1
|
Arzik Y, Kizilaslan M, Behrem S, Piel LMW, White SN, Çınar MU. Exploring Genetic Factors Associated with Moniezia spp. Tapeworm Resistance in Central Anatolian Merino Sheep via GWAS Approach. Animals (Basel) 2025; 15:812. [PMID: 40150341 PMCID: PMC11939720 DOI: 10.3390/ani15060812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/19/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Gastrointestinal parasite (GIP) infections pose significant challenges in pasture-based sheep farming, leading to economic losses and welfare concerns. This study aimed to uncover the genetic basis of resistance to Moniezia spp. infections in Central Anatolian Merino (CAM) sheep. Genome-Wide Association Analysis (GWAS) was conducted between Moniezia spp. egg burden and genomic data from 226 CAM lambs. Thirteen significant Single-Nucleotide Polymorphisms (SNPs) were identified, with five surpassing the genome-wide threshold and eight exceeding the chromosome-wide threshold. Functional annotation revealed associations with genes involved in immune function, notably CD79A and MAP3K7. CD79A, integral to B-cell activation and antibody production, plays a key role in the immune response against parasitic infections. Its interaction with helminth-derived proteins modulates B-cell function, highlighting its potential as a therapeutic target. MAP3K7, a central regulator of immune signaling pathways, modulates host responses to helminth infections by influencing NF-κB activity. Additionally, it regulates macrophage function in bacterial infections, showcasing its versatility in mediating immune responses against diverse pathogens. From a practical perspective, the findings of the current research underscore the potential of integrating genomic information into breeding programs to bolster disease resilience in livestock populations for sustainable production purposes. However, further research is needed to elucidate the functional significance of identified SNPs and associated genes. This study underscores the potential of genomic approaches in combating parasitic diseases and promoting sustainable agriculture in sheep production systems.
Collapse
Affiliation(s)
- Yunus Arzik
- Department of Animal Science, Faculty of Veterinary Medicine, Aksaray University, 68000 Aksaray, Türkiye; (Y.A.); (S.B.)
| | - Mehmet Kizilaslan
- Department of Animal Science, Faculty of Agriculture, Erciyes University, 38039 Kayseri, Türkiye;
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Sedat Behrem
- Department of Animal Science, Faculty of Veterinary Medicine, Aksaray University, 68000 Aksaray, Türkiye; (Y.A.); (S.B.)
| | - Lindsay M. W. Piel
- USDA-ARS Animal Disease Research 3003 ADBF, Washington State University, Pullman, WA 99164, USA;
| | - Stephen N. White
- USDA-ARS Poultry Microbiological Safety and Processing Research, US National Poultry Research Center, Athens, GA 30605, USA;
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Mehmet Ulaş Çınar
- Department of Animal Science, Faculty of Agriculture, Erciyes University, 38039 Kayseri, Türkiye;
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
2
|
Young WJ, van der Most PJ, Bartz TM, Bos MM, Biino G, Duong T, Foco L, Lominchar JT, Müller‐Nurasyid M, Nardone GG, Pecori A, Ramirez J, Repetto L, Schramm K, Shen X, van Duijvenboden S, van Heemst D, Weiss S, Yao J, Benjamins J, Alonso A, Spedicati B, Biggs ML, Brody JA, Dörr M, Fuchsberger C, Gögele M, Guo X, Ikram MA, Jukema JW, Kääb S, Kanters JK, Lin HJ, Linneberg A, Nauck M, Nolte IM, Pianigiani G, Santin A, Soliman EZ, Tesolin P, Vaccargiu S, Waldenberger M, van der Harst P, Verweij N, Arking DE, Concas MP, De Grandi A, Girotto G, Grarup N, Kavousi M, Mook‐Kanamori DO, Navarro P, Orini M, Padmanabhan S, Pattaro C, Peters A, Pirastu M, Pramstaller PP, Heckbert SR, Sinner M, Snieder H, Völker U, Wilson JF, Gauderman WJ, Lambiase PD, Sotoodehnia N, Tinker A, Warren HR, Noordam R, Munroe PB. Genome-Wide Interaction Analyses of Serum Calcium on Ventricular Repolarization Time in 125 393 Participants. J Am Heart Assoc 2024; 13:e034760. [PMID: 39206732 PMCID: PMC11646519 DOI: 10.1161/jaha.123.034760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Ventricular repolarization time (ECG QT and JT intervals) is associated with malignant arrhythmia. Genome-wide association studies have identified 230 independent loci for QT and JT; however, 50% of their heritability remains unexplained. Previous work supports a causal effect of lower serum calcium concentrations on longer ventricular repolarization time. We hypothesized calcium interactions with QT and JT variant associations could explain a proportion of the missing heritability. METHODS AND RESULTS We performed genome-wide calcium interaction analyses for QT and JT intervals. Participants were stratified by their calcium level relative to the study distribution (top or bottom 20%). We performed a 2-stage analysis (genome-wide discovery [N=62 532] and replication [N=59 861] of lead variants) and a single-stage genome-wide meta-analysis (N=122 393, [European ancestry N=117 581, African ancestry N=4812]). We also calculated 2-degrees of freedom joint main and interaction and 1-degree of freedom interaction P values. In 2-stage and single-stage analyses, 50 and 98 independent loci, respectively, were associated with either QT or JT intervals (2-degrees of freedom joint main and interaction P value <5×10-8). No lead variant had a significant interaction result after correcting for multiple testing and sensitivity analyses provided similar findings. Two loci in the single-stage meta-analysis were not reported previously (SPPL2B and RFX6). CONCLUSIONS We have found limited support for an interaction effect of serum calcium on QT and JT variant associations despite sample sizes with suitable power to detect relevant effects. Therefore, such effects are unlikely to explain a meaningful proportion of the heritability of QT and JT, and factors including rare variation and other environmental interactions need to be considered.
Collapse
Affiliation(s)
- William J. Young
- Clinical Pharmacology and Precision MedicineWilliam Harvey Research Institute, Queen Mary University of LondonUnited Kingdom
- Barts Heart CentreSt Bartholomew’s Hospital, Barts Health NHS TrustLondonUnited Kingdom
| | - Peter J. van der Most
- Department of EpidemiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Traci M. Bartz
- Cardiovascular Health Research Unit, Department of Biostatistics and MedicineUniversity of WashingtonSeattleWAUSA
| | - Maxime M. Bos
- Department of EpidemiologyErasmus MC University Medical CenterRotterdamNetherlands
| | - Ginevra Biino
- Institute of Molecular Genetics, National Research Council of ItalyPaviaItaly
| | - ThuyVy Duong
- Department of Genetic MedicineMcKusick‐Nathans Institute, Johns Hopkins University School of MedicineBaltimoreMDUSA
| | - Luisa Foco
- Eurac ResearchInstitute for Biomedicine (Affiliated with the University of Lübeck)BolzanoItaly
| | - Jesus T. Lominchar
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical SciencesUniversity of CopenhagenDenmark
| | - Martina Müller‐Nurasyid
- German Research Center for Environmental HealthInstitute of Genetic Epidemiology, Helmholtz Zentrum MünchenNeuherbergGermany
- IBE, Faculty of Medicine, LMU MunichMunichGermany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg UniversityMainzGermany
| | | | - Alessandro Pecori
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”TriesteItaly
| | - Julia Ramirez
- Clinical Pharmacology and Precision MedicineWilliam Harvey Research Institute, Queen Mary University of LondonUnited Kingdom
- Aragon Institute of Engineering Research, University of ZaragozaSpain
- Centro de Investigación Biomédica en Red—Bioingeniería, Biomateriales y NanomedicinaZaragozaSpain
| | - Linda Repetto
- Centre for Global Health ResearchUsher Institute, University of EdinburghScotland
| | - Katharina Schramm
- German Research Center for Environmental HealthInstitute of Genetic Epidemiology, Helmholtz Zentrum MünchenNeuherbergGermany
- IBE, Faculty of Medicine, LMU MunichMunichGermany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg UniversityMainzGermany
| | - Xia Shen
- Centre for Global Health ResearchUsher Institute, University of EdinburghScotland
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
- Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan UniversityGuangzhouChina
| | - Stefan van Duijvenboden
- Clinical Pharmacology and Precision MedicineWilliam Harvey Research Institute, Queen Mary University of LondonUnited Kingdom
- Institute of Cardiovascular Sciences, University of College LondonLondonUnited Kingdom
- Nuffield Department of Population HealthUniversity of OxfordUnited Kingdom
| | - Diana van Heemst
- Department of Internal Medicine, Section of Gerontology and GeriatricsLeiden University Medical CenterLeidenThe Netherlands
| | - Stefan Weiss
- DZHK (German Centre for Cardiovascular Research), partner site GreifswaldGreifswaldGermany
- Interfaculty Institute for Genetics and Functional Genomics; Department of Functional GenomicsUniversity Medicine GreifswaldGreifswaldGermany
| | - Jie Yao
- Department of PediatricsThe Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor‐UCLA Medical CenterTorranceCAUSA
| | - Jan‐Walter Benjamins
- Department of CardiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Alvaro Alonso
- Department of EpidemiologyRollins School of Public Health, Emory UniversityAtlantaGAUSA
| | - Beatrice Spedicati
- Department of Medicine, Surgery and Health SciencesUniversity of TriesteItaly
| | - Mary L. Biggs
- Cardiovascular Health Research Unit, Department of MedicineUniversity of WashingtonSeattleWAUSA
- Department of BiostatisticsUniversity of WashingtonSeattleWAUSA
| | - Jennifer A. Brody
- Cardiovascular Health Research Unit, Department of MedicineUniversity of WashingtonSeattleWAUSA
| | - Marcus Dörr
- DZHK (German Centre for Cardiovascular Research), partner site GreifswaldGreifswaldGermany
- Department of Internal Medicine B—Cardiology, Pneumology, Infectious Diseases, Intensive Care MedicineUniversity Medicine GreifswaldGreifswaldGermany
| | - Christian Fuchsberger
- Eurac ResearchInstitute for Biomedicine (Affiliated with the University of Lübeck)BolzanoItaly
- Department of BiostatisticsUniversity of Michigan School of Public HealthAnn ArborMIUSA
- Center for Statistical GeneticsUniversity of Michigan School of Public HealthAnn ArborMIUSA
| | - Martin Gögele
- Eurac ResearchInstitute for Biomedicine (Affiliated with the University of Lübeck)BolzanoItaly
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences/The Lundquist Institute at Harbor‐UCLA Medical CenterTorranceCAUSA
- Department of PediatricsDavid Geffen School of Medicine at UCLALos AngelesCAUSA
| | - M. Arfan Ikram
- Department of EpidemiologyErasmus MC University Medical CenterRotterdamNetherlands
| | - J. Wouter Jukema
- Department of CardiologyLeiden University Medical CenterLeidenThe Netherlands
- Netherlands Heart InstituteUtrechtThe Netherlands
| | - Stefan Kääb
- Department of CardiologyUniversity Hospital, LMU MunichMunichGermany
- DZHK (German Centre for Cardiovascular Research), partner site: Munich Heart AllianceMunichGermany
| | - Jørgen K. Kanters
- Laboratory of Experimental Cardiology, Department of Biomedical SciencesUniversity of CopenhagenDenmark
| | | | - Henry J. Lin
- Department of PediatricsThe Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor‐UCLA Medical CenterTorranceCAUSA
- Department of PediatricsDavid Geffen School of Medicine at UCLALos AngelesCAUSA
- Department of Pediatrics/Harbor‐UCLA Medical CenterTorranceCAUSA
| | - Allan Linneberg
- Center for Clinical Research and PreventionBispebjerg and Frederiksberg Hospital, The Capital RegionCopenhagenDenmark
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenDenmark
| | - Matthias Nauck
- DZHK (German Centre for Cardiovascular Research), partner site GreifswaldGreifswaldGermany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine GreifswaldGreifswaldGermany
| | - Ilja M. Nolte
- Department of EpidemiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Giulia Pianigiani
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”TriesteItaly
| | - Aurora Santin
- Department of Medicine, Surgery and Health SciencesUniversity of TriesteItaly
| | - Elsayed Z. Soliman
- Epidemiological Cardiology Research Center (EPICARE)Wake Forest School of MedicineWinston SalemUSA
| | - Paola Tesolin
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”TriesteItaly
| | - Simona Vaccargiu
- Institute for Genetic and Biomedical Research, National Research Council of ItalyCagliariItaly
| | - Melanie Waldenberger
- DZHK (German Centre for Cardiovascular Research), partner site: Munich Heart AllianceMunichGermany
- Research Unit Molecular EpidemiologyInstitute of Epidemiology, Helmholtz Zentrum München—German Research Center for Environmental HealthNeuherbergGermany
| | - Pim van der Harst
- Department of CardiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
- Department of Cardiology, Heart and Lung DivisionUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Niek Verweij
- Department of CardiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Dan E. Arking
- Department of Genetic MedicineMcKusick‐Nathans Institute, Johns Hopkins University School of MedicineBaltimoreMDUSA
| | - Maria Pina Concas
- Institute for Maternal and Child Health—IRCCS “Burlo Garofolo”TriesteItaly
| | - Alessandro De Grandi
- Eurac ResearchInstitute for Biomedicine (Affiliated with the University of Lübeck)BolzanoItaly
| | - Giorgia Girotto
- Department of Medicine, Surgery and Health SciencesUniversity of TriesteItaly
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical SciencesUniversity of CopenhagenDenmark
| | - Maryam Kavousi
- Department of EpidemiologyErasmus MC University Medical CenterRotterdamNetherlands
| | - Dennis O. Mook‐Kanamori
- Department of Clinical EpidemiologyLeiden University Medical CenterLeidenThe Netherlands
- Department of Public Health and Primary CareLeiden University Medical CenterLeidenThe Netherlands
| | - Pau Navarro
- MRC Human Genetics UnitInstitute of Genetics and Cancer, University of EdinburghScotland
| | - Michele Orini
- Barts Heart CentreSt Bartholomew’s Hospital, Barts Health NHS TrustLondonUnited Kingdom
- Institute of Cardiovascular Sciences, University of College LondonLondonUnited Kingdom
| | | | - Cristian Pattaro
- Eurac ResearchInstitute for Biomedicine (Affiliated with the University of Lübeck)BolzanoItaly
| | - Annette Peters
- German Research Center for Environmental HealthInstitute of Genetic Epidemiology, Helmholtz Zentrum MünchenNeuherbergGermany
- IBE, Faculty of Medicine, LMU MunichMunichGermany
- DZHK (German Centre for Cardiovascular Research), partner site: Munich Heart AllianceMunichGermany
| | - Mario Pirastu
- Institute for Genetic and Biomedical Research, Sassari Unit, National Research Council of ItalySassariItaly
| | - Peter P. Pramstaller
- Eurac ResearchInstitute for Biomedicine (Affiliated with the University of Lübeck)BolzanoItaly
- Department of NeurologyUniversity of LübeckGermany
| | - Susan R. Heckbert
- Cardiovascular Health Research Unit, Department of MedicineUniversity of WashingtonSeattleWAUSA
- Department of EpidemiologyUniversity of WashingtonSeattleWAUSA
| | - Mortiz Sinner
- Department of CardiologyUniversity Hospital, LMU MunichMunichGermany
- DZHK (German Centre for Cardiovascular Research), partner site: Munich Heart AllianceMunichGermany
| | - Harold Snieder
- Department of EpidemiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Uwe Völker
- DZHK (German Centre for Cardiovascular Research), partner site GreifswaldGreifswaldGermany
- Interfaculty Institute for Genetics and Functional Genomics; Department of Functional GenomicsUniversity Medicine GreifswaldGreifswaldGermany
| | - James F. Wilson
- Centre for Global Health ResearchUsher Institute, University of EdinburghScotland
- MRC Human Genetics UnitInstitute of Genetics and Cancer, University of EdinburghScotland
| | - W. James Gauderman
- Department of population and public health sciencesUniversity of Southern CaliforniaLos AngelesCAUSA
| | - Pier D. Lambiase
- Barts Heart CentreSt Bartholomew’s Hospital, Barts Health NHS TrustLondonUnited Kingdom
- Institute of Cardiovascular Sciences, University of College LondonLondonUnited Kingdom
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Division of Cardiology, Department of MedicineUniversity of WashingtonSeattleWAUSA
| | - Andrew Tinker
- Clinical Pharmacology and Precision MedicineWilliam Harvey Research Institute, Queen Mary University of LondonUnited Kingdom
- NIHR Barts Biomedical Research CentreBarts and The London Faculty of Medicine and Dentistry, Queen Mary University of LondonUnited Kingdom
| | - Helen R. Warren
- Clinical Pharmacology and Precision MedicineWilliam Harvey Research Institute, Queen Mary University of LondonUnited Kingdom
- NIHR Barts Biomedical Research CentreBarts and The London Faculty of Medicine and Dentistry, Queen Mary University of LondonUnited Kingdom
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and GeriatricsLeiden University Medical CenterLeidenThe Netherlands
| | - Patricia B. Munroe
- Clinical Pharmacology and Precision MedicineWilliam Harvey Research Institute, Queen Mary University of LondonUnited Kingdom
- NIHR Barts Biomedical Research CentreBarts and The London Faculty of Medicine and Dentistry, Queen Mary University of LondonUnited Kingdom
| |
Collapse
|
3
|
Nemkov T, Key A, Stephenson D, Earley EJ, Keele GR, Hay A, Amireault P, Casimir M, Dussiot M, Dzieciatkowska M, Reisz JA, Deng X, Stone M, Kleinman S, Spitalnik SL, Hansen KC, Norris PJ, Churchill GA, Busch MP, Roubinian N, Page GP, Zimring JC, Arduini A, D’Alessandro A. Genetic regulation of carnitine metabolism controls lipid damage repair and aging RBC hemolysis in vivo and in vitro. Blood 2024; 143:2517-2533. [PMID: 38513237 PMCID: PMC11208298 DOI: 10.1182/blood.2024023983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/22/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024] Open
Abstract
ABSTRACT Recent large-scale multiomics studies suggest that genetic factors influence the chemical individuality of donated blood. To examine this concept, we performed metabolomics analyses of 643 blood units from volunteers who donated units of packed red blood cells (RBCs) on 2 separate occasions. These analyses identified carnitine metabolism as the most reproducible pathway across multiple donations from the same donor. We also measured l-carnitine and acyl-carnitines in 13 091 packed RBC units from donors in the Recipient Epidemiology and Donor Evaluation study. Genome-wide association studies against 879 000 polymorphisms identified critical genetic factors contributing to interdonor heterogeneity in end-of-storage carnitine levels, including common nonsynonymous polymorphisms in genes encoding carnitine transporters (SLC22A16, SLC22A5, and SLC16A9); carnitine synthesis (FLVCR1 and MTDH) and metabolism (CPT1A, CPT2, CRAT, and ACSS2), and carnitine-dependent repair of lipids oxidized by ALOX5. Significant associations between genetic polymorphisms on SLC22 transporters and carnitine pools in stored RBCs were validated in 525 Diversity Outbred mice. Donors carrying 2 alleles of the rs12210538 SLC22A16 single-nucleotide polymorphism exhibited the lowest l-carnitine levels, significant elevations of in vitro hemolysis, and the highest degree of vesiculation, accompanied by increases in lipid peroxidation markers. Separation of RBCs by age, via in vivo biotinylation in mice, and Percoll density gradients of human RBCs, showed age-dependent depletions of l-carnitine and acyl-carnitine pools, accompanied by progressive failure of the reacylation process after chemically induced membrane lipid damage. Supplementation of stored murine RBCs with l-carnitine boosted posttransfusion recovery, suggesting this could represent a viable strategy to improve RBC storage quality.
Collapse
Affiliation(s)
- Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO
- Omix Technologies Inc, Aurora, CO
| | - Alicia Key
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO
| | - Daniel Stephenson
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO
| | - Eric J. Earley
- Genomics and Translational Research Center, RTI International, Research Triangle Park, NC
| | - Gregory R. Keele
- Genomics and Translational Research Center, RTI International, Research Triangle Park, NC
- The Jackson Laboratory, Bar Harbor, ME
| | - Ariel Hay
- Department of Pathology, University of Virginia, Charlottesville, VA
| | - Pascal Amireault
- Université Paris Cité et Université des Antilles, INSERM, Biologie Intégrée du Globule Rouge, Paris, France
- Université Paris Cité, Institut Imagine, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM, Paris, France
| | - Madeleine Casimir
- Université Paris Cité et Université des Antilles, INSERM, Biologie Intégrée du Globule Rouge, Paris, France
- Université Paris Cité, Institut Imagine, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM, Paris, France
| | - Michaël Dussiot
- Université Paris Cité et Université des Antilles, INSERM, Biologie Intégrée du Globule Rouge, Paris, France
- Université Paris Cité, Institut Imagine, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM, Paris, France
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO
| | - Julie A. Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO
| | - Xutao Deng
- Vitalant Research Institute, San Francisco, CA
| | - Mars Stone
- Vitalant Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
| | - Steve Kleinman
- The University of British Columbia, Victoria, BC, Canada
| | | | - Kirk C. Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO
| | - Philip J. Norris
- Vitalant Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
| | | | - Michael P. Busch
- Vitalant Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
| | - Nareg Roubinian
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
- Kaiser Permanente Northern California Division of Research, Oakland, CA
| | - Grier P. Page
- Genomics and Translational Research Center, RTI International, Research Triangle Park, NC
| | - James C. Zimring
- Department of Pathology, University of Virginia, Charlottesville, VA
| | - Arduino Arduini
- Department of Research and Development, CoreQuest Sagl, Lugano, Switzerland
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO
- Omix Technologies Inc, Aurora, CO
| |
Collapse
|
4
|
Nemkov T, Stephenson D, Earley EJ, Keele GR, Hay A, Key A, Haiman Z, Erickson C, Dzieciatkowska M, Reisz JA, Moore A, Stone M, Deng X, Kleinman S, Spitalnik SL, Hod EA, Hudson KE, Hansen KC, Palsson BO, Churchill GA, Roubinian N, Norris PJ, Busch MP, Zimring JC, Page GP, D'Alessandro A. Biological and Genetic Determinants of Glycolysis: Phosphofructokinase Isoforms Boost Energy Status of Stored Red Blood Cells and Transfusion Outcomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.11.557250. [PMID: 38260479 PMCID: PMC10802247 DOI: 10.1101/2023.09.11.557250] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Mature red blood cells (RBCs) lack mitochondria, and thus exclusively rely on glycolysis to generate adenosine triphosphate (ATP) during aging in vivo or storage in the blood bank. Here we leveraged 13,029 volunteers from the Recipient Epidemiology and Donor Evaluation Study to identify an association between end-of-storage levels of glycolytic metabolites and donor age, sex, and ancestry-specific genetic polymorphisms in regions encoding phosphofructokinase 1, platelet (detected in mature RBCs), hexokinase 1, ADP-ribosyl cyclase 1 and 2 (CD38/BST1). Gene-metabolite associations were validated in fresh and stored RBCs from 525 Diversity Outbred mice, and via multi-omics characterization of 1,929 samples from 643 human RBC units during storage. ATP and hypoxanthine levels - and the genetic traits linked to them - were associated with hemolysis in vitro and in vivo, both in healthy autologous transfusion recipients and in 5,816 critically ill patients receiving heterologous transfusions, suggesting their potential as markers to improve transfusion outcomes. eTOC and Highlights Highlights Blood donor age and sex affect glycolysis in stored RBCs from 13,029 volunteers;Ancestry, genetic polymorphisms in PFKP, HK1, CD38/BST1 influence RBC glycolysis;Modeled PFKP effects relate to preventing loss of the total AXP pool in stored RBCs;ATP and hypoxanthine are biomarkers of hemolysis in vitro and in vivo.
