health Research Unit, Departments of Medicine and Biostatistics, University of Washington, Seattle, WA USA
| | - Jennifer A. Brody
- grid.34477.330000000122986657Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA USA
| | - May E. Montasser
- grid.411024.20000 0001 2175 4264Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD USA ,grid.411024.20000 0001 2175 4264Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD USA
| | - Jeff R. O’Connell
- grid.411024.20000 0001 2175 4264Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD USA ,grid.411024.20000 0001 2175 4264Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD USA
| | - Chen Yao
- grid.94365.3d0000 0001 2297 5165Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD USA
| | - Rui Xia
- grid.267308.80000 0000 9206 2401The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX USA
| | - Eric Boerwinkle
- grid.267308.80000 0000 9206 2401School of Public Health, University of Texas Health Science Center at Houston, Huston, TX USA
| | - Megan Grove
- grid.267308.80000 0000 9206 2401School of Public Health, University of Texas Health Science Center at Houston, Huston, TX USA
| | - Weihua Guan
- grid.17635.360000000419368657Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN USA
| | - Pfeiffer Liliane
- grid.4567.00000 0004 0483 2525Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany ,grid.4567.00000 0004 0483 2525Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Paula Singmann
- grid.4567.00000 0004 0483 2525Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany ,grid.4567.00000 0004 0483 2525Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Martina Müller-Nurasyid
- grid.4567.00000 0004 0483 2525Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany ,grid.5252.00000 0004 1936 973XIBE, Faculty of Medicine, LMU Munich, Munich, Germany ,grid.410607.4Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Thomas Meitinger
- grid.410607.4Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany ,grid.4567.00000 0004 0483 2525Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany ,grid.6936.a0000000123222966Institute of Human Genetics, Technical University Munich, Munich, Germany
| | - Christian Gieger
- grid.4567.00000 0004 0483 2525Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany ,grid.4567.00000 0004 0483 2525Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Annette Peters
- grid.4567.00000 0004 0483 2525Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany ,grid.410607.4Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Wei Zhao
- grid.214458.e0000000086837370Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI USA
| | - Erin B. Ware
- grid.214458.e0000000086837370Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI USA ,Survey Research Center, Institute for Social Research, Ann Arbor, MI USA
| | - Jennifer A. Smith
- grid.214458.e0000000086837370Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI USA ,Survey Research Center, Institute for Social Research, Ann Arbor, MI USA
| | - Klodian Dhana
- grid.240684.c0000 0001 0705 3621Department of Internal Medicine, Rush University Medical Center, Chicago, IL USA
| | - Joyce van Meurs
- grid.5645.2000000040459992XDepartment of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Andre Uitterlinden
- grid.5645.2000000040459992XDepartment of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Mohammad Arfan Ikram
- grid.5645.2000000040459992XDepartment of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Mohsen Ghanbari
- grid.5645.2000000040459992XDepartment of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Deugi Zhi
- grid.267308.80000 0000 9206 2401School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX USA
| | - Stefan Gustafsson
- grid.8993.b0000 0004 1936 9457Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Lars Lind
- grid.8993.b0000 0004 1936 9457Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Shengxu Li
- grid.265219.b0000 0001 2217 8588Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA USA
| | - Dianjianyi Sun
- grid.265219.b0000 0001 2217 8588Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA USA ,grid.11135.370000 0001 2256 9319Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Tim D. Spector
- grid.13097.3c0000 0001 2322 6764Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King’s College London, London, UK
| | - Yii-der Ida Chen
- grid.239844.00000 0001 0157 6501Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute, and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA USA
| | - Coleen Damcott
- grid.411024.20000 0001 2175 4264Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD USA ,grid.411024.20000 0001 2175 4264Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD USA
| | - Alan R. Shuldiner
- grid.411024.20000 0001 2175 4264Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD USA ,grid.411024.20000 0001 2175 4264Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD USA
| | - Devin M. Absher
- grid.417691.c0000 0004 0408 3720HudsonAlpha Institute for Biotechnology, Huntsville, AL USA
| | - Steve Horvath
- grid.19006.3e0000 0000 9632 6718Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718Department of Biostatistics, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA USA
| | - Philip S. Tsao
- grid.168010.e0000000419368956Department of Medicine, Stanford University School of Medicine, Stanford, CA USA ,grid.280747.e0000 0004 0419 2556VA Palo Alto Healthcare System, Palo Alto, CA USA ,grid.168010.e0000000419368956Stanford Cardiovascular Institute, Stanford University, Stanford, CA USA
| | - Sharon Kardia
- grid.214458.e0000000086837370Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI USA
| | - Bruce M. Psaty