Collapse
|
5
|
D'Alessandro A, Keele GR, Hay A, Nemkov T, Earley EJ, Stephenson D, Vincent M, Deng X, Stone M, Dzieciatkowska M, Hansen KC, Kleinman S, Spitalnik SL, Roubinian NH, Norris PJ, Busch MP, Page GP, Stockwell BR, Churchill GA, Zimring JC. Ferroptosis regulates hemolysis in stored murine and human red blood cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598512. [PMID: 38915523 PMCID: PMC11195277 DOI: 10.1101/2024.06.11.598512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Red blood cell (RBC) metabolism regulates hemolysis during aging in vivo and in the blood bank. Here, we leveraged a diversity outbred mouse population to map the genetic drivers of fresh/stored RBC metabolism and extravascular hemolysis upon storage and transfusion in 350 mice. We identify the ferrireductase Steap3 as a critical regulator of a ferroptosis-like process of lipid peroxidation. Steap3 polymorphisms were associated with RBC iron content, in vitro hemolysis, and in vivo extravascular hemolysis both in mice and 13,091 blood donors from the Recipient Epidemiology and Donor evaluation Study. Using metabolite Quantitative Trait Loci analyses, we identified a network of gene products (FADS1/2, EPHX2 and LPCAT3) - enriched in donors of African descent - associated with oxylipin metabolism in stored human RBCs and related to Steap3 or its transcriptional regulator, the tumor protein TP53. Genetic variants were associated with lower in vivo hemolysis in thousands of single-unit transfusion recipients. Highlights Steap3 regulates lipid peroxidation and extravascular hemolysis in 350 diversity outbred miceSteap3 SNPs are linked to RBC iron, hemolysis, vesiculation in 13,091 blood donorsmQTL analyses of oxylipins identified ferroptosis-related gene products FADS1/2, EPHX2, LPCAT3Ferroptosis markers are linked to hemoglobin increments in transfusion recipients. Graphical abstract
Collapse
|
6
|
Verhoef E, Allegrini AG, Jansen PR, Lange K, Wang CA, Morgan AT, Ahluwalia TS, Symeonides C, Eising E, Franken MC, Hypponen E, Mansell T, Olislagers M, Omerovic E, Rimfeld K, Schlag F, Selzam S, Shapland CY, Tiemeier H, Whitehouse AJO, Saffery R, Bønnelykke K, Reilly S, Pennell CE, Wake M, Cecil CAM, Plomin R, Fisher SE, St Pourcain B. Genome-Wide Analyses of Vocabulary Size in Infancy and Toddlerhood: Associations With Attention-Deficit/Hyperactivity Disorder, Literacy, and Cognition-Related Traits. Biol Psychiatry 2024; 95:859-869. [PMID: 38070845 DOI: 10.1016/j.biopsych.2023.11.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 11/26/2023] [Accepted: 11/29/2023] [Indexed: 02/17/2024]
Abstract
BACKGROUND The number of words children produce (expressive vocabulary) and understand (receptive vocabulary) changes rapidly during early development, partially due to genetic factors. Here, we performed a meta-genome-wide association study of vocabulary acquisition and investigated polygenic overlap with literacy, cognition, developmental phenotypes, and neurodevelopmental conditions, including attention-deficit/hyperactivity disorder (ADHD). METHODS We studied 37,913 parent-reported vocabulary size measures (English, Dutch, Danish) for 17,298 children of European descent. Meta-analyses were performed for early-phase expressive (infancy, 15-18 months), late-phase expressive (toddlerhood, 24-38 months), and late-phase receptive (toddlerhood, 24-38 months) vocabulary. Subsequently, we estimated single nucleotide polymorphism-based heritability (SNP-h2) and genetic correlations (rg) and modeled underlying factor structures with multivariate models. RESULTS Early-life vocabulary size was modestly heritable (SNP-h2 = 0.08-0.24). Genetic overlap between infant expressive and toddler receptive vocabulary was negligible (rg = 0.07), although each measure was moderately related to toddler expressive vocabulary (rg = 0.69 and rg = 0.67, respectively), suggesting a multifactorial genetic architecture. Both infant and toddler expressive vocabulary were genetically linked to literacy (e.g., spelling: rg = 0.58 and rg = 0.79, respectively), underlining genetic similarity. However, a genetic association of early-life vocabulary with educational attainment and intelligence emerged only during toddlerhood (e.g., receptive vocabulary and intelligence: rg = 0.36). Increased ADHD risk was genetically associated with larger infant expressive vocabulary (rg = 0.23). Multivariate genetic models in the ALSPAC (Avon Longitudinal Study of Parents and Children) cohort confirmed this finding for ADHD symptoms (e.g., at age 13; rg = 0.54) but showed that the association effect reversed for toddler receptive vocabulary (rg = -0.74), highlighting developmental heterogeneity. CONCLUSIONS The genetic architecture of early-life vocabulary changes during development, shaping polygenic association patterns with later-life ADHD, literacy, and cognition-related traits.
Collapse
Affiliation(s)
- Ellen Verhoef
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands.
| | - Andrea G Allegrini
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Philip R Jansen
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit, Amsterdam, the Netherlands; Section Clinical Genetics, Department Human Genetics, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Katherine Lange
- Murdoch Children's Research Institute, Parkville, Victoria, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Carol A Wang
- School of Medicine and Public Health, The University of Newcastle, Newcastle, New South Wales, Australia; Mothers and Babies Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Angela T Morgan
- Murdoch Children's Research Institute, Parkville, Victoria, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia; Department of Audiology and Speech Pathology, University of Melbourne, Parkville, Victoria, Australia; Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Tarunveer S Ahluwalia
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Herlev, Denmark; Bioinformatics Center, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Christos Symeonides
- Murdoch Children's Research Institute, Parkville, Victoria, Australia; Royal Children's Hospital, Melbourne, Victoria, Australia; Minderoo Foundation, Perth, Western Australia, Australia
| | - Else Eising
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
| | - Marie-Christine Franken
- Erasmus University Medical Center, Sophia Children's Hospital, Department of Otorhinolaryngology and Head and Neck Surgery, Rotterdam, the Netherlands
| | - Elina Hypponen
- Australian Centre for Precision Health, Unit of Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia; South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Toby Mansell
- Murdoch Children's Research Institute, Parkville, Victoria, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Mitchell Olislagers
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands; Department of Urology, Erasmus University Medical Center, Erasmus University Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - Emina Omerovic
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Kaili Rimfeld
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Psychology, Royal Holloway University of London, London, UK
| | - Fenja Schlag
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
| | - Saskia Selzam
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Chin Yang Shapland
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Population Health Sciences, University of Bristol, Bristol, UK
| | - Henning Tiemeier
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, Rotterdam, the Netherlands; Harvard, T.H. Chan School of Public Health, Boston, Massachusetts
| | - Andrew J O Whitehouse
- Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Richard Saffery
- Murdoch Children's Research Institute, Parkville, Victoria, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia; Chongqing Medical University, Chongqing, China
| | - Klaus Bønnelykke
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Sheena Reilly
- Murdoch Children's Research Institute, Parkville, Victoria, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia; Menzies Health Institute Queensland, Griffith University, Brisbane, Queensland, Australia
| | - Craig E Pennell
- School of Medicine and Public Health, The University of Newcastle, Newcastle, New South Wales, Australia; Mothers and Babies Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia; Maternity and Gynaecology John Hunter Hospital, Newcastle, New South Wales, Australia
| | - Melissa Wake
- Murdoch Children's Research Institute, Parkville, Victoria, Australia; Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia; Liggins Institute, The University of Auckland, Grafton, New Zealand
| | - Charlotte A M Cecil
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands; Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Robert Plomin
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands
| | - Beate St Pourcain
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands; Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK; Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands.
| |
Collapse
|
7
|
Nemkov T, Stephenson D, Erickson C, Dzieciatkowska M, Key A, Moore A, Earley EJ, Page GP, Lacroix IS, Stone M, Deng X, Raife T, Kleinman S, Zimring JC, Roubinian N, Hansen KC, Busch MP, Norris PJ, D’Alessandro A. Regulation of kynurenine metabolism by blood donor genetics and biology impacts red cell hemolysis in vitro and in vivo. Blood 2024; 143:456-472. [PMID: 37976448 PMCID: PMC10862365 DOI: 10.1182/blood.2023022052] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/26/2023] [Accepted: 11/04/2023] [Indexed: 11/19/2023] Open
Abstract
ABSTRACT In the field of transfusion medicine, the clinical relevance of the metabolic markers of the red blood cell (RBC) storage lesion is incompletely understood. Here, we performed metabolomics of RBC units from 643 donors enrolled in the Recipient Epidemiology and Donor Evaluation Study, REDS RBC Omics. These units were tested on storage days 10, 23, and 42 for a total of 1929 samples and also characterized for end-of-storage hemolytic propensity after oxidative and osmotic insults. Our results indicate that the metabolic markers of the storage lesion poorly correlated with hemolytic propensity. In contrast, kynurenine was not affected by storage duration and was identified as the top predictor of osmotic fragility. RBC kynurenine levels were affected by donor age and body mass index and were reproducible within the same donor across multiple donations from 2 to 12 months apart. To delve into the genetic underpinnings of kynurenine levels in stored RBCs, we thus tested kynurenine levels in stored RBCs on day 42 from 13 091 donors from the REDS RBC Omics study, a population that was also genotyped for 879 000 single nucleotide polymorphisms. Through a metabolite quantitative trait loci analysis, we identified polymorphisms in SLC7A5, ATXN2, and a series of rate-limiting enzymes (eg, kynurenine monooxygenase, indoleamine 2,3-dioxygenase, and tryptophan dioxygenase) in the kynurenine pathway as critical factors affecting RBC kynurenine levels. By interrogating a donor-recipient linkage vein-to-vein database, we then report that SLC7A5 polymorphisms are also associated with changes in hemoglobin and bilirubin levels, suggestive of in vivo hemolysis in 4470 individuals who were critically ill and receiving single-unit transfusions.
Collapse
Affiliation(s)
- Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver-Anschutz Medical Campus, Aurora, CO
- Omix Technologies Inc, Aurora, CO
| | - Daniel Stephenson
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver-Anschutz Medical Campus, Aurora, CO
| | - Christopher Erickson
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver-Anschutz Medical Campus, Aurora, CO
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver-Anschutz Medical Campus, Aurora, CO
| | - Alicia Key
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver-Anschutz Medical Campus, Aurora, CO
| | - Amy Moore
- Research Triangle Institute International, Atlanta, GA
| | | | - Grier P. Page
- Research Triangle Institute International, Atlanta, GA
| | - Ian S. Lacroix
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver-Anschutz Medical Campus, Aurora, CO
| | - Mars Stone
- Vitalant Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
| | - Xutao Deng
- Vitalant Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
| | - Thomas Raife
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Steven Kleinman
- Department of Pathology, University of British Columbia, Victoria, BC, Canada
| | - James C. Zimring
- Department of Pathology, University of Virginia, Charlottesville, VA
| | - Nareg Roubinian
- Vitalant Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
- Kaiser Permanente Northern California Division of Research, Oakland, CA
| | - Kirk C. Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver-Anschutz Medical Campus, Aurora, CO
| | - Michael P. Busch
- Vitalant Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
| | - Philip J. Norris
- Vitalant Research Institute, San Francisco, CA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver-Anschutz Medical Campus, Aurora, CO
- Omix Technologies Inc, Aurora, CO
| |
Collapse
|
8
|
Nakamura R, Tohnai G, Nakatochi M, Atsuta N, Watanabe H, Ito D, Katsuno M, Hirakawa A, Izumi Y, Morita M, Hirayama T, Kano O, Kanai K, Hattori N, Taniguchi A, Suzuki N, Aoki M, Iwata I, Yabe I, Shibuya K, Kuwabara S, Oda M, Hashimoto R, Aiba I, Ishihara T, Onodera O, Yamashita T, Abe K, Mizoguchi K, Shimizu T, Ikeda Y, Yokota T, Hasegawa K, Tanaka F, Nakashima K, Kaji R, Niwa JI, Doyu M, Terao C, Ikegawa S, Fujimori K, Nakamura S, Ozawa F, Morimoto S, Onodera K, Ito T, Okada Y, Okano H, Sobue G. Genetic factors affecting survival in Japanese patients with sporadic amyotrophic lateral sclerosis: a genome-wide association study and verification in iPSC-derived motor neurons from patients. J Neurol Neurosurg Psychiatry 2023; 94:816-824. [PMID: 37142397 DOI: 10.1136/jnnp-2022-330851] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/18/2023] [Indexed: 05/06/2023]
Abstract
BACKGROUND Several genetic factors are associated with the pathogenesis of sporadic amyotrophic lateral sclerosis (ALS) and its phenotypes, such as disease progression. Here, in this study, we aimed to identify the genes that affect the survival of patients with sporadic ALS. METHODS We enrolled 1076 Japanese patients with sporadic ALS with imputed genotype data of 7 908 526 variants. We used Cox proportional hazards regression analysis with an additive model adjusted for sex, age at onset and the first two principal components calculated from genotyped data to conduct a genome-wide association study. We further analysed messenger RNA (mRNA) and phenotype expression in motor neurons derived from induced pluripotent stem cells (iPSC-MNs) of patients with ALS. RESULTS Three novel loci were significantly associated with the survival of patients with sporadic ALS-FGF1 at 5q31.3 (rs11738209, HR=2.36 (95% CI, 1.77 to 3.15), p=4.85×10-9), THSD7A at 7p21.3 (rs2354952, 1.38 (95% CI, 1.24 to 1.55), p=1.61×10-8) and LRP1 at 12q13.3 (rs60565245, 2.18 (95% CI, 1.66 to 2.86), p=2.35×10-8). FGF1 and THSD7A variants were associated with decreased mRNA expression of each gene in iPSC-MNs and reduced in vitro survival of iPSC-MNs obtained from patients with ALS. The iPSC-MN in vitro survival was reduced when the expression of FGF1 and THSD7A was partially disrupted. The rs60565245 was not associated with LRP1 mRNA expression. CONCLUSIONS We identified three loci associated with the survival of patients with sporadic ALS, decreased mRNA expression of FGF1 and THSD7A and the viability of iPSC-MNs from patients. The iPSC-MN model reflects the association between patient prognosis and genotype and can contribute to target screening and validation for therapeutic intervention.
Collapse
Affiliation(s)
- Ryoichi Nakamura
- Department of Neurology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Genki Tohnai
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Division of ALS Research, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Naoki Atsuta
- Department of Neurology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Hirohisa Watanabe
- Department of Neurology, Fujita Health University, Toyoake, Aichi, Japan
- Brain and Mind Research Center, Nagoya University, Nagoya, Aichi, Japan
| | - Daisuke Ito
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of Clinical Research Education, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Akihiro Hirakawa
- Department of Clinical Biostatistics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Yuishin Izumi
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Mitsuya Morita
- Division of Neurology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Takehisa Hirayama
- Department of Neurology, Toho University Faculty of Medicine, Ota-ku, Tokyo, Japan
| | - Osamu Kano
- Department of Neurology, Toho University Faculty of Medicine, Ota-ku, Tokyo, Japan
| | - Kazuaki Kanai
- Department of Neurology, Fukushima Medical University School of Medicine, Fukushima, Japan
- Department of Neurology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Akira Taniguchi
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
| | - Naoki Suzuki
- Department of Neurology, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Masashi Aoki
- Department of Neurology, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Ikuko Iwata
- Department of Neurology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Ichiro Yabe
- Department of Neurology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Kazumoto Shibuya
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Satoshi Kuwabara
- Department of Neurology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masaya Oda
- Department of Neurology, Vihara Hananosato Hospital, Miyoshi, Hiroshima, Japan
| | - Rina Hashimoto
- Department of Neurology, National Hospital Organization Higashinagoya National Hospital, Nagoya, Aichi, Japan
| | - Ikuko Aiba
- Department of Neurology, National Hospital Organization Higashinagoya National Hospital, Nagoya, Aichi, Japan
| | - Tomohiko Ishihara
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Toru Yamashita
- Department of Neurology, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Koji Abe
- Department of Neurology, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Kouichi Mizoguchi
- Department of Neurology, National Hospital Organization Shizuoka Medical Center, Shizuoka, Japan
| | - Toshio Shimizu
- Department of Neurology, Tokyo Metropolitan Neurological Hospital, Fuchu, Tokyo, Japan
| | - Yoshio Ikeda
- Department of Neurology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Takanori Yokota
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Kazuko Hasegawa
- Division of Neurology, National Hospital Organization, Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Fumiaki Tanaka
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa, Japan
| | - Kenji Nakashima
- Department of Neurology, National Hospital Organization, Matsue Medical Center, Matsue, Shimane, Japan
| | - Ryuji Kaji
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Jun-Ichi Niwa
- Department of Neurology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Manabu Doyu
- Department of Neurology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Shiro Ikegawa
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Koki Fujimori
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Shiho Nakamura
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Fumiko Ozawa
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Satoru Morimoto
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Kazunari Onodera
- Department of Neurology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Takuji Ito
- Department of Neurology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Yohei Okada
- Department of Neurology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Gen Sobue
- Brain and Mind Research Center, Nagoya University, Nagoya, Aichi, Japan
- Aichi Medical University, Nagakute, Aichi, Japan
| |
Collapse
|
9
|
Liu WW, Kinzy TG, Cooke Bailey JN, Xu Z, Hysi P, Wiggs JL. Mechanosensitive ion channel gene survey suggests potential roles in primary open angle glaucoma. Sci Rep 2023; 13:15871. [PMID: 37741866 PMCID: PMC10517927 DOI: 10.1038/s41598-023-43072-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/18/2023] [Indexed: 09/25/2023] Open
Abstract
Although glaucoma is a disease modulated by eye pressure, the mechanisms of pressure sensing in the eye are not well understood. Here, we investigated associations between mechanosensitive ion channel gene variants and primary open-angle glaucoma (POAG). Common (minor allele frequency > 5%) single nucleotide polymorphisms located within the genomic regions of 20 mechanosensitive ion channel genes in the K2P, TMEM63, PIEZO and TRP channel families were assessed using genotype data from the NEIGHBORHOOD consortium of 3853 cases and 33,480 controls. Rare (minor allele frequency < 1%) coding variants were assessed using exome array genotyping data for 2606 cases and 2606 controls. Association with POAG was analyzed using logistic regression adjusting for age and sex. Two rare PIEZO1 coding variants with protective effects were identified in the NEIGHBOR dataset: R1527H, (OR 0.17, P = 0.0018) and a variant that alters a canonical splice donor site, g.16-88737727-C-G Hg38 (OR 0.38, P = 0.02). Both variants showed similar effects in the UK Biobank and the R1527H also in the FinnGen database. Several common variants also reached study-specific thresholds for association in the NEIGHBORHOOD dataset. These results identify novel variants in several mechanosensitive channel genes that show associations with POAG, suggesting that these channels may be potential therapeutic targets.
Collapse
Affiliation(s)
- Wendy W Liu
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, 2370 Watson Court, Palo Alto, CA, 94303, USA.
| | - Tyler G Kinzy
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Jessica N Cooke Bailey
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Zihe Xu
- Department of Ophthalmology, King's College London, St. Thomas' Hospital, London, UK
| | - Pirro Hysi
- Department of Ophthalmology, King's College London, St. Thomas' Hospital, London, UK
- Department of Twin Research and Genetic Epidemiology, King's College London, St. Thomas' Hospital, London, UK
| | - Janey L Wiggs
- Massachusetts Eye and Ear, Harvard Medical School Boston, Boston, MA, USA
| |
Collapse
|
10
|
van de Vegte YJ, Eppinga RN, van der Ende MY, Hagemeijer YP, Mahendran Y, Salfati E, Smith AV, Tan VY, Arking DE, Ntalla I, Appel EV, Schurmann C, Brody JA, Rueedi R, Polasek O, Sveinbjornsson G, Lecoeur C, Ladenvall C, Zhao JH, Isaacs A, Wang L, Luan J, Hwang SJ, Mononen N, Auro K, Jackson AU, Bielak LF, Zeng L, Shah N, Nethander M, Campbell A, Rankinen T, Pechlivanis S, Qi L, Zhao W, Rizzi F, Tanaka T, Robino A, Cocca M, Lange L, Müller-Nurasyid M, Roselli C, Zhang W, Kleber ME, Guo X, Lin HJ, Pavani F, Galesloot TE, Noordam R, Milaneschi Y, Schraut KE, den Hoed M, Degenhardt F, Trompet S, van den Berg ME, Pistis G, Tham YC, Weiss S, Sim XS, Li HL, van der Most PJ, Nolte IM, Lyytikäinen LP, Said MA, Witte DR, Iribarren C, Launer L, Ring SM, de Vries PS, Sever P, Linneberg A, Bottinger EP, Padmanabhan S, Psaty BM, Sotoodehnia N, Kolcic I, Arnar DO, Gudbjartsson DF, Holm H, Balkau B, Silva CT, Newton-Cheh CH, Nikus K, Salo P, Mohlke KL, Peyser PA, Schunkert H, Lorentzon M, Lahti J, Rao DC, Cornelis MC, Faul JD, Smith JA, Stolarz-Skrzypek K, Bandinelli S, Concas MP, Sinagra G, Meitinger T, Waldenberger M, Sinner MF, et alvan de Vegte YJ, Eppinga RN, van der Ende MY, Hagemeijer YP, Mahendran Y, Salfati E, Smith AV, Tan VY, Arking DE, Ntalla I, Appel EV, Schurmann C, Brody JA, Rueedi R, Polasek O, Sveinbjornsson G, Lecoeur C, Ladenvall C, Zhao JH, Isaacs A, Wang L, Luan J, Hwang SJ, Mononen N, Auro K, Jackson AU, Bielak LF, Zeng L, Shah N, Nethander M, Campbell A, Rankinen T, Pechlivanis S, Qi L, Zhao W, Rizzi F, Tanaka T, Robino A, Cocca M, Lange L, Müller-Nurasyid M, Roselli C, Zhang W, Kleber ME, Guo X, Lin HJ, Pavani F, Galesloot TE, Noordam R, Milaneschi Y, Schraut KE, den Hoed M, Degenhardt F, Trompet S, van den Berg ME, Pistis G, Tham YC, Weiss S, Sim XS, Li HL, van der Most PJ, Nolte IM, Lyytikäinen LP, Said MA, Witte DR, Iribarren C, Launer L, Ring SM, de Vries PS, Sever P, Linneberg A, Bottinger EP, Padmanabhan S, Psaty BM, Sotoodehnia N, Kolcic I, Arnar DO, Gudbjartsson DF, Holm H, Balkau B, Silva CT, Newton-Cheh CH, Nikus K, Salo P, Mohlke KL, Peyser PA, Schunkert H, Lorentzon M, Lahti J, Rao DC, Cornelis MC, Faul JD, Smith JA, Stolarz-Skrzypek K, Bandinelli S, Concas MP, Sinagra G, Meitinger T, Waldenberger M, Sinner MF, Strauch K, Delgado GE, Taylor KD, Yao J, Foco L, Melander O, de Graaf J, de Mutsert R, de Geus EJC, Johansson Å, Joshi PK, Lind L, Franke A, Macfarlane PW, Tarasov KV, Tan N, Felix SB, Tai ES, Quek DQ, Snieder H, Ormel J, Ingelsson M, Lindgren C, Morris AP, Raitakari OT, Hansen T, Assimes T, Gudnason V, Timpson NJ, Morrison AC, Munroe PB, Strachan DP, Grarup N, Loos RJF, Heckbert SR, Vollenweider P, Hayward C, Stefansson K, Froguel P, Groop L, Wareham NJ, van Duijn CM, Feitosa MF, O'Donnell CJ, Kähönen M, Perola M, Boehnke M, Kardia SLR, Erdmann J, Palmer CNA, Ohlsson C, Porteous DJ, Eriksson JG, Bouchard C, Moebus S, Kraft P, Weir DR, Cusi D, Ferrucci L, Ulivi S, Girotto G, Correa A, Kääb S, Peters A, Chambers JC, Kooner JS, März W, Rotter JI, Hicks AA, Smith JG, Kiemeney LALM, Mook-Kanamori DO, Penninx BWJH, Gyllensten U, Wilson JF, Burgess S, Sundström J, Lieb W, Jukema JW, Eijgelsheim M, Lakatta ELM, Cheng CY, Dörr M, Wong TY, Sabanayagam C, Oldehinkel AJ, Riese H, Lehtimäki T, Verweij N, van der Harst P. Genetic insights into resting heart rate and its role in cardiovascular disease. Nat Commun 2023; 14:4646. [PMID: 37532724 PMCID: PMC10397318 DOI: 10.1038/s41467-023-39521-2] [Show More Authors] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 06/16/2023] [Indexed: 08/04/2023] Open
Abstract
Resting heart rate is associated with cardiovascular diseases and mortality in observational and Mendelian randomization studies. The aims of this study are to extend the number of resting heart rate associated genetic variants and to obtain further insights in resting heart rate biology and its clinical consequences. A genome-wide meta-analysis of 100 studies in up to 835,465 individuals reveals 493 independent genetic variants in 352 loci, including 68 genetic variants outside previously identified resting heart rate associated loci. We prioritize 670 genes and in silico annotations point to their enrichment in cardiomyocytes and provide insights in their ECG signature. Two-sample Mendelian randomization analyses indicate that higher genetically predicted resting heart rate increases risk of dilated cardiomyopathy, but decreases risk of developing atrial fibrillation, ischemic stroke, and cardio-embolic stroke. We do not find evidence for a linear or non-linear genetic association between resting heart rate and all-cause mortality in contrast to our previous Mendelian randomization study. Systematic alteration of key differences between the current and previous Mendelian randomization study indicates that the most likely cause of the discrepancy between these studies arises from false positive findings in previous one-sample MR analyses caused by weak-instrument bias at lower P-value thresholds. The results extend our understanding of resting heart rate biology and give additional insights in its role in cardiovascular disease development.
Collapse
Affiliation(s)
- Yordi J van de Vegte
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, 9700RB, the Netherlands
| | - Ruben N Eppinga
- Department of Cardiology, Isala Zwolle ziekenhuis, Zwolle, 8025 AB, the Netherlands
| | - M Yldau van der Ende
- Department of Cardiology, University medical Center Utrecht, Utrecht, 3584 Cx, the Netherlands
| | - Yanick P Hagemeijer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, 9700RB, the Netherlands
- Analytical Biochemistry, University of Groningen, Groningen, 9713 AV, the Netherlands
| | - Yuvaraj Mahendran
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medicine Science, University of Copenhagen, Copenhagen Ø, 2100, Denmark
| | - Elias Salfati
- Department of Medicine, Stanford University School of Medicine, Stanford, 94305, USA
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | - Albert V Smith
- Department of Biostatistics, University of Michigan, Ann Arbor, MI48109, USA
| | - Vanessa Y Tan
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, BS82BN, UK
- MRC Integrative Epidemiology, University of Bristol, Bristol, BS82BN, UK
| | - Dan E Arking
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, 21215, USA
| | - Ioanna Ntalla
- Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Emil V Appel
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medicine Science, University of Copenhagen, Copenhagen Ø, 2100, Denmark
| | - Claudia Schurmann
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, 10029, USA
| | | | - Rico Rueedi
- Department of Computational Biology, University of Lausanne, Lausanne, 1015, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
| | - Ozren Polasek
- Department of Public Health, University of Split School of Medicine, Split, 21000, Croatia
- Algebra LAB, Algebra University College, Zagreb, 10000, Croatia
| | | | - Cecile Lecoeur
- UMR 8199, University of Lille Nord de France, Lille, 59000, France
| | - Claes Ladenvall
- Clinial Genomics Uppsala, Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, 75185, Sweden
- Lund University Diabetes Center, Department of Clinical Sciences, Lund University, Malmö, 20502, Sweden
| | - Jing Hua Zhao
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, Victor Phillip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Cambridge, CB2 0BB, UK
| | - Aaron Isaacs
- CARIM School for Cardiovascular Diseases, Maastricht Centre for Systems Biology (MaCSBio), Department of Physiology, Maastricht University, Maastricht, 6229ER, Netherlands
| | - Lihua Wang
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63108-2212, Campus Box 8506, USA
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, CB2 0QQ, UK
| | - Shih-Jen Hwang
- Division of Intramural Research, National Heart Lung and Blood Institute, NIH, USA, Framingham, 1702, USA
| | - Nina Mononen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, FI-33014, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, FI-33014, Finland
| | - Kirsi Auro
- Department of Health, unit of genetics and biomarkers, , National Institute for Health and Welfare, Finland, Helsinki, FI-00290, Finland
- Department of molecular medicine, University of Helsinki, Helsinki, FI-00290, Finland
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Lawrence F Bielak
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Linyao Zeng
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, Munich, 80636, Germany
| | - Nabi Shah
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
- Pharmacogenetics Research Lab, Department of Pharmacy, COMSATS University Islamabad, Abbottabad, 22060, Pakistan
| | - Maria Nethander
- Sahlgrenska Osteoporosis Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 41345, Sweden
- Bioinformatics Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 40530, Sweden
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics & Cancer, University of Edinburgh, Edinburgh, EH4 2XU, UK
- Usher Institute for Population Health Sciences and Informatics, The University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Sonali Pechlivanis
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital of Essen, University Duisburg-Essen, Essen, 45122, Germany
| | - Lu Qi
- Department of Epidemiology, Tulane University, New Orleans, LA, 70112, USA
| | - Wei Zhao
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, 48109, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, 48104, USA
| | - Federica Rizzi
- Unit of Biomedicine, Bio4Dreams-Business Nursery for Life Sciences, Milano, 20121, Italy
| | - Toshiko Tanaka
- Longitudinal Study Section, National Institute on Aging, Baltimore, 21224, USA
| | - Antonietta Robino
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, 34137, Italy
| | - Massimiliano Cocca
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, 34137, Italy
| | - Leslie Lange
- Medicine, University of Colorado Anschutz Medical Campus, Aurora, 80045, USA
| | - Martina Müller-Nurasyid
- IBE, Ludwig-Maximilians-University Munich, LMU Munich, Munich, 81377, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, 55101, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Carolina Roselli
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, 02142, USA
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Imperial College London, London, W2 1PG, UK
- Department of Cardiology, Ealing Hospital, London North West University Healthcare NHS Trust, Middlesex, UB1 3HW, UK
| | - Marcus E Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, 68167, Germany
- SYNLAB MVZ Humangenetik Mannheim, Mannheim, 68163, Germany
| | - Xiuqing Guo
- Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA, Torrance, 90502, USA
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, 90502, USA
| | - Henry J Lin
- Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA, Torrance, 90502, USA
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, 90502, USA
| | - Francesca Pavani
- Institute for Biomedicine, Eurac Research, Bolzano, 39100, Italy
| | | | - Raymond Noordam
- Department of Internal Medicine, section Gerontology and Geriatrics, Leiden University Medical Center, Leiden, 2300 RC, the Netherlands
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam Public Health, Amsterdam UMC, Amsterdam UMC, Vrije Universiteit, Amsterdam, Amsterdam, 1081 HL, the Netherlands
| | - Katharina E Schraut
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, EH16 4TJ, Scotland, UK
| | - Marcel den Hoed
- The Beijer laboratory and Department of Immunology, Genetics and Pathology, Uppsala University and Science for Life Laboratory, Uppsala, 75237, Sweden
| | - Frauke Degenhardt
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, 24105, Germany
| | - Stella Trompet
- Department of Internal Medicine, section Gerontology and Geriatrics, Leiden University Medical Center, Leiden, 2300 RC, the Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, ZA, 2333, the Netherlands
| | - Marten E van den Berg
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, 3015GD, the Netherlands
| | - Giorgio Pistis
- Institute of Genetics and Biomedic Research (IRGB), Italian National Research Council (CNR), Monserrato, (CA), 9042, Italy
- Center for Statistical Genetics, University of Michigan, Ann Arbor, 48109, USA
| | - Yih-Chung Tham
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, 169856, Singapore
| | - Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, 17475, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, 17475, Germany
| | - Xueling S Sim
- Saw Swee Hock School of Public Health, National University Health System and National University of Singapore, Singapore, 117549, Singapore
| | - Hengtong L Li
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, 169856, Singapore
| | - Peter J van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, 9700 RB, The Netherlands
| | - Ilja M Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, 9700 RB, The Netherlands
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, FI-33014, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, FI-33014, Finland
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB2 0SL, UK
| | - M Abdullah Said
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, 9700RB, the Netherlands
| | - Daniel R Witte
- Department of Public Health, Aarhus University, Aarhus C, 8000, Denmark
| | - Carlos Iribarren
- Division of Research, Kaiser Permenente of Northern California, Oakland, 94612, USA
- The Scripps Research Institute, La Jolla, 10550, USA
| | | | - Susan M Ring
- Bristol Medical School, Population Health Sciences, University of Bristol, Bristol, BS82BN, UK
- MRC Integrative Epidemiology, University of Bristol, Bristol, BS82BN, UK
| | - Paul S de Vries
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, School of Public Health, Houston, 77030, USA
| | - Peter Sever
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Allan Linneberg
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Copenhagen, 2400, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Erwin P Bottinger
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, 10029, USA
- Department of Preventive Medicine, The Icahn School of Medicine at Mount Sinai, New York, 10029, USA
| | - Sandosh Padmanabhan
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Bruce M Psaty
- Departments of Medicine, Epidemiology and Health Systems and Population Health, University of Washington, Seattle, 98195, USA
| | - Nona Sotoodehnia
- Medicine and Epidemiology, University of Washington, Seattle, 98195, USA
| | - Ivana Kolcic
- Department of Public Health, University of Split School of Medicine, Split, 21000, Croatia
- Algebra LAB, Algebra University College, Zagreb, 10000, Croatia
| | - David O Arnar
- deCODE genetics / Amgen Inc., Reykjavik, 102, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, 101, Iceland
- Department of Medicine, Landspitali-The National University Hospital of Iceland, Reykjavik, 101, Iceland
| | - Daniel F Gudbjartsson
- deCODE genetics / Amgen Inc., Reykjavik, 102, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, 101, Iceland
| | - Hilma Holm
- deCODE genetics / Amgen Inc., Reykjavik, 102, Iceland
| | - Beverley Balkau
- Centre for Research in Epidemiology and Population Health, Institut national de la santé et de la recherche médicale, Villejuif, 94800, France
- UMRS 1018, University Versailles Saint-Quentin-en-Yvelines, Versailles, 78035, France
- UMRS 1018, University Paris Sud, Villejuif, 94807, France
| | - Claudia T Silva
- Genetic Epidemiology Unit, Dept. of Epidemiology, Erasmus University Medical Center, Rotterdam, 3000CA, Netherlands
| | | | - Kjell Nikus
- Department of Cardiology, Heart Center, Tampere University Hospital, Tampere, FI-33521, Finland
- Department of Cardiology, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, FI-33014, Finland
| | - Perttu Salo
- Department of Health, unit of genetics and biomarkers, , National Institute for Health and Welfare, Finland, Helsinki, FI-00290, Finland
- Department of molecular medicine, University of Helsinki, Helsinki, FI-00290, Finland
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Patricia A Peyser
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Heribert Schunkert
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, Munich, 80636, Germany
- Deutsches Zentrum für Herz- und Kreislauferkrankungen (DZHK), Partner Site Munich Heart Alliance, Munich, 80636, Germany
| | - Mattias Lorentzon
- Sahlgrenska Osteoporosis Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 41345, Sweden
- Region Västra Götaland, Geriatric Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Mölndal, 43180, Sweden
- Mary McKillop Institute for Health Research, Australian Catholic University, Melbourne, 3000, Australia
| | - Jari Lahti
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, 00014, Finland
| | - Dabeeru C Rao
- Division of Biostatistics, Washington University, St. Louis, MO, 63110, USA
| | | | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, 48104, USA
| | - Jennifer A Smith
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, 48109, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, 48104, USA
| | - Katarzyna Stolarz-Skrzypek
- Department of Cardiology, Interventional Electrocardiology and Hypertension, Jagiellonian University Medical College, Kraków, 31-008, Poland
| | - Stefania Bandinelli
- Geriatric Unit, Unità sanitaria locale Toscana Centro, Florence, 50142, Italy
| | - Maria Pina Concas
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, 34137, Italy
| | - Gianfranco Sinagra
- Cardiovascular Department, "Ospedali Riuniti and University of Trieste", Trieste, 34149, Italy
| | - Thomas Meitinger
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, München, 81675, Germany
- Institute of Human Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Melanie Waldenberger
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- German Centre for Cardiovascular Research (DZHK), partner site: Munich Heart Alliance, Munich, 80802, Germany
| | - Moritz F Sinner
- German Centre for Cardiovascular Research (DZHK), partner site: Munich Heart Alliance, Munich, 80802, Germany
- Department of Cardiology, University Hospital, LMU Munich, Munich, 81377, Germany
| | - Konstantin Strauch
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, 55101, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Chair of Genetic Epidemiology, IBE, Faculty of Medicine, LMU Munich, Munich, 81377, Germany
| | - Graciela E Delgado
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, 68167, Germany
| | - Kent D Taylor
- Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA, Torrance, 90502, USA
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, 90502, USA
| | - Jie Yao
- Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA, Torrance, 90502, USA
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, 90502, USA
| | - Luisa Foco
- Institute for Biomedicine, Eurac Research, Bolzano, 39100, Italy
| | - Olle Melander
- Department of Internal Medicine, Clinical Sciences, Lund University and Skåne University Hospital, Malmo, 221 85, Sweden
- Lund University Diabetes Center, Lund University, Malmö, 221 85, Sweden
| | | | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, 2300 RC, the Netherlands
| | - Eco J C de Geus
- Biological Psychology, EMGO+ Institute for Health and Care Research and Neuroscience Campus Amsterdam, VU University, Amsterdam, 1081 BT, the Netherlands
| | - Åsa Johansson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, 75108, Sweden
| | - Peter K Joshi
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, EH8 9AG, Scotland, UK
| | - Lars Lind
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University Hospital, Uppsala, 75237, Sweden
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, 24105, Germany
| | - Peter W Macfarlane
- Institute of Health and Wellbeing, Faculty of Medicine, University of Glasgow, Glasgow, G12 0XH, UK
| | - Kirill V Tarasov
- Laboratory of Cardiovascular Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Nicholas Tan
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, 169856, Singapore
| | - Stephan B Felix
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, 17475, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, 17475, Germany
| | - E-Shyong Tai
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, 169856, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Debra Q Quek
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, 169856, Singapore
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, 9700 RB, The Netherlands
| | - Johan Ormel
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, 9700 RB, The Netherlands
| | - Martin Ingelsson
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Uppsala University, Uppsala, 75237, Sweden
| | - Cecilia Lindgren
- Genetic and Genomic Epidemiology Unit, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Andrew P Morris
- Genetic and Genomic Epidemiology Unit, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Olli T Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, FI-20521, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, FI-20521, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, FI-20521, Finland
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medicine Science, University of Copenhagen, Copenhagen Ø, 2100, Denmark
| | - Themistocles Assimes
- Department of Medicine, Stanford University School of Medicine, Stanford, 94305, USA
| | - Vilmundur Gudnason
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
- Icelandic Heart Association, Kopavogur, 201, Iceland
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 2BN, UK
- Population Health Sciences, Bristol Medical School,, University of Bristol, Bristol, BS8 2BN, UK
| | - Alanna C Morrison
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, School of Public Health, Houston, 77030, USA
| | - Patricia B Munroe
- Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
- NIHR Barts Biomedical Research Centre, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - David P Strachan
- Population Health Research Institute, St George's, University of London, London, SW17 0RE, UK
| | - Niels Grarup
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medicine Science, University of Copenhagen, Copenhagen Ø, 2100, Denmark
| | - Ruth J F Loos
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medicine Science, University of Copenhagen, Copenhagen Ø, 2100, Denmark
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, 10029, USA
- The Mindich Child Health and Development Institute, The Icahn School of Medicine at Mount Sinai, New York, 10029, USA
| | - Susan R Heckbert
- Department of Epidemiology, University of Washington, Seattle, 98195, USA
| | - Peter Vollenweider
- Department of Medicine, Internal Medicine, Lausanne University hospital, Lausanne, 1015, Switzerland
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, Scotland, UK
| | - Kari Stefansson
- deCODE genetics / Amgen Inc., Reykjavik, 102, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, 101, Iceland
| | - Philippe Froguel
- Department of Metabolism, Imperial College London, London, W12 0HS, UK
- Inserm/CNRS UMR 1283/8199, Pasteur Institute of Lille, Lille University Hospital, EGID, Lille, 59000, France
- University of Lille, Lille, 59000, France
| | - Leif Groop
- Lund University Diabetes Center, Department of Clinical Sciences, Lund University, Malmö, 20502, Sweden
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00290, Finland
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, CB2 0QQ, UK
| | - Cornelia M van Duijn
- Genetic Epidemiology Unit, Dept. of Epidemiology, Erasmus University Medical Center, Rotterdam, 3000CA, Netherlands
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, 63108-2212, Campus Box 8506, USA
| | - Christopher J O'Donnell
- Cardiology Section, VA Boston Healthcare System, Harvard Medical School, Boston, MA, 02132, USA
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere, FI-33521, Finland
- Department of Clinical Physiology, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, FI-33521, Finland
| | - Markus Perola
- Department of Health, unit of genetics and biomarkers, , National Institute for Health and Welfare, Finland, Helsinki, FI-00290, Finland
- Department of molecular medicine, University of Helsinki, Helsinki, FI-00290, Finland
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sharon L R Kardia
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, Lübeck, 23562, Germany
| | - Colin N A Palmer
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
| | - Claes Ohlsson
- Sahlgrenska Osteoporosis Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 41345, Sweden
- Department of Drug Treatment, Sahlgrenska University Hospital, Gothenburg, 41345, Sweden
| | - David J Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Johan G Eriksson
- Department of General practice and primary care, University of Helsinki, Helsinki, 00014, Finland
- Department of Obstetrics and Gynecology, National University of Singapore, Singapore, 119228, Singapore
- Public health Research Program, Folkhalsan Research Center, Helsinki, 000250, Finland
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Susanne Moebus
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital of Essen, University Duisburg-Essen, Essen, 45122, Germany
- Centre for Urban Epidemiology, University Hospital of Essen, University Duisburg-Essen, Essen, 45122, Germany
| | - Peter Kraft
- Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02112, USA
| | - David R Weir
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, 48104, USA
| | - Daniele Cusi
- Unit of Biomedicine, Bio4Dreams-Business Nursery for Life Sciences, Milano, 20121, Italy
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, (MI), 20090, Italy
| | - Luigi Ferrucci
- Longitudinal Study Section, National Institute on Aging, Baltimore, 21224, USA
| | - Sheila Ulivi
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, 34137, Italy
| | - Giorgia Girotto
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, 34137, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, 34149, Italy
| | - Adolfo Correa
- Jackson Heart Study, University of Mississippi Medical Center, Jackson, 39216, USA
| | - Stefan Kääb
- German Centre for Cardiovascular Research (DZHK), partner site: Munich Heart Alliance, Munich, 80802, Germany
- Department of Cardiology, University Hospital, LMU Munich, Munich, 81377, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, 85764, Germany
- German Centre for Cardiovascular Research (DZHK), partner site: Munich Heart Alliance, Munich, 80802, Germany
- Chair of Epidemiology, Institute for Medical Information Processing, Biometry and Epidemiology, Ludwig-Maximilians-Universität München, Munich, 81377, Germany
| | - John C Chambers
- Department of Epidemiology and Biostatistics, Imperial College London, London, W2 1PG, UK
- Department of Cardiology, Ealing Hospital, London North West University Healthcare NHS Trust, Middlesex, UB1 3HW, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
| | - Jaspal S Kooner
- Department of Cardiology, Ealing Hospital, London North West University Healthcare NHS Trust, Middlesex, UB1 3HW, UK
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK
- Imperial College Healthcare NHS Trust, Imperial College London, London, W12 0HS, UK
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, 68167, Germany
- Synlab Academy, Synlab Holding Deutschland GmbH, Mannheim, 68161, Germany
| | - Jerome I Rotter
- Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA, Torrance, 90502, USA
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, 90502, USA
| | - Andrew A Hicks
- Institute for Biomedicine, Eurac Research, Bolzano, 39100, Italy
| | - J Gustav Smith
- Department of Cardiology, Clinical Sciences, Lund University and Skåne University Hospital, Lund, 221 85, Sweden
- Wallenberg Center for Molecular Medicine and Lund University Diabetes Center, Lund University, Lund, 221 84, Sweden
- The Wallenberg Laboratory/Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University and the Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, 413 45, Sweden
| | | | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, 2300 RC, the Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, 2300 RC, the Netherlands
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam Public Health, Amsterdam UMC, Amsterdam UMC, Vrije Universiteit, Amsterdam, Amsterdam, 1081 HL, the Netherlands
| | - Ulf Gyllensten
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, 75108, Sweden
| | - James F Wilson
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, EH8 9AG, Scotland, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU, Scotland, UK
| | - Stephen Burgess
- MRC Biostatistics Unit, University of Cambridge, Cambridge, CB2 0SR, UK
| | - Johan Sundström
- Department of Medical Sciences, Cardiovascular Epidemiology, Uppsala University Hospital, Uppsala, 75237, Sweden
| | - Wolfgang Lieb
- Institute of Epidemiology and Biobank PopGen, Kiel University, Kiel, 24105, Germany
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, ZA, 2333, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, ZA, 2333, the Netherlands
- Netherlands Heart Institute, Utrecht, 3511 EP, the Netherlands
| | - Mark Eijgelsheim
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, 3015GD, the Netherlands
- Department of Nephrology, University Medical Center Groningen, Groningen, 9700RB, the Netherlands
| | - Edward L M Lakatta
- Laboratory of Cardiovascular Sciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, 169856, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, 169857, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Marcus Dörr
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, 17475, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, 17475, Germany
| | - Tien-Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, 169856, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, 169857, Singapore
- Tsinghua Medicine, Tsinghua University, Beijing, 100084, China
| | - Charumathi Sabanayagam
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, 169856, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Albertine J Oldehinkel
- Interdisciplinary Center Psychopathology and Emotion Regulation, University of Groningen, University Medical Center Groningen, Groningen, 9700 RB, The Netherlands
| | - Harriette Riese
- Department of Psychiatry, University of Groningen, University Medical Center Groningen, Groningen, 9700 RB, The Netherlands
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, FI-33014, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, FI-33014, Finland
| | - Niek Verweij
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, 9700RB, the Netherlands
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, 9700RB, the Netherlands.