- grid.34477.330000000122986657Cardiovascular Health Research Unit, Departments of Epidemiology, Medicine, and Health Services, University of Washington, Seattle, WA USA ,grid.488833.c0000 0004 0615 7519Kaiser Permanente Washington Health Research Institute, Seattle, WA USA
| | - Nona Sotoodehnia
- grid.34477.330000000122986657Cardiovascular Health Research Unit, Division of Cardiology, University of Washington, Seattle, WA USA
| | - Jordana T. Bell
- grid.13097.3c0000 0001 2322 6764Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King’s College London, London, UK
| | - Erik Ingelsson
- grid.168010.e0000000419368956Department of Medicine, Stanford University School of Medicine, Stanford, CA USA ,grid.8993.b0000 0004 1936 9457Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden ,grid.168010.e0000000419368956Stanford Cardiovascular Institute, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Stanford Diabetes Research Center, Stanford University, Stanford, CA USA
| | - Wei Chen
- grid.265219.b0000 0001 2217 8588Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA USA
| | - Abbas Dehghan
- grid.5645.2000000040459992XDepartment of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands ,grid.7445.20000 0001 2113 8111Department of Biostatistics and Epidemiology, MRC Centre for Environment and Health, School of Public Health, Imperial College, London, UK
| | - Donna K. Arnett
- grid.265892.20000000106344187Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL USA
| | - Melanie Waldenberger
- grid.4567.00000 0004 0483 2525Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany ,grid.4567.00000 0004 0483 2525Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Lifang Hou
- grid.16753.360000 0001 2299 3507Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL USA
| | - Eric A. Whitsel
- grid.410711.20000 0001 1034 1720Department of Epidemiology, University of North Carolina, Chapel Hill, NC USA ,grid.410711.20000 0001 1034 1720Department of Medicine, University of North Carolina, Chapel Hill, NC USA
| | - Andrea Baccarelli
- grid.38142.3c000000041936754XDepartment of Environmental Health Sciences, Harvard T.H. Chan School of Public Health, Boston, MA USA ,grid.21729.3f0000000419368729Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY USA
| | - Daniel Levy
- grid.94365.3d0000 0001 2297 5165Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD USA ,grid.510954.c0000 0004 0444 3861Framingham Heart Study, Framingham, MA USA
| | - Myriam Fornage
- grid.267308.80000 0000 9206 2401The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX USA
| | - Marguerite R. Irvin
- grid.265892.20000000106344187Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL USA
| | - Themistocles L. Assimes
- grid.168010.e0000000419368956Department of Medicine, Stanford University School of Medicine, Stanford, CA USA ,grid.280747.e0000 0004 0419 2556VA Palo Alto Healthcare System, Palo Alto, CA USA ,grid.168010.e0000000419368956Stanford Cardiovascular Institute, Stanford University, Stanford, CA USA
| |
Collapse
|
200
|
Braz CU, Rowan TN, Schnabel RD, Decker JE. Genome-wide association analyses identify genotype-by-environment interactions of growth traits in Simmental cattle. Sci Rep 2021; 11:13335. [PMID: 34172761 PMCID: PMC8233360 DOI: 10.1038/s41598-021-92455-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 06/07/2021] [Indexed: 02/06/2023] Open
Abstract
Understanding genotype-by-environment interactions (G × E) is crucial to understand environmental adaptation in mammals and improve the sustainability of agricultural production. Here, we present an extensive study investigating the interaction of genome-wide SNP markers with a vast assortment of environmental variables and searching for SNPs controlling phenotypic variance (vQTL) using a large beef cattle dataset. We showed that G × E contribute 10.1%, 3.8%, and 2.8% of the phenotypic variance of birth weight, weaning weight, and yearling weight, respectively. G × E genome-wide association analysis (GWAA) detected a large number of G × E loci affecting growth traits, which the traditional GWAA did not detect, showing that functional loci may have non-additive genetic effects regardless of differences in genotypic means. Further, variance-heterogeneity GWAA detected loci enriched with G × E effects without requiring prior knowledge of the interacting environmental factors. Functional annotation and pathway analysis of G × E genes revealed biological mechanisms by which cattle respond to changes in their environment, such as neurotransmitter activity, hypoxia-induced processes, keratinization, hormone, thermogenic and immune pathways. We unraveled the relevance and complexity of the genetic basis of G × E underlying growth traits, providing new insights into how different environmental conditions interact with specific genes influencing adaptation and productivity in beef cattle and potentially across mammals.
Collapse
Affiliation(s)
- Camila U Braz
- Division of Animal Sciences, University of Missouri, Columbia, MO, 65211, USA
| | - Troy N Rowan
- Division of Animal Sciences, University of Missouri, Columbia, MO, 65211, USA
- Genetics Area Program, University of Missouri, Columbia, MO, 65211, USA
| | - Robert D Schnabel
- Division of Animal Sciences, University of Missouri, Columbia, MO, 65211, USA
- Genetics Area Program, University of Missouri, Columbia, MO, 65211, USA
- Informatics Institute, University of Missouri, Columbia, MO, 65211, USA
| | - Jared E Decker
- Division of Animal Sciences, University of Missouri, Columbia, MO, 65211, USA.
- Genetics Area Program, University of Missouri, Columbia, MO, 65211, USA.
- Informatics Institute, University of Missouri, Columbia, MO, 65211, USA.
| |
Collapse
|