- Department of Cardiology, University medical Center Utrecht, Utrecht, 3584 Cx, the Netherlands.
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, 9700RB, the Netherlands.
| |
Collapse
|
11
|
Van Buren E, Radicioni G, Lester S, O’Neal WK, Dang H, Kasela S, Garudadri S, Curtis JL, Han MK, Krishnan JA, Wan ES, Silverman EK, Hastie A, Ortega VE, Lappalainen T, Nawijn MC, van den Berge M, Christenson SA, Li Y, Cho MH, Kesimer M, Kelada SNP. Genetic regulators of sputum mucin concentration and their associations with COPD phenotypes. PLoS Genet 2023; 19:e1010445. [PMID: 37352370 PMCID: PMC10325042 DOI: 10.1371/journal.pgen.1010445] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 07/06/2023] [Accepted: 04/26/2023] [Indexed: 06/25/2023] Open
Abstract
Hyper-secretion and/or hyper-concentration of mucus is a defining feature of multiple obstructive lung diseases, including chronic obstructive pulmonary disease (COPD). Mucus itself is composed of a mixture of water, ions, salt and proteins, of which the gel-forming mucins, MUC5AC and MUC5B, are the most abundant. Recent studies have linked the concentrations of these proteins in sputum to COPD phenotypes, including chronic bronchitis (CB) and acute exacerbations (AE). We sought to determine whether common genetic variants influence sputum mucin concentrations and whether these variants are also associated with COPD phenotypes, specifically CB and AE. We performed a GWAS to identify quantitative trait loci for sputum mucin protein concentration (pQTL) in the Sub-Populations and InteRmediate Outcome Measures in COPD Study (SPIROMICS, n = 708 for total mucin, n = 215 for MUC5AC, MUC5B). Subsequently, we tested for associations of mucin pQTL with CB and AE using regression modeling (n = 822-1300). Replication analysis was conducted using data from COPDGene (n = 5740) and by examining results from the UK Biobank. We identified one genome-wide significant pQTL for MUC5AC (rs75401036) and two for MUC5B (rs140324259, rs10001928). The strongest association for MUC5B, with rs140324259 on chromosome 11, explained 14% of variation in sputum MUC5B. Despite being associated with lower MUC5B, the C allele of rs140324259 conferred increased risk of CB (odds ratio (OR) = 1.42; 95% confidence interval (CI): 1.10-1.80) as well as AE ascertained over three years of follow up (OR = 1.41; 95% CI: 1.02-1.94). Associations between rs140324259 and CB or AE did not replicate in COPDGene. However, in the UK Biobank, rs140324259 was associated with phenotypes that define CB, namely chronic mucus production and cough, again with the C allele conferring increased risk. We conclude that sputum MUC5AC and MUC5B concentrations are associated with common genetic variants, and the top locus for MUC5B may influence COPD phenotypes, in particular CB.
Collapse
Affiliation(s)
- Eric Van Buren
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Giorgia Radicioni
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Sarah Lester
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Wanda K. O’Neal
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Hong Dang
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Silva Kasela
- New York Genome Center, New York, New York, United States of America
- Department of Systems Biology, Columbia University, New York, New York, United States of America
| | - Suresh Garudadri
- Division of Pulmonary, Critical Care, Allergy, & Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Jeffrey L. Curtis
- Pulmonary & Critical Care Medicine Division, University of Michigan, Ann Arbor, Michigan, United States of America
- Medical Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan, United States of America
| | - MeiLan K. Han
- Pulmonary & Critical Care Medicine Division, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jerry A. Krishnan
- Breathe Chicago Center, University of Illinois, Chicago, Illinois, United States of America
| | - Emily S. Wan
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- VA Boston Healthcare System, Jamaica Plain, Massachusetts, United States of America
| | - Edwin K. Silverman
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Annette Hastie
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Victor E. Ortega
- Department of Internal Medicine, Division of Respiratory Medicine, Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Tuuli Lappalainen
- New York Genome Center, New York, New York, United States of America
- Department of Systems Biology, Columbia University, New York, New York, United States of America
| | - Martijn C. Nawijn
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, the Netherlands
| | - Maarten van den Berge
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, the Netherlands
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Stephanie A. Christenson
- Division of Pulmonary, Critical Care, Allergy, & Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Yun Li
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Michael H. Cho
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Mehmet Kesimer
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Samir N. P. Kelada
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
12
|
Moore A, Busch MP, Dziewulska K, Francis RO, Hod EA, Zimring JC, D’Alessandro A, Page GP. Genome-wide metabolite quantitative trait loci analysis (mQTL) in red blood cells from volunteer blood donors. J Biol Chem 2022; 298:102706. [PMID: 36395887 PMCID: PMC9763692 DOI: 10.1016/j.jbc.2022.102706] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
The red blood cell (RBC)-Omics study, part of the larger NHLBI-funded Recipient Epidemiology and Donor Evaluation Study (REDS-III), aims to understand the genetic contribution to blood donor RBC characteristics. Previous work identified donor demographic, behavioral, genetic, and metabolic underpinnings to blood donation, storage, and (to a lesser extent) transfusion outcomes, but none have yet linked the genetic and metabolic bodies of work. We performed a genome-wide association (GWA) analysis using RBC-Omics study participants with generated untargeted metabolomics data to identify metabolite quantitative trait loci in RBCs. We performed GWA analyses of 382 metabolites in 243 individuals imputed using the 1000 Genomes Project phase 3 all-ancestry reference panel. Analyses were conducted using ProbABEL and adjusted for sex, age, donation center, number of whole blood donations in the past 2 years, and first 10 principal components of ancestry. Our results identified 423 independent genetic loci associated with 132 metabolites (p < 5×10-8). Potentially novel locus-metabolite associations were identified for the region encoding heme transporter FLVCR1 and choline and for lysophosphatidylcholine acetyltransferase LPCAT3 and lysophosphatidylserine 16.0, 18.0, 18.1, and 18.2; these associations are supported by published rare disease and mouse studies. We also confirmed previous metabolite GWA results for associations, including N(6)-methyl-L-lysine and protein PYROXD2 and various carnitines and transporter SLC22A16. Association between pyruvate levels and G6PD polymorphisms was validated in an independent cohort and novel murine models of G6PD deficiency (African and Mediterranean variants). We demonstrate that it is possible to perform metabolomics-scale GWA analyses with a modest, trans-ancestry sample size.
Collapse
Affiliation(s)
- Amy Moore
- Division of Biostatistics and Epidemiology, RTI International, Atlanta, Georgia, USA
| | | | - Karolina Dziewulska
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Richard O. Francis
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, USA
| | - Eldad A. Hod
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, USA
| | - James C. Zimring
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Angelo D’Alessandro
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York, USA,Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Anschutz Medical Campus, Aurora, CO, USA,For correspondence: Grier P. Page; Angelo D’Alessandro
| | - Grier P. Page
- Division of Biostatistics and Epidemiology, RTI International, Atlanta, Georgia, USA,For correspondence: Grier P. Page; Angelo D’Alessandro
| |
Collapse
|
13
|
Liang X, Cao X, Sha Q, Zhang S. HCLC-FC: A novel statistical method for phenome-wide association studies. PLoS One 2022; 17:e0276646. [PMID: 36350801 PMCID: PMC9645610 DOI: 10.1371/journal.pone.0276646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 10/11/2022] [Indexed: 11/11/2022] Open
Abstract
The emergence of genetic data coupled to longitudinal electronic medical records (EMRs) offers the possibility of phenome-wide association studies (PheWAS). In PheWAS, the whole phenome can be divided into numerous phenotypic categories according to the genetic architecture across phenotypes. Currently, statistical analyses for PheWAS are mainly univariate analyses, which test the association between one genetic variant and one phenotype at a time. In this article, we derived a novel and powerful multivariate method for PheWAS. The proposed method involves three steps. In the first step, we apply the bottom-up hierarchical clustering method to partition a large number of phenotypes into disjoint clusters within each phenotypic category. In the second step, the clustering linear combination method is used to combine test statistics within each category based on the phenotypic clusters and obtain p-values from each phenotypic category. In the third step, we propose a new false discovery rate (FDR) control approach. We perform extensive simulation studies to compare the performance of our method with that of other existing methods. The results show that our proposed method controls FDR very well and outperforms other methods we compared with. We also apply the proposed approach to a set of EMR-based phenotypes across more than 300,000 samples from the UK Biobank. We find that the proposed approach not only can well-control FDR at a nominal level but also successfully identify 1,244 significant SNPs that are reported to be associated with some phenotypes in the GWAS catalog. Our open-access tools and instructions on how to implement HCLC-FC are available at https://github.com/XiaoyuLiang/HCLCFC.
Collapse
Affiliation(s)
- Xiaoyu Liang
- Department of Preventive Medicine, Division of Biostatistics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Xuewei Cao
- Department of Mathematical Sciences, Michigan Technological University, Houghton, Michigan, United States of America
| | - Qiuying Sha
- Department of Mathematical Sciences, Michigan Technological University, Houghton, Michigan, United States of America
| | - Shuanglin Zhang
- Department of Mathematical Sciences, Michigan Technological University, Houghton, Michigan, United States of America
| |
Collapse
|
14
|
Chen Y, Chen W. Genome-Wide Integration of Genetic and Genomic Studies of Atopic Dermatitis: Insights into Genetic Architecture and Pathogenesis. J Invest Dermatol 2022; 142:2958-2967.e8. [PMID: 35577104 DOI: 10.1016/j.jid.2022.04.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 12/23/2022]
Abstract
Atopic dermatitis (AD) is a common heterogeneous, chronic, itching, and inflammatory skin disease. Genetic studies have identified multiple AD susceptibility genes. However, the genetic architecture of AD has not been elucidated. In this study, we conducted a large-scale meta-analysis of AD (35,647 cases and 1,013,885 controls) to characterize the genetic basis of AD. The heritability of AD in different datasets varied from 0.6 to 7.1%. We identified 31 previously unreported genes by integrating multiomics data. Among the 31 genes, MCL1 was identified as a potential treatment target for AD by mediating gene‒drug interactions. Tissue enrichment analyses and phenome-wide association study provided strong support for the role of the hemic and immune systems in AD. Across 1,207 complex traits and diseases, genetic correlations indicated that AD shared links with multiple respiratory phenotypes. The phenome-wide Mendelian randomization analysis (Mendelian randomization‒phenome-wide association study) revealed that the age of onset of diabetes exhibited a positive causal effect on AD (inverse-variance weighted β = 0.39, SEM = 0.09, P = 2.77 × 10-5). Overall, these results provide important insights into the genetic architecture of AD and will lead to a more thorough and complete understanding of the molecular mechanisms underlying AD.
Collapse
Affiliation(s)
- Yanxuan Chen
- Department of General Medicine, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Wenyan Chen
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, China.
| |
Collapse
|
15
|
Baca SC, Singler C, Zacharia S, Seo JH, Morova T, Hach F, Ding Y, Schwarz T, Huang CCF, Anderson J, Fay AP, Kalita C, Groha S, Pomerantz MM, Wang V, Linder S, Sweeney CJ, Zwart W, Lack NA, Pasaniuc B, Takeda DY, Gusev A, Freedman ML. Genetic determinants of chromatin reveal prostate cancer risk mediated by context-dependent gene regulation. Nat Genet 2022; 54:1364-1375. [PMID: 36071171 PMCID: PMC9784646 DOI: 10.1038/s41588-022-01168-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 07/19/2022] [Indexed: 12/25/2022]
Abstract
Many genetic variants affect disease risk by altering context-dependent gene regulation. Such variants are difficult to study mechanistically using current methods that link genetic variation to steady-state gene expression levels, such as expression quantitative trait loci (eQTLs). To address this challenge, we developed the cistrome-wide association study (CWAS), a framework for identifying genotypic and allele-specific effects on chromatin that are also associated with disease. In prostate cancer, CWAS identified regulatory elements and androgen receptor-binding sites that explained the association at 52 of 98 known prostate cancer risk loci and discovered 17 additional risk loci. CWAS implicated key developmental transcription factors in prostate cancer risk that are overlooked by eQTL-based approaches due to context-dependent gene regulation. We experimentally validated associations and demonstrated the extensibility of CWAS to additional epigenomic datasets and phenotypes, including response to prostate cancer treatment. CWAS is a powerful and biologically interpretable paradigm for studying variants that influence traits by affecting transcriptional regulation.
Collapse
Affiliation(s)
- Sylvan C. Baca
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA,The Eli and Edythe L. Broad Institute, Cambridge, MA, USA
| | - Cassandra Singler
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Soumya Zacharia
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ji-Heui Seo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Tunc Morova
- Vancouver Prostate Centre University of British Columbia, Vancouver, BC, Canada
| | - Faraz Hach
- Vancouver Prostate Centre University of British Columbia, Vancouver, BC, Canada
| | - Yi Ding
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA
| | - Tommer Schwarz
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA
| | | | - Jacob Anderson
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - André P. Fay
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Cynthia Kalita
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Division of Genetics, Brigham & Women’s Hospital, Boston, MA, USA
| | - Stefan Groha
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,The Eli and Edythe L. Broad Institute, Cambridge, MA, USA
| | - Mark M. Pomerantz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Victoria Wang
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA,Department of Biostatistics, Harvard School of Public Health, Boston, MA, USA
| | - Simon Linder
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands,Laboratory of Chemical Biology and Institute for Complex Molecular Systems, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | | | - Wilbert Zwart
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands,Laboratory of Chemical Biology and Institute for Complex Molecular Systems, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Nathan A. Lack
- Vancouver Prostate Centre University of British Columbia, Vancouver, BC, Canada,School of Medicine, Koç University, Istanbul, Turkey
| | - Bogdan Pasaniuc
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA,Department of Computational Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA USA,Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA,Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - David Y. Takeda
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Alexander Gusev
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA,The Eli and Edythe L. Broad Institute, Cambridge, MA, USA,Division of Genetics, Brigham & Women’s Hospital, Boston, MA, USA,These authors jointly supervised this work. Correspondence should be directed to M.L.F or A.G. ()
| | - 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,The Eli and Edythe L. Broad Institute, Cambridge, MA, USA,These authors jointly supervised this work. Correspondence should be directed to M.L.F or A.G. ()
| |
Collapse
|
16
|
Pacheco HA, Rossoni A, Cecchinato A, Peñagaricano F. Deciphering the genetic basis of male fertility in Italian Brown Swiss dairy cattle. Sci Rep 2022; 12:10575. [PMID: 35732705 PMCID: PMC9217806 DOI: 10.1038/s41598-022-14889-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022] Open
Abstract
Improving reproductive performance remains a major goal in dairy cattle worldwide. Service sire has been recognized as an important factor affecting herd fertility. The main objective of this study was to reveal the genetic basis of male fertility in Italian Brown Swiss dairy cattle. Dataset included 1102 Italian Brown Swiss bulls with sire conception rate records genotyped with 454k single nucleotide polymorphisms. The analysis included whole-genome scans and gene-set analyses to identify genomic regions, individual genes and genetic mechanisms affecting Brown Swiss bull fertility. One genomic region on BTA1 showed significant additive effects. This region harbors gene RABL3 which is implicated cell proliferation and motility. Two genomic regions, located on BTA6 and BTA26, showed marked non-additive effects. These regions harbor genes, such as WDR19 and ADGRA1, that are directly involved in male fertility, including sperm motility, acrosome reaction, and embryonic development. The gene-set analysis revealed functional terms related to cell adhesion, cellular signaling, cellular transport, immune system, and embryonic development. Remarkably, a gene-set analysis also including Holstein and Jersey data, revealed significant processes that are common to the three dairy breeds, including cell migration, cell-cell interaction, GTPase activity, and the immune function. Overall, this comprehensive study contributes to a better understanding of the genetic basis of male fertility in cattle. In addition, our findings may guide the development of novel genomic strategies for improving service sire fertility in Brown Swiss cattle.
Collapse
Affiliation(s)
- Hendyel A Pacheco
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Attilio Rossoni
- Italian Brown Breeders Association, Bussolengo, 37012, Verona, Italy
| | - Alessio Cecchinato
- Department of Agronomy, Food, Natural Resources, Animals and Environment, University of Padova, 35020, Legnaro, Padua, Italy
| | - Francisco Peñagaricano
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| |
Collapse
|
17
|
Aung N, Vargas JD, Yang C, Fung K, Sanghvi MM, Piechnik SK, Neubauer S, Manichaikul A, Rotter JI, Taylor KD, Lima JAC, Bluemke DA, Kawut SM, Petersen SE, Munroe PB. Genome-wide association analysis reveals insights into the genetic architecture of right ventricular structure and function. Nat Genet 2022; 54:783-791. [PMID: 35697868 PMCID: PMC11929962 DOI: 10.1038/s41588-022-01083-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 04/22/2022] [Indexed: 01/03/2023]
Abstract
Right ventricular (RV) structure and function influence the morbidity and mortality from coronary artery disease (CAD), dilated cardiomyopathy (DCM), pulmonary hypertension and heart failure. Little is known about the genetic basis of RV measurements. Here we perform genome-wide association analyses of four clinically relevant RV phenotypes (RV end-diastolic volume, RV end-systolic volume, RV stroke volume, RV ejection fraction) from cardiovascular magnetic resonance images, using a state-of-the-art deep learning algorithm in 29,506 UK Biobank participants. We identify 25 unique loci associated with at least one RV phenotype at P < 2.27 ×10-8, 17 of which are validated in a combined meta-analysis (n = 41,830). Several candidate genes overlap with Mendelian cardiomyopathy genes and are involved in cardiac muscle contraction and cellular adhesion. The RV polygenic risk scores (PRSs) are associated with DCM and CAD. The findings substantially advance our understanding of the genetic underpinning of RV measurements.
Collapse
Affiliation(s)
- Nay Aung
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research, Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London, UK
| | - Jose D Vargas
- Veterans Affairs Medical Center, Washington, DC, USA
- Georgetown University, Washington, DC, USA
| | - Chaojie Yang
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Kenneth Fung
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research, Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London, UK
| | - Mihir M Sanghvi
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research, Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London, UK
| | - Stefan K Piechnik
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Joao A C Lima
- Division of Cardiology, Johns Hopkins University, Baltimore, MD, USA
| | - David A Bluemke
- Department of Radiology, University of Wisconsin, Madison, WI, USA
| | - Steven M Kawut
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Steffen E Petersen
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
- National Institute for Health Research, Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK.
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London, UK.
| | - Patricia B Munroe
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
- National Institute for Health Research, Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK.
| |
Collapse
|
18
|
Huang Z, Chang X, Wang L, Liu J, Heng CK, Khor CC, Yuan JM, Koh WP, Dorajoo R. Interaction between cigarette smoking and genetic polymorphisms on the associations with age of natural menopause and reproductive lifespan: the Singapore Chinese Health Study. Hum Reprod 2022; 37:1351-1359. [PMID: 35413122 PMCID: PMC9156846 DOI: 10.1093/humrep/deac075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 03/09/2022] [Indexed: 11/12/2022] Open
Abstract
STUDY QUESTION Are there genetic variants that interact with smoking to reduce reproductive lifespan in East-Asian women? SUMMARY ANSWER Our study corroborates several recently identified genetic loci associated with reproductive lifespan and highlights specific genetic predispositions that may interact with smoking status to adversely affect reproductive lifespan in East-Asian women. WHAT IS KNOWN ALREADY Epidemiological data as well as evaluations on genetic predisposition to smoke indicate on the importance of smoking in adverse effects on reproductive lifespan in women. However, there are no previous smoking and gene interaction studies for reproductive traits in East-Asian women. STUDY DESIGN, SIZE, DURATION This population-based prospective cohort study comprised 11 643 East-Asian Chinese women with overlapping genome-wide genotyping and reproductive data. PARTICIPANTS/MATERIALS, SETTING, METHODS We performed a genome-wide association study for reproductive lifespan in women (n = 11 643) from the Singapore Chinese Health Study (SCHS) and carried out a genome-wide interaction study to identify loci that interacted with smoking status to affect age of natural menopause and reproductive-time. MAIN RESULTS AND THE ROLE OF CHANCE Two known loci associated with menopause, rs113430717 (near HMCES, chromosome 3, Pmeta = 5.72 × 10-15) and rs3020136 (near RAD21, chromosome 8, Pmeta = 1.38 × 10-8) were observed beyond genome-wide levels of association with age at menopause in this study. For reproductive lifespan, the genome-wide association observed at rs79784106 (chromosome 3, Pmeta = 5.05 × 10-12) was in linkage disequilibrium with the menopause lead single-nucleotide polymorphism (SNP) (rs113430717). Four additional loci, first reported to be associated with menopause, were also associated with reproductive lifespan in our study (PAdj between 7.42 × 10-5 to 4.51 × 10-3). A significant interaction was observed between smoking and an East-Asian specific SNP, rs140146885, for reduced reproductive lifespan, per copy of the minor C allele (beta = -1.417 years, Pinteraction = 2.31 × 10-10). This interaction was successfully replicated in additional independent samples (beta = -1.389 years, Pinteraction = 6.78 × 10-3). Another known variant associated with menopause, rs11031006 (near FSHB), was also observed to interact with smoking status to reduce age at menopause in our dataset (beta = -0.450 years, Padj = 0.042). LIMITATIONS, REASONS FOR CAUTION The modest sample size of the replication datasets used likely affected the statistical power to firmly replicate all identified novel loci observed in our smoking interaction analyses. WIDER IMPLICATIONS OF THE FINDINGS Age of natural menopause and reproductive lifespan have clear genetic predispositions with distinct ethnic differences, and they may be adversely truncated by lifestyle factors such as smoking, which can pose a significant impact on the reproductive lifespan and future health outcomes in women. STUDY FUNDING/COMPETING INTEREST(S) The Singapore Chinese Health Study is funded by the National Medical Research Council, Singapore (NMRC/CIRG/1456/2016), National Institutes of Health (R01 CA144034 and UM1 CA182876) and National Research Foundation, Singapore (Project Number 370062002). W.-P.K. is supported by the National Medical Research Council, Singapore (MOH-CSASI19nov-0001). The corresponding author had full access to all the data in the study and had final responsibility for the decision to submit for publication. The authors do not report conflicts of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Zhongwei Huang
- Institute of Molecular and Cell Biology, Agency of Science Research and Technology, Singapore, Singapore
- Department of Obstetrics & Gynaecology, National University Health Systems, Singapore, Singapore
- NUS Bia-Echo Asia Centre of Reproductive Longevity and Equality, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xuling Chang
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Khoo Teck Puat—National University Children’s Medical Institute, National University Health System, Singapore, Singapore
- Department of Infectious Diseases, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Ling Wang
- Genome Institute of Singapore, Agency of Science Research and Technology, Singapore, Singapore
| | - Jianjun Liu
- Genome Institute of Singapore, Agency of Science Research and Technology, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chew-Kiat Heng
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Khoo Teck Puat—National University Children’s Medical Institute, National University Health System, Singapore, Singapore
| | - Chiea-Chuen Khor
- Genome Institute of Singapore, Agency of Science Research and Technology, Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - 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
| | - Woon-Puay Koh
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Singapore Institute of Clinical Sciences, Agency of Science Research and Technology, Singapore, Singapore
| | - Rajkumar Dorajoo
- Genome Institute of Singapore, Agency of Science Research and Technology, Singapore, Singapore
- Health Services and Systems Research, Duke-NUS Medical School Singapore, Singapore, Singapore
| |
Collapse
|
19
|
Lee JM, Huang Y, Orth M, Gillis T, Siciliano J, Hong E, Mysore JS, Lucente D, Wheeler VC, Seong IS, McLean ZL, Mills JA, McAllister B, Lobanov SV, Massey TH, Ciosi M, Landwehrmeyer GB, Paulsen JS, Dorsey ER, Shoulson I, Sampaio C, Monckton DG, Kwak S, Holmans P, Jones L, MacDonald ME, Long JD, Gusella JF. Genetic modifiers of Huntington disease differentially influence motor and cognitive domains. Am J Hum Genet 2022; 109:885-899. [PMID: 35325614 DOI: 10.1016/j.ajhg.2022.03.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/01/2022] [Indexed: 12/12/2022] Open
Abstract
Genome-wide association studies (GWASs) of Huntington disease (HD) have identified six DNA maintenance gene loci (among others) as modifiers and implicated a two step-mechanism of pathogenesis: somatic instability of the causative HTT CAG repeat with subsequent triggering of neuronal damage. The largest studies have been limited to HD individuals with a rater-estimated age at motor onset. To capitalize on the wealth of phenotypic data in several large HD natural history studies, we have performed algorithmic prediction by using common motor and cognitive measures to predict age at other disease landmarks as additional phenotypes for GWASs. Combined with imputation with the Trans-Omics for Precision Medicine reference panel, predictions using integrated measures provided objective landmark phenotypes with greater power to detect most modifier loci. Importantly, substantial differences in the relative modifier signal across loci, highlighted by comparing common modifiers at MSH3 and FAN1, revealed that individual modifier effects can act preferentially in the motor or cognitive domains. Individual components of the DNA maintenance modifier mechanisms may therefore act differentially on the neuronal circuits underlying the corresponding clinical measures. In addition, we identified additional modifier effects at the PMS1 and PMS2 loci and implicated a potential second locus on chromosome 7. These findings indicate that broadened discovery and characterization of HD genetic modifiers based on additional quantitative or qualitative phenotypes offers not only the promise of in-human validated therapeutic targets but also a route to dissecting the mechanisms and cell types involved in both the somatic instability and toxicity components of HD pathogenesis.
Collapse
|
20
|
Fang F, Hazegh K, Mast AE, Triulzi DJ, Spencer BR, Gladwin MT, Busch MP, Kanias T, Page GP. Sex-specific genetic modifiers identified susceptibility of cold stored red blood cells to osmotic hemolysis. BMC Genomics 2022; 23:227. [PMID: 35321643 PMCID: PMC8941732 DOI: 10.1186/s12864-022-08461-4] [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: 12/09/2021] [Accepted: 03/04/2022] [Indexed: 11/19/2022] Open
Abstract
Background Genetic variants have been found to influence red blood cell (RBC) susceptibility to hemolytic stress and affect transfusion outcomes and the severity of blood diseases. Males have a higher susceptibility to hemolysis than females, but little is known about the genetic mechanism contributing to the difference. Results To investigate the sex differences in RBC susceptibility to hemolysis, we conducted a sex-stratified genome-wide association study and a genome-wide gene-by-sex interaction scan in a multi-ethnic dataset with 12,231 blood donors who have in vitro osmotic hemolysis measurements during routine blood storage. The estimated SNP-based heritability for osmotic hemolysis was found to be significantly higher in males than in females (0.46 vs. 0.41). We identified SNPs associated with sex-specific susceptibility to osmotic hemolysis in five loci (SPTA1, KCNA6, SLC4A1, SUMO1P1, and PAX8) that impact RBC function and hemolysis. Conclusion Our study established a best practice to identify sex-specific genetic modifiers for sexually dimorphic traits in datasets with mixed ancestries, providing evidence of different genetic regulations of RBC susceptibility to hemolysis between sexes. These and other variants may help explain observed sex differences in the severity of hemolytic diseases, such as sickle cell and malaria, as well as the viability of red cell storage and recovery. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08461-4.
Collapse
Affiliation(s)
- Fang Fang
- GenOmics, Bioinformatics, and Translational Research Center, RTI International, Research Triangle Park, Durham, NC, USA.
| | | | - Alan E Mast
- Versiti Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI, USA.,Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Darrell J Triulzi
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Mark T Gladwin
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.,Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Michael P Busch
- Vitalant Research Institute, San Francisco, CA, USA.,Department of Laboratory Medicine, UCSF, San Francisco, CA, USA
| | - Tamir Kanias
- Vitalant Research Institute, Denver, CO, USA.,Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Grier P Page
- GenOmics, Bioinformatics, and Translational Research Center, RTI International, Research Triangle Park, Durham, NC, USA.,Division of Biostatistics and Epidemiology, RTI International, GA, Atlanta, USA
| |
Collapse
|
21
|
Genome-wide pharmacogenetics of anti-drug antibody response to bococizumab highlights key residues in HLA DRB1 and DQB1. Sci Rep 2022; 12:4266. [PMID: 35277540 PMCID: PMC8917227 DOI: 10.1038/s41598-022-07997-5] [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: 12/03/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
In this largest to-date genetic analysis of anti-drug antibody (ADA) response to a therapeutic monoclonal antibody (MAb), genome-wide association was performed for five measures of ADA status among 8844 individuals randomized to bococizumab, which targets PCSK9 for LDL-C lowering and cardiovascular protection. Index associations prioritized specific amino acid substitutions at the DRB1 and DQB1 MHC class II genes rather than canonical haplotypes. Two clusters of missense variants at DRB1 were associated with general ADA measures (residues 9, 11, 13; and 96, 112, 120, 180) and a third cluster of missense variants in DQB1 was associated with ADA measures including neutralizing antibody (NAb) titers (residues 66, 67, 71, 74, 75). The structural disposition of the missense substitutions implicates peptide antigen binding and CD4 effector function, mechanisms that are potentially generalizable to other therapeutic mAbs. Clinicaltrials.gov: NCT01968954, NCT01968967, NCT01968980, NCT01975376, NCT01975389, NCT02100514.
Collapse
|
22
|
Dai H, Chu X, Liang Q, Wang M, Li L, Zhou Y, Zheng Z, Wang W, Wang Z, Li H, Wang J, Zheng H, Zhao Y, Liu L, Yao H, Luo M, Wang Q, Kang S, Li Y, Wang K, Song F, Zhang R, Wu X, Cheng X, Zhang W, Wei Q, Li MJ, Chen K. Genome-wide association and functional interrogation identified a variant at 3p26.1 modulating ovarian cancer survival among Chinese women. Cell Discov 2021; 7:121. [PMID: 34930913 PMCID: PMC8688503 DOI: 10.1038/s41421-021-00342-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 09/23/2021] [Indexed: 12/03/2022] Open
Abstract
Ovarian cancer survival varies considerably among patients, to which germline variation may also contribute in addition to mutational signatures. To identify genetic markers modulating ovarian cancer outcome, we performed a genome-wide association study in 2130 Chinese ovarian cancer patients and found a hitherto unrecognized locus at 3p26.1 to be associated with the overall survival (Pcombined = 8.90 × 10−10). Subsequent statistical fine-mapping, functional annotation, and eQTL mapping prioritized a likely casual SNP rs9311399 in the non-coding regulatory region. Mechanistically, rs9311399 altered its enhancer activity through an allele-specific transcription factor binding and a long-range interaction with the promoter of a lncRNA BHLHE40-AS1. Deletion of the rs9311399-associated enhancer resulted in expression changes in several oncogenic signaling pathway genes and a decrease in tumor growth. Thus, we have identified a novel genetic locus that is associated with ovarian cancer survival possibly through a long-range gene regulation of oncogenic pathways.
Collapse
Affiliation(s)
- Hongji Dai
- Department of Epidemiology and Biostatistics, National Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Xinlei Chu
- Department of Epidemiology and Biostatistics, National Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Qian Liang
- Department of Pharmacology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Mengyun Wang
- Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lian Li
- Department of Epidemiology and Biostatistics, National Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Yao Zhou
- Department of Pharmacology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zhanye Zheng
- Department of Pharmacology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Wei Wang
- Department of Epidemiology and Biostatistics, National Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Zhao Wang
- Department of Pharmacology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Haixin Li
- Cancer Biobank, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Jianhua Wang
- Department of Pharmacology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hong Zheng
- Department of Epidemiology and Biostatistics, National Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Yanrui Zhao
- Department of Epidemiology and Biostatistics, National Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Luyang Liu
- Department of Epidemiology and Biostatistics, National Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Hongcheng Yao
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Menghan Luo
- Department of Pharmacology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Qiong Wang
- Department of Epidemiology and Biostatistics, National Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Shan Kang
- Department of Obstetrics and Gynaecology, Hebei Medical University, Fourth Hospital, Shijiazhuang, China
| | - Yan Li
- Department of Molecular Biology, Hebei Medical University, Fourth Hospital, Shijiazhuang, China
| | - Ke Wang
- Department of Gynecologic Oncology, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Fengju Song
- Department of Epidemiology and Biostatistics, National Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Ruoxin Zhang
- Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaohua Wu
- Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xi Cheng
- Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Zhang
- Center for Cancer Genomics and Precision Oncology, Wake Forest Baptist Comprehensive Cancer Center, Wake Forest Baptist Medical Center, Winston-Salem, NC, USA.,Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Qingyi Wei
- Cancer Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China. .,Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA. .,Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, USA.
| | - Mulin Jun Li
- Department of Epidemiology and Biostatistics, National Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China. .,Department of Pharmacology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, National Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
23
|
Li-Gao R, Hughes DA, van Klinken JB, de Mutsert R, Rosendaal FR, Mook-Kanamori DO, Timpson NJ, Willems van Dijk K. Genetic Studies of Metabolomics Change After a Liquid Meal Illuminate Novel Pathways for Glucose and Lipid Metabolism. Diabetes 2021; 70:2932-2946. [PMID: 34610981 DOI: 10.2337/db21-0397] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 09/27/2021] [Indexed: 11/13/2022]
Abstract
Humans spend the greater part of the day in a postprandial state. However, the genetic basis of postprandial blood measures is relatively uncharted territory. We examined the genetics of variation in concentrations of postprandial metabolites (t = 150 min) in response to a liquid mixed meal through genome-wide association studies (GWAS) performed in the Netherlands Epidemiology of Obesity (NEO) study (n = 5,705). The metabolite response GWAS identified an association between glucose change and rs10830963:G in the melatonin receptor 1B (β [SE] -0.23 [0.03], P = 2.15 × 10-19). In addition, the ANKRD55 locus led by rs458741:C showed strong associations with extremely large VLDL (XXLVLDL) particle response (XXLVLDL total cholesterol: β [SE] 0.17 [0.03], P = 5.76 × 10-10; XXLVLDL cholesterol ester: β [SE] 0.17 [0.03], P = 9.74 × 10-10), which also revealed strong associations with body composition and diabetes in the UK Biobank (P < 5 × 10-8). Furthermore, the associations between XXLVLDL response and insulinogenic index, HOMA-β, Matsuda insulin sensitivity index, and HbA1c in the NEO study implied the role of chylomicron synthesis in diabetes (with false discovery rate-corrected q <0.05). To conclude, genetic studies of metabolomics change after a liquid meal illuminate novel pathways for glucose and lipid metabolism. Further studies are warranted to corroborate biological pathways of the ANKRD55 locus underlying diabetes.
Collapse
Affiliation(s)
- Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - David A Hughes
- Medical Research Council Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, U.K
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, U.K
| | - Jan B van Klinken
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, the Netherlands
| | - Nicholas J Timpson
- Medical Research Council Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, U.K
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, U.K
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
24
|
Guo Y, Daghlas I, Gormley P, Giulianini F, Ridker PM, Mora S, Kurth T, Rist PM, Chasman DI. Phenotypic and Genotypic Associations Between Migraine and Lipoprotein Subfractions. Neurology 2021; 97:e2223-e2235. [PMID: 34635557 DOI: 10.1212/wnl.0000000000012919] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 09/20/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVE To evaluate phenotypic and genetic relationships between migraine and lipoprotein subfractions. METHODS We evaluated phenotypic associations between migraine and 19 lipoprotein subfraction measures in the Women's Genome Health Study (n = 22,788). We then investigated genetic relationships between these traits using summary statistics from the International Headache Genetics Consortium for migraine (ncase = 54,552, ncontrol = 297,970) and combined summary data for lipoprotein subfractions (n up to 47,713). RESULTS There was a significant phenotypic association (odds ratio 1.27 [95% confidence interval 1.12-1.44]) and a significant genetic correlation at 0.18 (p = 0.001) between migraine and triglyceride-rich lipoproteins (TRLPs) concentration but not for low-density lipoprotein or high-density lipoprotein subfractions. Mendelian randomization (MR) estimates were largely null, implying that pleiotropy rather than causality underlies the genetic correlation between migraine and lipoprotein subfractions. Pleiotropy was further supported in cross-trait meta-analysis, revealing significant shared signals at 4 loci (chr2p21 harboring THADA, chr5q13.3 harboring HMGCR, chr6q22.31 harboring HEY2, and chr7q11.23 harboring MLXIPL) between migraine and lipoprotein subfractions. Three of these loci were replicated for migraine (p < 0.05) in a smaller sample from the UK Biobank. The shared signal at chr5q13.3 colocalized with expression of HMGCR, ANKDD1B, and COL4A3BP in multiple tissues. CONCLUSIONS The study supports the association between certain lipoprotein subfractions, especially for TRLP, and migraine in populations of European ancestry. The corresponding shared genetic components may help identify potential targets for future migraine therapeutics. CLASSIFICATION OF EVIDENCE This study provides Class I evidence that migraine is significantly associated with some lipoprotein subfractions.
Collapse
Affiliation(s)
- Yanjun Guo
- From the Division of Preventive Medicine (Y.G., I.D., F.G., P.M. Ridker, S.M., P.M. Rist, D.I.C.), Center for Cardiovascular Disease Prevention (P.M. Ridker, S.M., D.I.C.), and Center for Lipid Metabolomics (S.M.), Brigham and Women's Hospital; Harvard Medical School (Y.G., I.D., P.M. Ridker, S.M., P.M. Rist, D.I.C.); Department of Epidemiology (Y.G., T.K., P.R., D.I.C.), Harvard T.H. Chan School of Public Health; Genetics and Pharmacogenomics (P.G.), Merck & Co., Inc., Boston, MA; and Institute of Public Health (T.K.), Charité Universitätsmedizin Berlin, Germany
| | - Iyas Daghlas
- From the Division of Preventive Medicine (Y.G., I.D., F.G., P.M. Ridker, S.M., P.M. Rist, D.I.C.), Center for Cardiovascular Disease Prevention (P.M. Ridker, S.M., D.I.C.), and Center for Lipid Metabolomics (S.M.), Brigham and Women's Hospital; Harvard Medical School (Y.G., I.D., P.M. Ridker, S.M., P.M. Rist, D.I.C.); Department of Epidemiology (Y.G., T.K., P.R., D.I.C.), Harvard T.H. Chan School of Public Health; Genetics and Pharmacogenomics (P.G.), Merck & Co., Inc., Boston, MA; and Institute of Public Health (T.K.), Charité Universitätsmedizin Berlin, Germany
| | - Padhraig Gormley
- From the Division of Preventive Medicine (Y.G., I.D., F.G., P.M. Ridker, S.M., P.M. Rist, D.I.C.), Center for Cardiovascular Disease Prevention (P.M. Ridker, S.M., D.I.C.), and Center for Lipid Metabolomics (S.M.), Brigham and Women's Hospital; Harvard Medical School (Y.G., I.D., P.M. Ridker, S.M., P.M. Rist, D.I.C.); Department of Epidemiology (Y.G., T.K., P.R., D.I.C.), Harvard T.H. Chan School of Public Health; Genetics and Pharmacogenomics (P.G.), Merck & Co., Inc., Boston, MA; and Institute of Public Health (T.K.), Charité Universitätsmedizin Berlin, Germany
| | - Franco Giulianini
- From the Division of Preventive Medicine (Y.G., I.D., F.G., P.M. Ridker, S.M., P.M. Rist, D.I.C.), Center for Cardiovascular Disease Prevention (P.M. Ridker, S.M., D.I.C.), and Center for Lipid Metabolomics (S.M.), Brigham and Women's Hospital; Harvard Medical School (Y.G., I.D., P.M. Ridker, S.M., P.M. Rist, D.I.C.); Department of Epidemiology (Y.G., T.K., P.R., D.I.C.), Harvard T.H. Chan School of Public Health; Genetics and Pharmacogenomics (P.G.), Merck & Co., Inc., Boston, MA; and Institute of Public Health (T.K.), Charité Universitätsmedizin Berlin, Germany
| | - Paul M Ridker
- From the Division of Preventive Medicine (Y.G., I.D., F.G., P.M. Ridker, S.M., P.M. Rist, D.I.C.), Center for Cardiovascular Disease Prevention (P.M. Ridker, S.M., D.I.C.), and Center for Lipid Metabolomics (S.M.), Brigham and Women's Hospital; Harvard Medical School (Y.G., I.D., P.M. Ridker, S.M., P.M. Rist, D.I.C.); Department of Epidemiology (Y.G., T.K., P.R., D.I.C.), Harvard T.H. Chan School of Public Health; Genetics and Pharmacogenomics (P.G.), Merck & Co., Inc., Boston, MA; and Institute of Public Health (T.K.), Charité Universitätsmedizin Berlin, Germany
| | - Samia Mora
- From the Division of Preventive Medicine (Y.G., I.D., F.G., P.M. Ridker, S.M., P.M. Rist, D.I.C.), Center for Cardiovascular Disease Prevention (P.M. Ridker, S.M., D.I.C.), and Center for Lipid Metabolomics (S.M.), Brigham and Women's Hospital; Harvard Medical School (Y.G., I.D., P.M. Ridker, S.M., P.M. Rist, D.I.C.); Department of Epidemiology (Y.G., T.K., P.R., D.I.C.), Harvard T.H. Chan School of Public Health; Genetics and Pharmacogenomics (P.G.), Merck & Co., Inc., Boston, MA; and Institute of Public Health (T.K.), Charité Universitätsmedizin Berlin, Germany
| | - Tobias Kurth
- From the Division of Preventive Medicine (Y.G., I.D., F.G., P.M. Ridker, S.M., P.M. Rist, D.I.C.), Center for Cardiovascular Disease Prevention (P.M. Ridker, S.M., D.I.C.), and Center for Lipid Metabolomics (S.M.), Brigham and Women's Hospital; Harvard Medical School (Y.G., I.D., P.M. Ridker, S.M., P.M. Rist, D.I.C.); Department of Epidemiology (Y.G., T.K., P.R., D.I.C.), Harvard T.H. Chan School of Public Health; Genetics and Pharmacogenomics (P.G.), Merck & Co., Inc., Boston, MA; and Institute of Public Health (T.K.), Charité Universitätsmedizin Berlin, Germany
| | - Pamela M Rist
- From the Division of Preventive Medicine (Y.G., I.D., F.G., P.M. Ridker, S.M., P.M. Rist, D.I.C.), Center for Cardiovascular Disease Prevention (P.M. Ridker, S.M., D.I.C.), and Center for Lipid Metabolomics (S.M.), Brigham and Women's Hospital; Harvard Medical School (Y.G., I.D., P.M. Ridker, S.M., P.M. Rist, D.I.C.); Department of Epidemiology (Y.G., T.K., P.R., D.I.C.), Harvard T.H. Chan School of Public Health; Genetics and Pharmacogenomics (P.G.), Merck & Co., Inc., Boston, MA; and Institute of Public Health (T.K.), Charité Universitätsmedizin Berlin, Germany
| | - Daniel I Chasman
- From the Division of Preventive Medicine (Y.G., I.D., F.G., P.M. Ridker, S.M., P.M. Rist, D.I.C.), Center for Cardiovascular Disease Prevention (P.M. Ridker, S.M., D.I.C.), and Center for Lipid Metabolomics (S.M.), Brigham and Women's Hospital; Harvard Medical School (Y.G., I.D., P.M. Ridker, S.M., P.M. Rist, D.I.C.); Department of Epidemiology (Y.G., T.K., P.R., D.I.C.), Harvard T.H. Chan School of Public Health; Genetics and Pharmacogenomics (P.G.), Merck & Co., Inc., Boston, MA; and Institute of Public Health (T.K.), Charité Universitätsmedizin Berlin, Germany.
| |
Collapse
|
25
|
Wang H, Noordam R, Cade BE, Schwander K, Winkler TW, Lee J, Sung YJ, Bentley AR, Manning AK, Aschard H, Kilpeläinen TO, Ilkov M, Brown MR, Horimoto AR, Richard M, Bartz TM, Vojinovic D, Lim E, Nierenberg JL, Liu Y, Chitrala K, Rankinen T, Musani SK, Franceschini N, Rauramaa R, Alver M, Zee PC, Harris SE, van der Most PJ, Nolte IM, Munroe PB, Palmer ND, Kühnel B, Weiss S, Wen W, Hall KA, Lyytikäinen LP, O'Connell J, Eiriksdottir G, Launer LJ, de Vries PS, Arking DE, Chen H, Boerwinkle E, Krieger JE, Schreiner PJ, Sidney S, Shikany JM, Rice K, Chen YDI, Gharib SA, Bis JC, Luik AI, Ikram MA, Uitterlinden AG, Amin N, Xu H, Levy D, He J, Lohman KK, Zonderman AB, Rice TK, Sims M, Wilson G, Sofer T, Rich SS, Palmas W, Yao J, Guo X, Rotter JI, Biermasz NR, Mook-Kanamori DO, Martin LW, Barac A, Wallace RB, Gottlieb DJ, Komulainen P, Heikkinen S, Mägi R, Milani L, Metspalu A, Starr JM, Milaneschi Y, Waken RJ, Gao C, Waldenberger M, Peters A, Strauch K, Meitinger T, Roenneberg T, Völker U, Dörr M, Shu XO, Mukherjee S, Hillman DR, Kähönen M, Wagenknecht LE, Gieger C, Grabe HJ, Zheng W, et alWang H, Noordam R, Cade BE, Schwander K, Winkler TW, Lee J, Sung YJ, Bentley AR, Manning AK, Aschard H, Kilpeläinen TO, Ilkov M, Brown MR, Horimoto AR, Richard M, Bartz TM, Vojinovic D, Lim E, Nierenberg JL, Liu Y, Chitrala K, Rankinen T, Musani SK, Franceschini N, Rauramaa R, Alver M, Zee PC, Harris SE, van der Most PJ, Nolte IM, Munroe PB, Palmer ND, Kühnel B, Weiss S, Wen W, Hall KA, Lyytikäinen LP, O'Connell J, Eiriksdottir G, Launer LJ, de Vries PS, Arking DE, Chen H, Boerwinkle E, Krieger JE, Schreiner PJ, Sidney S, Shikany JM, Rice K, Chen YDI, Gharib SA, Bis JC, Luik AI, Ikram MA, Uitterlinden AG, Amin N, Xu H, Levy D, He J, Lohman KK, Zonderman AB, Rice TK, Sims M, Wilson G, Sofer T, Rich SS, Palmas W, Yao J, Guo X, Rotter JI, Biermasz NR, Mook-Kanamori DO, Martin LW, Barac A, Wallace RB, Gottlieb DJ, Komulainen P, Heikkinen S, Mägi R, Milani L, Metspalu A, Starr JM, Milaneschi Y, Waken RJ, Gao C, Waldenberger M, Peters A, Strauch K, Meitinger T, Roenneberg T, Völker U, Dörr M, Shu XO, Mukherjee S, Hillman DR, Kähönen M, Wagenknecht LE, Gieger C, Grabe HJ, Zheng W, Palmer LJ, Lehtimäki T, Gudnason V, Morrison AC, Pereira AC, Fornage M, Psaty BM, van Duijn CM, Liu CT, Kelly TN, Evans MK, Bouchard C, Fox ER, Kooperberg C, Zhu X, Lakka TA, Esko T, North KE, Deary IJ, Snieder H, Penninx BWJH, Gauderman WJ, Rao DC, Redline S, van Heemst D. Multi-ancestry genome-wide gene-sleep interactions identify novel loci for blood pressure. Mol Psychiatry 2021; 26:6293-6304. [PMID: 33859359 PMCID: PMC8517040 DOI: 10.1038/s41380-021-01087-0] [Show More Authors] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 03/18/2021] [Accepted: 03/29/2021] [Indexed: 02/02/2023]
Abstract
Long and short sleep duration are associated with elevated blood pressure (BP), possibly through effects on molecular pathways that influence neuroendocrine and vascular systems. To gain new insights into the genetic basis of sleep-related BP variation, we performed genome-wide gene by short or long sleep duration interaction analyses on four BP traits (systolic BP, diastolic BP, mean arterial pressure, and pulse pressure) across five ancestry groups in two stages using 2 degree of freedom (df) joint test followed by 1df test of interaction effects. Primary multi-ancestry analysis in 62,969 individuals in stage 1 identified three novel gene by sleep interactions that were replicated in an additional 59,296 individuals in stage 2 (stage 1 + 2 Pjoint < 5 × 10-8), including rs7955964 (FIGNL2/ANKRD33) that increases BP among long sleepers, and rs73493041 (SNORA26/C9orf170) and rs10406644 (KCTD15/LSM14A) that increase BP among short sleepers (Pint < 5 × 10-8). Secondary ancestry-specific analysis identified another novel gene by long sleep interaction at rs111887471 (TRPC3/KIAA1109) in individuals of African ancestry (Pint = 2 × 10-6). Combined stage 1 and 2 analyses additionally identified significant gene by long sleep interactions at 10 loci including MKLN1 and RGL3/ELAVL3 previously associated with BP, and significant gene by short sleep interactions at 10 loci including C2orf43 previously associated with BP (Pint < 10-3). 2df test also identified novel loci for BP after modeling sleep that has known functions in sleep-wake regulation, nervous and cardiometabolic systems. This study indicates that sleep and primary mechanisms regulating BP may interact to elevate BP level, suggesting novel insights into sleep-related BP regulation.
Collapse
Affiliation(s)
- Heming Wang
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA.
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Brian E Cade
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Karen Schwander
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Thomas W Winkler
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Jiwon Lee
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Joint Carnegie Mellon University-University of Pittsburgh PhD Program in Computational Biology, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Pittsburgh Center for Evolutionary Biology and Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yun Ju Sung
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alisa K Manning
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hugues Aschard
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
- Centre de Bioinformatique, Biostatistique et Biologie Intégrative (C3BI), Institut Pasteur, Paris, France
| | - Tuomas O Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Environmental Medicine and Public Health, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Michael R Brown
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Andrea R Horimoto
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Melissa Richard
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Dina Vojinovic
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Elise Lim
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Jovia L Nierenberg
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Yongmei Liu
- Division of Cardiology, Department of Medicine, Duke Molecular Physiology Institute Duke University School of Medicine, Durham, NC, USA
| | - Kumaraswamynaidu Chitrala
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Solomon K Musani
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Rainer Rauramaa
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Maris Alver
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | - Phyllis C Zee
- Division of Sleep Medicine, Department of Neurology, Northwestern University, Chicago, IL, USA
| | - Sarah E Harris
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Peter J van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ilja M Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Patricia B Munroe
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Unit, Queen Mary University of London, London, London, UK
| | | | - Brigitte Kühnel
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- German Center for Cardiovascular Research (DZHK), partner site Greifswald, Greifswald, Germany
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Kelly A Hall
- School of Public Health, The University of Adelaide, Adelaide, SA, Australia
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jeff O'Connell
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
- Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Dan E Arking
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Han Chen
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Precision Health, School of Public Health & School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Jose E Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Pamela J Schreiner
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | | | - James M Shikany
- Division of Preventive Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kenneth Rice
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Sina A Gharib
- Computational Medicine Core, Center for Lung Biology, UW Medicine Sleep Center, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Annemarie I Luik
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Najaf Amin
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Hanfei Xu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Daniel Levy
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, MA, USA
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Kurt K Lohman
- Division of Cardiology, Department of Medicine, Duke Molecular Physiology Institute Duke University School of Medicine, Durham, NC, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Treva K Rice
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Mario Sims
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Gregory Wilson
- JHS Graduate Training and Education Center, Jackson State University, Jackson, MS, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Walter Palmas
- Division of General Medicine, Department of Medicine, Columbia University, New York, NY, USA
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Nienke R Biermasz
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, The Netherlands
| | - Lisa W Martin
- George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Ana Barac
- MedStar Heart and Vascular Institute, Washington, DC, USA
| | - Robert B Wallace
- Department of Epidemiology, University of Iowa College of Public Health, Iowa City, IA, USA
| | - Daniel J Gottlieb
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - Pirjo Komulainen
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Sami Heikkinen
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus, Finland
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Reedik Mägi
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Lili Milani
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Andres Metspalu
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - John M Starr
- Alzheimer Scotland Dementia Research Centre, The University of Edinburgh, Edinburgh, UK
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam UMC, Vrije Universiteit, Amsterdam, HJ, The Netherlands
| | - R J Waken
- Division of Cardiology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Chuan Gao
- Molecular Genetics and Genomics Program, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Annette Peters
- 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, Neuherberg, Germany
| | - Konstantin Strauch
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
- 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
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Till Roenneberg
- Institute and Polyclinic for Occupational-, Social- and Environmental Medicine, LMU Munich, Munich, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- German Center for Cardiovascular Research (DZHK), partner site Greifswald, Greifswald, Germany
| | - Marcus Dörr
- German Center for Cardiovascular Research (DZHK), partner site Greifswald, Greifswald, Germany
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Sutapa Mukherjee
- Sleep Health Service, Respiratory and Sleep Services, Southern Adelaide Local Health Network, Adelaide, SA, Australia
- Adelaide Institute for Sleep Health, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - David R Hillman
- Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
- Department of Clinical Physiology, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Lynne E Wagenknecht
- Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Hans J Grabe
- Department Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Lyle J Palmer
- School of Public Health, The University of Adelaide, Adelaide, SA, Australia
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Alexandre C Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Myriam Fornage
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Cardiovascular Health Research Unit, Departments of Epidemiology and Health Services, University of Washington, Seattle, WA, USA
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Tanika N Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Ervin R Fox
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Xiaofeng Zhu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Timo A Lakka
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus, Finland
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Tõnu Esko
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Ian J Deary
- Lothian Birth Cohorts group, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam UMC, Vrije Universiteit, Amsterdam, HJ, The Netherlands
| | - W James Gauderman
- Division of Biostatistics, Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Dabeeru C Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Pulmonary Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Diana van Heemst
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
26
|
McDonough CW, Warren HR, Jack JR, Motsinger-Reif AA, Armstrong ND, Bis JC, House JS, Singh S, El Rouby NM, Gong Y, Mychaleckyj JC, Rotroff DM, Benavente OR, Caulfield MJ, Doria A, Pepine CJ, Psaty BM, Glorioso V, Glorioso N, Hiltunen TP, Kontula KK, Arnett DK, Buse JB, Irvin MR, Johnson JA, Munroe PB, Wagner MJ, Cooper-DeHoff RM. Adverse Cardiovascular Outcomes and Antihypertensive Treatment: A Genome-Wide Interaction Meta-Analysis in the International Consortium for Antihypertensive Pharmacogenomics Studies. Clin Pharmacol Ther 2021; 110:723-732. [PMID: 34231218 PMCID: PMC8672325 DOI: 10.1002/cpt.2355] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 06/11/2021] [Indexed: 01/01/2023]
Abstract
We sought to identify genome-wide variants influencing antihypertensive drug response and adverse cardiovascular outcomes, utilizing data from four randomized controlled trials in the International Consortium for Antihypertensive Pharmacogenomics Studies (ICAPS). Genome-wide antihypertensive drug-single nucleotide polymorphism (SNP) interaction tests for four drug classes (β-blockers, n = 9,195; calcium channel blockers (CCBs), n = 10,511; thiazide/thiazide-like diuretics, n = 3,516; ACE-inhibitors/ARBs, n = 2,559) and cardiovascular outcomes (incident myocardial infarction, stroke, or death) were analyzed among patients with hypertension of European ancestry. Top SNPs from the meta-analyses were tested for replication of cardiovascular outcomes in an independent Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) study (n = 21,267), blood pressure (BP) response in independent ICAPS studies (n = 1,552), and ethnic validation in African Americans from the Genetics of Hypertension Associated Treatment study (GenHAT; n = 5,115). One signal reached genome-wide significance in the β-blocker-SNP interaction analysis (rs139945292, Interaction P = 1.56 × 10-8 ). rs139945292 was validated through BP response to β-blockers, with the T-allele associated with less BP reduction (systolic BP response P = 6 × 10-4 , Beta = 3.09, diastolic BP response P = 5 × 10-3 , Beta = 1.53). The T-allele was also associated with increased adverse cardiovascular risk within the β-blocker treated patients' subgroup (P = 2.35 × 10-4 , odds ratio = 1.57, 95% confidence interval = 1.23-1.99). The locus showed nominal replication in CHARGE, and consistent directional trends in β-blocker treated African Americans. rs139945292 is an expression quantitative trait locus for the 50 kb upstream gene NTM (neurotrimin). No SNPs attained genome-wide significance for any other drugs classes. Top SNPs were located near CALB1 (CCB), FLJ367777 (ACE-inhibitor), and CES5AP1 (thiazide). The NTM region is associated with increased risk for adverse cardiovascular outcomes and less BP reduction in β-blocker treated patients. Further investigation into this region is warranted.
Collapse
Affiliation(s)
- Caitrin W. McDonough
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Helen R. Warren
- Clinical Pharmacology Department, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- NIHR Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - John R. Jack
- Bioinformatics Research Center, Department of Statistics, North Carolina State University, Raleigh, North Carolina, USA
| | - Alison A. Motsinger-Reif
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| | - Nicole D. Armstrong
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Joshua C. Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - John S. House
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| | - Sonal Singh
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Nihal M. El Rouby
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Yan Gong
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Joesyf C. Mychaleckyj
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Daniel M. Rotroff
- Bioinformatics Research Center, Department of Statistics, North Carolina State University, Raleigh, North Carolina, USA
| | - Oscar R. Benavente
- Department of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mark J. Caulfield
- National Institute for Health Research, Barts Cardiovascular Biomedical Research Center, Queen Mary University of London, London, UK
| | - Alessandrio Doria
- Research Division, Joslin Diabetes Center; and Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Carl J. Pepine
- Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Valeria Glorioso
- Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milano, Italy
| | - Nicola Glorioso
- Department of Clinical, Surgical and Experimental Science, University of Sassari, Medical School, Sassari, Italy
| | - Timo P. Hiltunen
- Department of Medicine and Research Program for Clinical and Molecular Metabolism, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Kimmo K. Kontula
- Department of Medicine and Research Program for Clinical and Molecular Metabolism, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Donna K. Arnett
- College of Public Health, Dean’s Office, University of Kentucky, Lexington, Kentucky, USA
| | - John B. Buse
- Division of Endocrinology, Department of Medicine, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Marguerite R. Irvin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Julie A. Johnson
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
- Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Patricia B. Munroe
- Clinical Pharmacology Department, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- NIHR Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Michael J. Wagner
- Center for Pharmacogenomics and Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Rhonda M. Cooper-DeHoff
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
- Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
27
|
Freitag-Wolf S, Munz M, Junge O, Graetz C, Jockel-Schneider Y, Staufenbiel I, Bruckmann C, Lieb W, Franke A, Loos BG, Jepsen S, Dommisch H, Schaefer AS. Sex-specific genetic factors affect the risk of early-onset periodontitis in Europeans. J Clin Periodontol 2021; 48:1404-1413. [PMID: 34409643 DOI: 10.1111/jcpe.13538] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 07/03/2021] [Indexed: 12/21/2022]
Abstract
AIMS Various studies have reported that young European women are more likely to develop early-onset periodontitis compared to men. A potential explanation for the observed variations in sex and age of disease onset is the natural genetic variation within the autosomal genomes. We hypothesized that genotype-by-sex (G × S) interactions contribute to the increased prevalence and severity. MATERIALS AND METHODS Using the case-only design, we tested for differences in genetic effects between men and women in 896 North-West European early-onset cases, using imputed genotypes from the OmniExpress genotyping array. Population-representative 6823 controls were used to verify that the interacting variables G and S were uncorrelated in the general population. RESULTS In total, 20 loci indicated G × S associations (P < 0.0005), 3 of which were previously suggested as risk genes for periodontitis (ABLIM2, CDH13, and NELL1). We also found independent G × S interactions of the related gene paralogs MACROD1/FLRT1 (chr11) and MACROD2/FLRT3 (chr20). G × S-associated SNPs at CPEB4, CDH13, MACROD1, and MECOM were genome-wide-associated with heel bone mineral density (CPEB4, MECOM), waist-to-hip ratio (CPEB4, MACROD1), and blood pressure (CPEB4, CDH13). CONCLUSIONS Our results indicate that natural genetic variation affects the different heritability of periodontitis among sexes and suggest genes that contribute to inter-sex phenotypic variation in early-onset periodontitis.
Collapse
Affiliation(s)
- Sandra Freitag-Wolf
- Institute of Medical Informatics and Statistics, Kiel University, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Matthias Munz
- Department of Periodontology, Oral Medicine and Oral Surgery, Charité - University Medicine Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute for Dental and Craniofacial Sciences, Berlin, Germany
| | - Olaf Junge
- Institute of Medical Informatics and Statistics, Kiel University, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Christian Graetz
- Department of Conservative Dentistry, Unit of Periodontology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Yvonne Jockel-Schneider
- Department of Periodontology, Clinic of Preventive Dentistry and Periodontology, University Medical Center of the Julius-Maximilians-University, Würzburg, Germany
| | - Ingmar Staufenbiel
- Department of Conservative Dentistry, Periodontology and Preventive Dentistry, Hannover Medical School, Hannover, Germany
| | - Corinna Bruckmann
- Department of Conservative Dentistry and Periodontology, School of Dentistry, Medical University Vienna, Vienna, Austria
| | - Wolfgang Lieb
- Institute of Epidemiology, Christian-Albrechts University, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts University, Kiel, Germany
| | - Bruno G Loos
- Department of Periodontology and Oral Biochemistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Søren Jepsen
- Department of Periodontology, Operative and Preventive Dentistry, University of Bonn, Bonn, Germany
| | - Henrik Dommisch
- Department of Periodontology, Oral Medicine and Oral Surgery, Charité - University Medicine Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute for Dental and Craniofacial Sciences, Berlin, Germany
| | - Arne S Schaefer
- Department of Periodontology, Oral Medicine and Oral Surgery, Charité - University Medicine Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute for Dental and Craniofacial Sciences, Berlin, Germany
| |
Collapse
|
28
|
Ruggiero D, Nutile T, Nappo S, Tirozzi A, Bellenguez C, Leutenegger AL, Ciullo M. Genetics of PlGF plasma levels highlights a role of its receptors and supports the link between angiogenesis and immunity. Sci Rep 2021; 11:16821. [PMID: 34413389 PMCID: PMC8376970 DOI: 10.1038/s41598-021-96256-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 07/28/2021] [Indexed: 11/29/2022] Open
Abstract
Placental growth factor (PlGF) is a member of the vascular endothelial growth factor family and is involved in bone marrow-derived cell activation, endothelial stimulation and pathological angiogenesis. High levels of PlGF have been observed in several pathological conditions especially in cancer, cardiovascular, autoimmune and inflammatory diseases. Little is known about the genetics of circulating PlGF levels. Indeed, although the heritability of circulating PlGF levels is around 40%, no studies have assessed the relation between PlGF plasma levels and genetic variants at a genome-wide level. In the current study, PlGF plasma levels were measured in a population-based sample of 2085 adult individuals from three isolated populations of South Italy. A GWAS was performed in a discovery cohort (N = 1600), followed by a de novo replication (N = 468) from the same populations. The meta-analysis of the discovery and replication samples revealed one signal significantly associated with PlGF circulating levels. This signal was mapped to the PlGF co-receptor coding gene NRP1, indicating its important role in modulating the PlGF plasma levels. Two additional signals, at the PlGF receptor coding gene FLT1 and RAPGEF5 gene, were identified at a suggestive level. Pathway and TWAS analyses highlighted genes known to be involved in angiogenesis and immune response, supporting the link between these processes and PlGF regulation. Overall, these data improve our understanding of the genetic variation underlying circulating PlGF levels. This in turn could lead to new preventive and therapeutic strategies for a wide variety of PlGF-related pathologies.
Collapse
Affiliation(s)
- Daniela Ruggiero
- Institute of Genetics and Biophysics "A. Buzzati-Traverso", National Research Council of Italy (CNR), Via Pietro Castellino, 111, 80131, Naples, Italy.
- IRCCS Neuromed, Pozzilli, Isernia, Italy.
| | - Teresa Nutile
- Institute of Genetics and Biophysics "A. Buzzati-Traverso", National Research Council of Italy (CNR), Via Pietro Castellino, 111, 80131, Naples, Italy
| | | | | | - Celine Bellenguez
- CHU Lille, U1167 - Labex DISTALZ - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Inserm, Institut Pasteur de Lille, Univ. Lille, 59000, Lille, France
| | - Anne-Louise Leutenegger
- UMR 946, Genetic Variation and Human Diseases, Inserm, 75010, Paris, France
- UMR946, Université Paris-Diderot, Sorbonne Paris Cité, 75010, Paris, France
| | - Marina Ciullo
- Institute of Genetics and Biophysics "A. Buzzati-Traverso", National Research Council of Italy (CNR), Via Pietro Castellino, 111, 80131, Naples, Italy.
- IRCCS Neuromed, Pozzilli, Isernia, Italy.
| |
Collapse
|
29
|
Sliz E, Shin J, Syme C, Patel Y, Parker N, Richer L, Gaudet D, Bennett S, Paus T, Pausova Z. A variant near DHCR24 associates with microstructural properties of white matter and peripheral lipid metabolism in adolescents. Mol Psychiatry 2021; 26:3795-3805. [PMID: 31900429 PMCID: PMC7332371 DOI: 10.1038/s41380-019-0640-9] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 12/01/2019] [Accepted: 12/12/2019] [Indexed: 12/17/2022]
Abstract
Visceral adiposity has been associated with altered microstructural properties of white matter in adolescents. Previous evidence suggests that circulating phospholipid PC(16:0/2:0) may mediate this association. To investigate the underlying biology, we performed a genome-wide association study (GWAS) of the shared variance of visceral fat, PC(16:0/2:0), and white matter microstructure in 872 adolescents from the Saguenay Youth Study. We further studied the metabolomic profile of the GWAS-lead variant in 931 adolescents. Visceral fat and white matter microstructure were assessed with magnetic resonance imaging. Circulating metabolites were quantified with serum lipidomics and metabolomics. We identified a genome-wide significant association near DHCR24 (Seladin-1) encoding a cholesterol-synthesizing enzyme (rs588709, p = 3.6 × 10-8); rs588709 was also associated nominally with each of the three traits (white matter microstructure: p = 2.1 × 10-6, PC(16:0/2:0): p = 0.005, visceral fat: p = 0.010). We found that the metabolic profile associated with rs588709 resembled that of a TM6SF2 variant impacting very low-density lipoprotein (VLDL) secretion and was only partially similar to that of a HMGCR variant. This suggests that the effect of rs588709 on VLDL lipids may arise due to altered phospholipid rather than cholesterol metabolism. The rs588709 was also nominally associated with circulating concentrations of omega-3 fatty acids in interaction with visceral fat and PC(16:0/2:0), and these fatty acid measures showed robust associations with white matter microstructure. Overall, the present study provides evidence that the DHCR24 locus may link peripheral metabolism to brain microstructure, an association with implications for cognitive impairment.
Collapse
Affiliation(s)
- Eeva Sliz
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON, Canada
- Center for Life-Course Health Research and Computational Medicine, Faculty of Medicine, University of Oulu, and Biocenter Oulu, Oulu, Finland
| | - Jean Shin
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Catriona Syme
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Yash Patel
- Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, ON, Canada
| | - Nadine Parker
- Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, ON, Canada
| | - Louis Richer
- Department of Health Sciences, Université du Québec à Chicoutimi, Chicoutimi, QC, Canada
| | - Daniel Gaudet
- Clinical Lipidology and rare lipid disorders Unit, Community Genetic Medicine Center, Department of Medicine, Université de Montréal, ECOGENE-21, Chicoutimi, QC, Canada
| | - Steffany Bennett
- Neural Regeneration Laboratory, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Tomas Paus
- Bloorview Research Institute, Holland Bloorview Kids Rehabilitation Hospital, Toronto, ON, Canada
- Departments of Psychology and Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Zdenka Pausova
- The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada.
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
30
|
Wang Y, Ji M, Zhu M, Fan J, Xie J, Huang Y, Wei X, Jiang X, Xu J, Chen L, Yin R, Wang C, Zhang R, Zhao Y, Dai J, Jin G, Hu Z, Christiani DC, Ma H, Xu L, Shen H. Genome-wide gene-smoking interaction study identified novel susceptibility loci for non-small cell lung cancer in Chinese populations. Carcinogenesis 2021; 42:1154-1161. [PMID: 34297049 DOI: 10.1093/carcin/bgab064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/27/2021] [Accepted: 07/22/2021] [Indexed: 12/24/2022] Open
Abstract
Gene-smoking interactions play important roles in the development of non-small cell lung cancer (NSCLC). To identify single nucleotide polymorphisms (SNPs) that modify the association of smoking behavior with NSCLC risk, we conducted a genome-wide gene-smoking interaction study in Chinese populations. The genome-wide interaction analysis between SNPs and smoking status (ever- versus never-smokers) was carried out using genome-wide association studies (GWAS) of NSCLC, which included 13,327 cases and 13,328 controls. Stratified analysis by histological subtypes was also conducted. We used a genome-wide significance threshold of 5×10 -8 for identifying significant gene-smoking interactions and 1×10 -6 for identifying suggestive results. Functional annotation was performed to identify potential functional SNPs and target genes. We identified three novel loci with significant or suggestive gene-smoking interaction. For NSCLC, the interaction between rs2746087 (20q11.23) and smoking status reached genome-wide significance threshold (OR = 0.63, 95%CI: 0.54-0.74, P = 3.31×10 -8), and the interaction between rs11912498 (22q12.1) and smoking status reached suggestive significance threshold (OR = 0.72, 95%CI: 0.63-0.82, P = 8.10×10 -7). Stratified analysis by histological subtypes identified suggestive interactions between rs459724 (5q11.2) and smoking status (OR = 0.61, 95%CI: 0.51-0.73, P = 7.55×10 -8) in the risk of lung squamous cell carcinoma. Functional annotation indicated that both classic and novel biological processes, including nicotine addiction and airway clearance, may modulate the susceptibility to NSCLC. These novel loci provide new insights into the biological mechanisms underlying NSCLC risk. Independent replication in large-scale studies is needed and experimental studies are warranted to functionally validate these associations.
Collapse
Affiliation(s)
- Yuzhuo Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Mengmeng Ji
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Epidemiology, School of Public Health, Southeast University, Nanjing, China
| | - Meng Zhu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Jingyi Fan
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Junxing Xie
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yanqian Huang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiaoxia Wei
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiangxiang Jiang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jing Xu
- Department of Thoracic Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liang Chen
- Department of Thoracic Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rong Yin
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Cheng Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Ruyang Zhang
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America.,China International Cooperation Center for Environment and Human Health, Nanjing Medical University, Nanjing, China
| | - Yang Zhao
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Juncheng Dai
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Guangfu Jin
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Zhibin Hu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - David C Christiani
- Department of Environmental Health, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America.,Pulmonary and Critical Care Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Hongxia Ma
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Lin Xu
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Hongbing Shen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Research Units of Cohort Study on Cardiovascular Diseases and Cancers, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
31
|
Page GP, Kanias T, Guo YJ, Lanteri MC, Zhang X, Mast AE, Cable RG, Spencer BR, Kiss JE, Fang F, Endres-Dighe SM, Brambilla D, Nouraie M, Gordeuk VR, Kleinman S, Busch MP, Gladwin MT. Multiple-ancestry genome-wide association study identifies 27 loci associated with measures of hemolysis following blood storage. J Clin Invest 2021; 131:146077. [PMID: 34014839 DOI: 10.1172/jci146077] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 05/13/2021] [Indexed: 12/17/2022] Open
Abstract
BackgroundThe evolutionary pressure of endemic malaria and other erythrocytic pathogens has shaped variation in genes encoding erythrocyte structural and functional proteins, influencing responses to hemolytic stress during transfusion and disease.MethodsWe sought to identify such genetic variants in blood donors by conducting a genome-wide association study (GWAS) of 12,353 volunteer donors, including 1,406 African Americans, 1,306 Asians, and 945 Hispanics, whose stored erythrocytes were characterized by quantitative assays of in vitro osmotic, oxidative, and cold-storage hemolysis.ResultsGWAS revealed 27 significant loci (P < 5 × 10-8), many in candidate genes known to modulate erythrocyte structure, metabolism, and ion channels, including SPTA1, ALDH2, ANK1, HK1, MAPKAPK5, AQP1, PIEZO1, and SLC4A1/band 3. GWAS of oxidative hemolysis identified variants in genes encoding antioxidant enzymes, including GLRX, GPX4, G6PD, and SEC14L4 (Golgi-transport protein). Genome-wide significant loci were also tested for association with the severity of steady-state (baseline) in vivo hemolytic anemia in patients with sickle cell disease, with confirmation of identified SNPs in HBA2, G6PD, PIEZO1, AQP1, and SEC14L4.ConclusionsMany of the identified variants, such as those in G6PD, have previously been shown to impair erythrocyte recovery after transfusion, associate with anemia, or cause rare Mendelian human hemolytic diseases. Candidate SNPs in these genes, especially in polygenic combinations, may affect RBC recovery after transfusion and modulate disease severity in hemolytic diseases, such as sickle cell disease and malaria.
Collapse
Affiliation(s)
- Grier P Page
- Division of Biostatistics and Epidemiology, RTI International, Atlanta, Georgia, USA
| | - Tamir Kanias
- Vitalant Research Institute, Denver, Colorado, USA
| | - Yuelong J Guo
- Division of Biostatistics and Epidemiology, RTI International, Durham, North Carolina, USA
| | - Marion C Lanteri
- Vitalant Research Institute and the Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Xu Zhang
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Alan E Mast
- Blood Research Institute, Blood Center of Wisconsin, and Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | | | - Joseph E Kiss
- Vitalant Northeast Division, Pittsburgh, Pennsylvania, USA
| | - Fang Fang
- Division of Biostatistics and Epidemiology, RTI International, Durham, North Carolina, USA
| | - Stacy M Endres-Dighe
- Division of Biostatistics and Epidemiology, RTI International, Rockville, Maryland, USA
| | - Donald Brambilla
- Division of Biostatistics and Epidemiology, RTI International, Rockville, Maryland, USA
| | - Mehdi Nouraie
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pennsylvania, USA
| | - Victor R Gordeuk
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Steve Kleinman
- University of British Columbia, Victoria, British Columbia, Canada
| | - Michael P Busch
- Vitalant Research Institute and the Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Mark T Gladwin
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
32
|
Westerman KE, Pham DT, Hong L, Chen Y, Sevilla-González M, Sung YJ, Sun YV, Morrison AC, Chen H, Manning AK. CLUE: Exact maximal reduction of kinetic models by constrained lumping of differential equations. Bioinformatics 2021; 37:btab223. [PMID: 34037712 PMCID: PMC8545347 DOI: 10.1093/bioinformatics/btab223] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 03/09/2021] [Accepted: 04/07/2021] [Indexed: 01/19/2023] Open
Abstract
MOTIVATION Detailed mechanistic models of biological processes can pose significant challenges for analysis and parameter estimations due to the large number of equations used to track the dynamics of all distinct configurations in which each involved biochemical species can be found. Model reduction can help tame such complexity by providing a lower-dimensional model in which each macro-variable can be directly related to the original variables. RESULTS We present CLUE, an algorithm for exact model reduction of systems of polynomial differential equations by constrained linear lumping. It computes the smallest dimensional reduction as a linear mapping of the state space such that the reduced model preserves the dynamics of user-specified linear combinations of the original variables. Even though CLUE works with nonlinear differential equations, it is based on linear algebra tools, which makes it applicable to high-dimensional models. Using case studies from the literature, we show how CLUE can substantially lower model dimensionality and help extract biologically intelligible insights from the reduction. AVAILABILITY An implementation of the algorithm and relevant resources to replicate the experiments herein reported are freely available for download at https://github.com/pogudingleb/CLUE. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Kenneth E Westerman
- Department of Medicine, Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA 02114, USA
- Metabolism Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Duy T Pham
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Liang Hong
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ye Chen
- Department of Medicine, Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Magdalena Sevilla-González
- Department of Medicine, Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA 02114, USA
- Metabolism Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Yun Ju Sung
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63130, USA
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Yan V Sun
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA 30322, USA
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Han Chen
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Center for Precision Health, School of Public Health and School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Alisa K Manning
- Department of Medicine, Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA 02114, USA
- Metabolism Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
33
|
Tchio C, Musani SK, Quarshie A, Tosini G. Association between MTNR1B polymorphisms and obesity in African American: findings from the Jackson Heart Study. BMC Med Genomics 2021; 14:136. [PMID: 34020621 PMCID: PMC8138980 DOI: 10.1186/s12920-021-00983-2] [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] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 05/13/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Melatonin is a hormone that is secreted at night by the pineal gland. It exerts its function by binding to the MT1 and MT2 receptors, which are encoded by the MTNR1A and MTNR1B genes, respectively. Previous studies reveal that MTNR1B variants are associated with insulin secretion impairments and an increased body mass index (BMI) in individuals of European and Asian ancestries. Obesity is highly prevalent in the US and disproportionately affects African Americans. Here, we hypothesized that common single nucleotide polymorphisms (SNPs) imputed in 1000 Genomes in the MTNR1B gene are associated with adiposity in African American adult men and women and that the association is modified by insomnia. METHODS We used an additive genetic model to describe the association between the adiposity traits (BMI and waist circumference) and selected MTNR1B variants in 3,029 Jackson Heart Study participants, with an average age of 55.13 ± 12.84 years, and 62% were women. We regressed the adiposity measures on the estimated allelic or genotypic dosage at every selected SNP and adjusted for age, sex, population stratification, and insomnia. Thirty common SNPs, spanning the MTNR1B gene, with a minor allele frequency ≥ 5%, a call rate ≥ 90%, a Hardy-Weinberg equilibrium p value > 10-6, were available for the analysis. RESULTS The allele T of rs76371840 was associated with adiposity (OR = 1.47 [1.13-1.82]; PFDR-adjusted = 0.0499), and the allele A of rs8192552 showed a significant association with waist circumference (β = 0.023 ± 0.007; PFDR-adjusted = 0.0077) after correcting for multiple testing. When insomnia was included in the adiposity analysis model, the following four variants became significantly associated with adiposity: rs6483208; rs4388843; rs4601728; and rs12804291. CONCLUSIONS Our data indicate that polymorphisms in the MTNR1B gene are associated with obesity traits in African Americans. To the best of our knowledge, this is the first study to explore the effect of insomnia on the association between the circadian MTNR1B genetic variants and metabolic traits in an African American sample population. We observed that insomnia affected the association between the MTNR1B variants and adiposity.
Collapse
Affiliation(s)
- Cynthia Tchio
- Circadian Rhythms and Sleep Disorders Program, Neuroscience Institute, Morehouse School of Medicine, 720 Westview Dr. SW, Atlanta, GA, 30130, USA
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Solomon K Musani
- Jackson Heart Study, University of Mississippi Medical Center, Jackson, MS, USA
| | - Alexander Quarshie
- Clinical Research Center, Morehouse School of Medicine, Atlanta, GA, USA
| | - Gianluca Tosini
- Circadian Rhythms and Sleep Disorders Program, Neuroscience Institute, Morehouse School of Medicine, 720 Westview Dr. SW, Atlanta, GA, 30130, USA.
- Department of Pharmacology and Toxicology, Morehouse School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
34
|
Xu H, Schwander K, Brown MR, Wang W, Waken RJ, Boerwinkle E, Cupples LA, de las Fuentes L, van Heemst D, Osazuwa-Peters O, de Vries PS, van Dijk KW, Sung YJ, Zhang X, Morrison AC, Rao DC, Noordam R, Liu CT. Lifestyle Risk Score: handling missingness of individual lifestyle components in meta-analysis of gene-by-lifestyle interactions. Eur J Hum Genet 2021; 29:839-850. [PMID: 33500576 PMCID: PMC8110957 DOI: 10.1038/s41431-021-00808-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 11/30/2020] [Accepted: 01/05/2021] [Indexed: 01/29/2023] Open
Abstract
Recent studies consider lifestyle risk score (LRS), an aggregation of multiple lifestyle exposures, in identifying association of gene-lifestyle interaction with disease traits. However, not all cohorts have data on all lifestyle factors, leading to increased heterogeneity in the environmental exposure in collaborative meta-analyses. We compared and evaluated four approaches (Naïve, Safe, Complete and Moderator Approaches) to handle the missingness in LRS-stratified meta-analyses under various scenarios. Compared to "benchmark" results with all lifestyle factors available for all cohorts, the Complete Approach, which included only cohorts with all lifestyle components, was underpowered due to lower sample size, and the Naïve Approach, which utilized all available data and ignored the missingness, was slightly inflated. The Safe Approach, which used all data in LRS-exposed group and only included cohorts with all lifestyle factors available in the LRS-unexposed group, and the Moderator Approach, which handled missingness via moderator meta-regression, were both slightly conservative and yielded almost identical p values. We also evaluated the performance of the Safe Approach under different scenarios. We observed that the larger the proportion of cohorts without missingness included, the more accurate the results compared to "benchmark" results. In conclusion, we generally recommend the Safe Approach, a straightforward and non-inflated approach, to handle heterogeneity among cohorts in the LRS based genome-wide interaction meta-analyses.
Collapse
Affiliation(s)
- Hanfei Xu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA.
| | - Karen Schwander
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael R Brown
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, the University of Texas School of Public health, Houston, TX, USA
| | - Wenyi Wang
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - R J Waken
- Field and Environmental Data Science, Benson Hill Inc, St. Louis, MO, USA
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, the University of Texas School of Public health, Houston, TX, USA
- The Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - L Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- NHLBI and Boston University Framingham Heart Study, Framingham, MA, USA
| | - Lisa de las Fuentes
- Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Diana van Heemst
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, the University of Texas School of Public health, Houston, TX, USA
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Leiden Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Yun Ju Sung
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaoyu Zhang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, the University of Texas School of Public health, Houston, TX, USA
| | - D C Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Raymond Noordam
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA.
| |
Collapse
|
35
|
Chang X, Dorajoo R, Sun Y, Wang L, Ong CN, Liu J, Khor CC, Yuan JM, Koh WP, Friedlander Y, Heng CK. Effect of plasma polyunsaturated fatty acid levels on leukocyte telomere lengths in the Singaporean Chinese population. Nutr J 2020; 19:119. [PMID: 33126880 PMCID: PMC7602302 DOI: 10.1186/s12937-020-00626-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 09/15/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Shorter telomere length (TL) has been associated with poor health behaviors, increased risks of chronic diseases and early mortality. Excessive shortening of telomere is a marker of accelerated aging and can be influenced by oxidative stress and nutritional deficiency. Plasma n6:n3 polyunsaturated fatty acid (PUFA) ratio may impact cell aging. Increased dietary intake of marine n-3 PUFA is associated with reduced telomere attrition. However, the effect of plasma PUFA on leukocyte telomere length (LTL) and its interaction with genetic variants are not well established. METHODS A nested coronary artery disease (CAD) case-control study comprising 711 cases and 638 controls was conducted within the Singapore Chinese Health Study (SCHS). Samples genotyped with the Illumina ZhongHua-8 array. Plasma n-3 and n-6 PUFA were quantified using mass spectrometry (MS). LTL was measured with quantitative PCR method. Linear regression was used to test the association between PUFA and LTL. The interaction between plasma PUFAs and genetic variants was assessed by introducing an additional term (PUFA×genetic variant) in the regression model. Analysis was carried out in cases and controls separately and subsequently meta-analyzed using the inverse-variance weighted method. We further assessed the association of PUFA and LTL with CAD risk by Cox Proportional-Hazards model and whether the effect of PUFA on CAD was mediated through LTL by using structural equation modeling. RESULTS Higher n6:n3 ratio was significantly associated with shorter LTL (p = 0.018) and increased CAD risk (p = 0.005). These associations were mainly driven by elevated plasma total n-3 PUFAs, especially eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) (p < 0.05). There was a statistically significant interaction for an intergenic single nucleotide polymorphism (SNP) rs529143 with plasma total n-3 PUFA and DHA on LTL beyond the genome-wide threshold (p < 5 × 10- 8). Mediation analysis showed that PUFA and LTL affected CAD risk independently. CONCLUSIONS Higher plasma n6:n3 PUFA ratio, and lower EPA and DHA n-3 PUFAs were associated with shorter LTL and increased CAD risk in this Chinese population. Furthermore, genetic variants may modify the effect of PUFAs on LTL. PUFA and LTL had independent effect on CAD risk in our study population.
Collapse
Affiliation(s)
- Xuling Chang
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block, Level 12, 1E Kent Ridge Road, Singapore, 119228, Singapore
- Khoo Teck Puat - National University Children's Medical Institute, National University Health System, Singapore, Singapore
| | - Rajkumar Dorajoo
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Ye Sun
- Nestlé Research Singapore Hub, Singapore, 21 Biopolis Drive, Nucleos, Singapore, Singapore
| | - Ling Wang
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Choon Nam Ong
- Saw Swee Hock School of Public Health, National University of Singapore, National University Health System, Singapore, Singapore
- NUS Environmental Research Institute, National University of Singapore, Singapore, Singapore
| | - 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, Singapore, Singapore
| | - Chiea Chuen Khor
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - 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
| | - Woon Puay Koh
- Saw Swee Hock School of Public Health, National University of Singapore, National University Health System, Singapore, Singapore
- Health Systems and Services Research, Duke-NUS Medical School Singapore, Singapore, Singapore
| | - Yechiel Friedlander
- School of Public Health and Community Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
- Unit of Epidemiology, Hebrew University-Hadassah Braun School of Public Health, POB 12272, 91120, Jerusalem, Israel.
| | - Chew-Kiat Heng
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block, Level 12, 1E Kent Ridge Road, Singapore, 119228, Singapore.
- Khoo Teck Puat - National University Children's Medical Institute, National University Health System, Singapore, Singapore.
| |
Collapse
|
36
|
Leal-Gutiérrez JD, Rezende FM, Reecy JM, Kramer LM, Peñagaricano F, Mateescu RG. Whole Genome Sequence Data Provides Novel Insights Into the Genetic Architecture of Meat Quality Traits in Beef. Front Genet 2020; 11:538640. [PMID: 33101375 PMCID: PMC7500205 DOI: 10.3389/fgene.2020.538640] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 08/12/2020] [Indexed: 12/12/2022] Open
Abstract
Tenderness is a major quality attribute for fresh beef steaks in the United States, and meat quality traits in general are suitable candidates for genomic research. The objectives of the present analysis were to (1) perform genome-wide association (GWA) analysis for marbling, Warner-Bratzler shear force (WBSF), tenderness, and connective tissue using whole-genome data in an Angus population, (2) identify enriched pathways in each GWA analysis; (3) construct a protein-protein interaction network using the associated genes and (4) perform a μ-calpain proteolysis assessment for associated structural proteins. An Angus-sired population of 2,285 individuals was assessed. Animals were transported to a commercial packing plant and harvested at an average age of 457 ± 46 days. After 48 h postmortem, marbling was recorded by graders' visual appraisal. Two 2.54-cm steaks were sampled from each muscle for recording of WBSF, and tenderness, and connective tissue by a sensory panel. The relevance of additive effects on marbling, WBSF, tenderness, and connective tissue was evaluated on a genome-wide scale using a two-step mixed model-based approach in single-trait analysis. A tissue-restricted gene enrichment was performed for each GWA where all polymorphisms with an association p-value lower than 1 × 10-3 were included. The genes identified as associated were included in a protein-protein interaction network and a candidate structural protein assessment of proteolysis analyses. A total of 1,867, 3,181, 3,926, and 3,678 polymorphisms were significantly associated with marbling, WBSF, tenderness, and connective tissue, respectively. The associate region on BTA29 (36,432,655-44,313,046 bp) harbors 13 highly significant markers for meat quality traits. Enrichment for the GO term GO:0005634 (Nucleus), which includes transcription factors, was evident. The final protein-protein network included 431 interations between 349 genes. The 42 most important genes based on significance that encode structural proteins were included in a proteolysis analysis, and 81% of these proteins were potential μ-Calpain substrates. Overall, this comprehensive study unraveled genetic variants, genes and mechanisms of action responsible for the variation in meat quality traits. Our findings can provide opportunities for improving meat quality in beef cattle via marker-assisted selection.
Collapse
Affiliation(s)
| | - Fernanda M. Rezende
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
- Faculdade de Medicina Veterinária, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - James M. Reecy
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Luke M. Kramer
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Francisco Peñagaricano
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
- University of Florida Genetics Institute, University of Florida, Gainesville, FL, United States
| | - Raluca G. Mateescu
- Department of Animal Sciences, University of Florida, Gainesville, FL, United States
| |
Collapse
|
37
|
Osazuwa-Peters OL, Waken RJ, Schwander KL, Sung YJ, de Vries PS, Hartz SM, Chasman DI, Morrison AC, Bierut LJ, Xiong C, de las Fuentes L, Rao DC. Identifying blood pressure loci whose effects are modulated by multiple lifestyle exposures. Genet Epidemiol 2020; 44:629-641. [PMID: 32227373 PMCID: PMC7717887 DOI: 10.1002/gepi.22292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/30/2019] [Accepted: 03/06/2020] [Indexed: 12/27/2022]
Abstract
Although multiple lifestyle exposures simultaneously impact blood pressure (BP) and cardiovascular health, most analysis so far has considered each single lifestyle exposure (e.g., smoking) at a time. Here, we exploit gene-multiple lifestyle exposure interactions to find novel BP loci. For each of 6,254 Framingham Heart Study participants, we computed lifestyle risk score (LRS) value by aggregating the risk of four lifestyle exposures (smoking, alcohol, education, and physical activity) on BP. Using the LRS, we performed genome-wide gene-environment interaction analysis in systolic and diastolic BP using the joint 2 degree of freedom (DF) and 1 DF interaction tests. We identified one genome-wide significant (p < 5 × 10-8 ) and 11 suggestive (p < 1 × 10-6 ) loci. Gene-environment analysis using single lifestyle exposures identified only one of the 12 loci. Nine of the 12 BP loci detected were novel. Loci detected by the LRS were located within or nearby genes with biologically plausible roles in the pathophysiology of hypertension, including KALRN, VIPR2, SNX1, and DAPK2. Our results suggest that simultaneous consideration of multiple lifestyle exposures in gene-environment interaction analysis can identify additional loci missed by single lifestyle approaches.
Collapse
Affiliation(s)
| | - R J Waken
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Karen L Schwander
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Yun Ju Sung
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Sarah M Hartz
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - Daniel I Chasman
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Laura J Bierut
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Lisa de las Fuentes
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
- Cardiovascular Division, Department of Medicine, Washington University, St. Louis, Missouri
| | - D C Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
38
|
Xu H, Li X, Yang Y, Li Y, Pinheiro J, Sasser K, Hamadeh H, Steven X, Yuan M. High-throughput and efficient multilocus genome-wide association study on longitudinal outcomes. Bioinformatics 2020; 36:3004-3010. [PMID: 32096821 DOI: 10.1093/bioinformatics/btaa120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/16/2020] [Accepted: 02/18/2020] [Indexed: 11/12/2022] Open
Abstract
MOTIVATION With the emerging of high-dimensional genomic data, genetic analysis such as genome-wide association studies (GWAS) have played an important role in identifying disease-related genetic variants and novel treatments. Complex longitudinal phenotypes are commonly collected in medical studies. However, since limited analytical approaches are available for longitudinal traits, these data are often underutilized. In this article, we develop a high-throughput machine learning approach for multilocus GWAS using longitudinal traits by coupling Empirical Bayesian Estimates from mixed-effects modeling with a novel ℓ0-norm algorithm. RESULTS Extensive simulations demonstrated that the proposed approach not only provided accurate selection of single nucleotide polymorphisms (SNPs) with comparable or higher power but also robust control of false positives. More importantly, this novel approach is highly scalable and could be approximately >1000 times faster than recently published approaches, making genome-wide multilocus analysis of longitudinal traits possible. In addition, our proposed approach can simultaneously analyze millions of SNPs if the computer memory allows, thereby potentially allowing a true multilocus analysis for high-dimensional genomic data. With application to the data from Alzheimer's Disease Neuroimaging Initiative, we confirmed that our approach can identify well-known SNPs associated with AD and were much faster than recently published approaches (≥6000 times). AVAILABILITY AND IMPLEMENTATION The source code and the testing datasets are available at https://github.com/Myuan2019/EBE_APML0. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Huang Xu
- Department of Statistics and Finance, University of Science and Technology of China, Hefei 230026, China
| | - Xiang Li
- Janssen Research and Development, Raritan, NJ 08869, USA
| | - Yaning Yang
- Department of Statistics and Finance, University of Science and Technology of China, Hefei 230026, China
| | - Yi Li
- Department of Statistics and Finance, University of Science and Technology of China, Hefei 230026, China
| | - Jose Pinheiro
- Janssen Research and Development, Raritan, NJ 08869, USA
| | | | | | - Xu Steven
- Genmab US, Inc., Princeton, NJ 08540, USA
| | - Min Yuan
- School of Public Health Administration, Anhui Medical University, Hefei 230032, China
| | | |
Collapse
|
39
|
Manry J, Vincent QB, Johnson C, Chrabieh M, Lorenzo L, Theodorou I, Ardant MF, Marion E, Chauty A, Marsollier L, Abel L, Alcaïs A. Genome-wide association study of Buruli ulcer in rural Benin highlights role of two LncRNAs and the autophagy pathway. Commun Biol 2020; 3:177. [PMID: 32313116 PMCID: PMC7171125 DOI: 10.1038/s42003-020-0920-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 03/24/2020] [Indexed: 02/07/2023] Open
Abstract
Buruli ulcer, caused by Mycobacterium ulcerans and characterized by devastating necrotizing skin lesions, is the third mycobacterial disease worldwide. The role of host genetics in susceptibility to Buruli ulcer has long been suggested. We conduct the first genome-wide association study of Buruli ulcer on a sample of 1524 well characterized patients and controls from rural Benin. Two-stage analyses identify two variants located within LncRNA genes: rs9814705 in ENSG00000240095.1 (P = 2.85 × 10−7; odds ratio = 1.80 [1.43–2.27]), and rs76647377 in LINC01622 (P = 9.85 × 10−8; hazard ratio = 0.41 [0.28–0.60]). Furthermore, we replicate the protective effect of allele G of a missense variant located in ATG16L1, previously shown to decrease bacterial autophagy (rs2241880, P = 0.003; odds ratio = 0.31 [0.14–0.68]). Our results suggest LncRNAs and the autophagy pathway as critical factors in the development of Buruli ulcer. Jeremy Manry, Quentin Vincent et al. report a genome-wide association study for susceptibility to Buruli ulcer in a rural population from the West African country of Benin. They identify two independently associated variants within LncRNA genes and confirm the protective effect of a missense variant in the bacterial autophagy gene ATG16L1.
Collapse
Affiliation(s)
- Jeremy Manry
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1163, Paris, France. .,Université de Paris, Imagine Institute, Paris, France.
| | - Quentin B Vincent
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1163, Paris, France.,Université de Paris, Imagine Institute, Paris, France
| | - Christian Johnson
- Fondation Raoul Follereau, Paris, France.,Centre Interfacultaire de Formation et de Recherche en Environnement pour le Développement Durable. Université d'Abomey, Calavi, Benin
| | - Maya Chrabieh
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1163, Paris, France.,Université de Paris, Imagine Institute, Paris, France
| | - Lazaro Lorenzo
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1163, Paris, France.,Université de Paris, Imagine Institute, Paris, France
| | - Ioannis Theodorou
- Center for Immunology and Infectious Diseases, INSERM UMR S 1135, Pierre and Marie Curie University, and AP-HP Laboratoire d'Immunologie et Histocompatibilité Hôpital Saint-Louis, Paris, France
| | - Marie-Françoise Ardant
- Fondation Raoul Follereau, Paris, France.,Centre de Dépistage et de Traitement de la Lèpre et de l'Ulcère de Buruli (CDTLUB), Pobè, Benin
| | - Estelle Marion
- INSERM UMR-U892 and CNRS U6299, team 7, Angers University, Angers University Hospital, Angers, France
| | - Annick Chauty
- Fondation Raoul Follereau, Paris, France.,Centre de Dépistage et de Traitement de la Lèpre et de l'Ulcère de Buruli (CDTLUB), Pobè, Benin
| | - Laurent Marsollier
- INSERM UMR-U892 and CNRS U6299, team 7, Angers University, Angers University Hospital, Angers, France
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1163, Paris, France.,Université de Paris, Imagine Institute, Paris, France.,St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Alexandre Alcaïs
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1163, Paris, France. .,Université de Paris, Imagine Institute, Paris, France.
| |
Collapse
|
40
|
Magnus MC, Guyatt AL, Lawn RB, Wyss AB, Trajanoska K, Küpers LK, Rivadeneira F, Tobin MD, London SJ, Lawlor DA, Millard LAC, Fraser A. Identifying potential causal effects of age at menarche: a Mendelian randomization phenome-wide association study. BMC Med 2020; 18:71. [PMID: 32200763 PMCID: PMC7087394 DOI: 10.1186/s12916-020-01515-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 02/10/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Age at menarche has been associated with various health outcomes. We aimed to identify potential causal effects of age at menarche on health-related traits in a hypothesis-free manner. METHODS We conducted a Mendelian randomization phenome-wide association study (MR-pheWAS) of age at menarche with 17,893 health-related traits in UK Biobank (n = 181,318) using PHESANT. The exposure of interest was the genetic risk score for age at menarche. We conducted a second MR-pheWAS after excluding SNPs associated with BMI from the genetic risk score, to examine whether results might be due to the genetic overlap between age at menarche and BMI. We followed up a subset of health-related traits to investigate MR assumptions and seek replication in independent study populations. RESULTS Of the 17,893 tests performed in our MR-pheWAS, we identified 619 associations with the genetic risk score for age at menarche at a 5% false discovery rate threshold, of which 295 were below a Bonferroni-corrected P value threshold. These included potential effects of younger age at menarche on lower lung function, higher heel bone-mineral density, greater burden of psychosocial/mental health problems, younger age at first birth, higher risk of childhood sexual abuse, poorer cardiometabolic health, and lower physical activity. After exclusion of variants associated with BMI, the genetic risk score for age at menarche was related to 37 traits at a 5% false discovery rate, of which 29 were below a Bonferroni-corrected P value threshold. We attempted to replicate findings for bone-mineral density, lung function, neuroticism, and childhood sexual abuse using 5 independent cohorts/consortia. While estimates for lung function, higher bone-mineral density, neuroticism, and childhood sexual abuse in replication cohorts were consistent with UK Biobank estimates, confidence intervals were wide and often included the null. CONCLUSIONS The genetic risk score for age at menarche was related to a broad range of health-related traits. Follow-up analyses indicated imprecise evidence of an effect of younger age at menarche on greater bone-mineral density, lower lung function, higher neuroticism score, and greater risk of childhood sexual abuse in the smaller replication samples available; hence, these findings need further exploration when larger independent samples become available.
Collapse
Affiliation(s)
- Maria C Magnus
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK.
- Population Health Sciences, Bristol Medical School, Bristol, UK.
- Centre for Fertility and Health, Norwegian Institute of Public Health, P.O. Box 222 Skøyen, 0213, Oslo, Norway.
| | - Anna L Guyatt
- Department of Health Sciences, University of Leicester, Leicester, UK
| | - Rebecca B Lawn
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, Bristol, UK
- School of Experimental Psychology, University of Bristol, Bristol, UK
| | - Annah B Wyss
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Katerina Trajanoska
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Leanne K Küpers
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, Bristol, UK
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Martin D Tobin
- Department of Health Sciences, University of Leicester, Leicester, UK
- National Institute for Health Research, Leicester Respiratory Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Stephanie J London
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Debbie A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, Bristol, UK
- NIHR Bristol Biomedical Research Centre at the University Hospitals Bristol NHS Foundation Trust and the University of Bristol, Bristol, UK
| | - Louise A C Millard
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, Bristol, UK
- Intelligent Systems Laboratory, Department of Computer Science, University of Bristol, Bristol, UK
| | - Abigail Fraser
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, Bristol, UK
- NIHR Bristol Biomedical Research Centre at the University Hospitals Bristol NHS Foundation Trust and the University of Bristol, Bristol, UK
| |
Collapse
|
41
|
Pacheco HA, Rezende FM, Peñagaricano F. Gene mapping and genomic prediction of bull fertility using sex chromosome markers. J Dairy Sci 2020; 103:3304-3311. [PMID: 32063375 DOI: 10.3168/jds.2019-17767] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 12/09/2019] [Indexed: 12/29/2022]
Abstract
Service sire has been recognized as an important factor affecting dairy herd fertility. Our group has reported promising results on gene mapping and genomic prediction of dairy bull fertility using autosomal SNP markers. Little is known, however, about the genetic contribution of sex chromosomes, which are enriched in genes related to sexual development and reproduction. As such, the main goal of this study was to investigate the effect of SNP markers on X and Y chromosomes (BTAX and BTAY, respectively) on sire conception rate (SCR) in US Holstein bulls. The analysis included a total of 5,014 bulls with SCR records and genotypes for roughly 291k SNP located on the autosomes, 1.5k SNP located on the pseudoautosomal region (PAR), 13.7k BTAX-specific SNP, and 24 BTAY-specific SNP. We first performed genomic scans of the sex chromosomes, and then we evaluated the genomic prediction of SCR including BTAX SNP markers in the predictive models. Two markers located on PAR and 3 markers located on the X-specific region showed significant associations with sire fertility. Interestingly, these regions harbor genes, such as FAM9B, TBL1X, and PIH1D3, that are directly implicated in testosterone concentration, spermatogenesis, and sperm motility. On the other hand, BTAY showed very low genetic variability, and none of the segregating markers were associated with SCR. Notably, model predictive ability was largely improved by including BTAX markers. Indeed, the combination of autosomal with BTAX SNP delivered predictive correlations around 0.343, representing an increase in accuracy of about 7.5% compared with the standard whole autosomal genome approach. Overall, this study provides evidence of the importance of both PAR and X-specific regions in male fertility in dairy cattle. These findings may help to improve conception rates in dairy herds through accurate genome-guided decisions on bull fertility.
Collapse
Affiliation(s)
- Hendyel A Pacheco
- Department of Animal Sciences, University of Florida, Gainesville 32611
| | - Fernanda M Rezende
- Department of Animal Sciences, University of Florida, Gainesville 32611; Faculdade de Medicina Veterinária, Universidade Federal de Uberlândia, Uberlândia MG 38400-902, Brazil
| | - Francisco Peñagaricano
- Department of Animal Sciences, University of Florida, Gainesville 32611; University of Florida Genetics Institute, University of Florida, Gainesville 32610.
| |
Collapse
|
42
|
Kerns SL, Fachal L, Dorling L, Barnett GC, Baran A, Peterson DR, Hollenberg M, Hao K, Narzo AD, Ahsen ME, Pandey G, Bentzen SM, Janelsins M, Elliott RM, Pharoah PDP, Burnet NG, Dearnaley DP, Gulliford SL, Hall E, Sydes MR, Aguado-Barrera ME, Gómez-Caamaño A, Carballo AM, Peleteiro P, Lobato-Busto R, Stock R, Stone NN, Ostrer H, Usmani N, Singhal S, Tsuji H, Imai T, Saito S, Eeles R, DeRuyck K, Parliament M, Dunning AM, Vega A, Rosenstein BS, West CML. Radiogenomics Consortium Genome-Wide Association Study Meta-Analysis of Late Toxicity After Prostate Cancer Radiotherapy. J Natl Cancer Inst 2020; 112:179-190. [PMID: 31095341 PMCID: PMC7019089 DOI: 10.1093/jnci/djz075] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/20/2019] [Accepted: 04/29/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND A total of 10%-20% of patients develop long-term toxicity following radiotherapy for prostate cancer. Identification of common genetic variants associated with susceptibility to radiotoxicity might improve risk prediction and inform functional mechanistic studies. METHODS We conducted an individual patient data meta-analysis of six genome-wide association studies (n = 3871) in men of European ancestry who underwent radiotherapy for prostate cancer. Radiotoxicities (increased urinary frequency, decreased urinary stream, hematuria, rectal bleeding) were graded prospectively. We used grouped relative risk models to test associations with approximately 6 million genotyped or imputed variants (time to first grade 2 or higher toxicity event). Variants with two-sided Pmeta less than 5 × 10-8 were considered statistically significant. Bayesian false discovery probability provided an additional measure of confidence. Statistically significant variants were evaluated in three Japanese cohorts (n = 962). All statistical tests were two-sided. RESULTS Meta-analysis of the European ancestry cohorts identified three genomic signals: single nucleotide polymorphism rs17055178 with rectal bleeding (Pmeta = 6.2 × 10-10), rs10969913 with decreased urinary stream (Pmeta = 2.9 × 10-10), and rs11122573 with hematuria (Pmeta = 1.8 × 10-8). Fine-scale mapping of these three regions was used to identify another independent signal (rs147121532) associated with hematuria (Pconditional = 4.7 × 10-6). Credible causal variants at these four signals lie in gene-regulatory regions, some modulating expression of nearby genes. Previously identified variants showed consistent associations (rs17599026 with increased urinary frequency, rs7720298 with decreased urinary stream, rs1801516 with overall toxicity) in new cohorts. rs10969913 and rs17599026 had similar effects in the photon-treated Japanese cohorts. CONCLUSIONS This study increases the understanding of the architecture of common genetic variants affecting radiotoxicity, points to novel radio-pathogenic mechanisms, and develops risk models for testing in clinical studies. Further multinational radiogenomics studies in larger cohorts are worthwhile.
Collapse
Affiliation(s)
- Sarah L Kerns
- Departments of Radiation Oncology and Surgery, University of Rochester Medical Center, Rochester, NY
| | | | | | - Gillian C Barnett
- Department of Public Health and Primary Care
- Centre for Cancer Genetic Epidemiology, Strangeways Research Laboratory, University of Cambridge, Cambridge, UK; Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Andrea Baran
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY
| | - Derick R Peterson
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY
| | | | - Ke Hao
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Antonio Di Narzo
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Mehmet Eren Ahsen
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Gaurav Pandey
- Department of Genetics and Genomic Sciences and Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Søren M Bentzen
- Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, University of Maryland Greenebaum Cancer Center, School of Medicine, University of Maryland, Baltimore
| | - Michelle Janelsins
- Departments of Radiation Oncology and Surgery, University of Rochester Medical Center, Rochester, NY
| | - Rebecca M Elliott
- Division of Cancer Sciences, the University of Manchester, Manchester Academic Health Science Centre, Christie Hospital, Manchester, UK
| | - Paul D P Pharoah
- Centre for Cancer Genetic Epidemiology, Strangeways Research Laboratory, University of Cambridge, Cambridge, UK; Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Neil G Burnet
- Division of Cancer Sciences, the University of Manchester, Manchester Academic Health Science Centre, Christie Hospital, Manchester, UK
| | - David P Dearnaley
- Academic Urooncology Unit, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, UK
| | - Sarah L Gulliford
- Academic Urooncology Unit, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, UK
| | - Emma Hall
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - Matthew R Sydes
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Miguel E Aguado-Barrera
- Fundación Pública Galega de Medicina Xenómica-Servizo Galego de Saude (SERGAS & Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | | | | | | | | | - Richard Stock
- Complexo Hospitalario Universitario de Santiago, SERGAS, Santiago de Compostela, Spain; Department of Radiation Oncology
| | | | - Harry Ostrer
- Icahn School of Medicine at Mount Sinai, New York, NY; Departments of Pathology and Genetics, Albert Einstein College of Medicine, Bronx, NY
| | - Nawaid Usmani
- Division of Radiation Oncology, Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Canada
| | - Sandeep Singhal
- Department of Pathology and Cell Biology, Columbia University, New York, NY
| | - Hiroshi Tsuji
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Takashi Imai
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Shiro Saito
- Department of Urology, National Tokyo Medical Center, Tokyo, Japan
| | - Rosalind Eeles
- Division of Genetics and Epidemiology, Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, UK
| | - Kim DeRuyck
- Departments of Basic Medical Sciences and Radiotherapy, Ghent University Hospital, Ghent, Belgium
| | - Matthew Parliament
- Division of Radiation Oncology, Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Canada
| | | | - Ana Vega
- Fundación Pública Galega de Medicina Xenómica-Servizo Galego de Saude (SERGAS & Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Grupo de Medicina Xenómica, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Barry S Rosenstein
- Departments of Radiation Oncology & Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Catharine M L West
- Division of Cancer Sciences, the University of Manchester, Manchester Academic Health Science Centre, Christie Hospital, Manchester, UK
| |
Collapse
|
43
|
Shah S, Henry A, Roselli C, Lin H, Sveinbjörnsson G, Fatemifar G, Hedman ÅK, Wilk JB, Morley MP, Chaffin MD, Helgadottir A, Verweij N, Dehghan A, Almgren P, Andersson C, Aragam KG, Ärnlöv J, Backman JD, Biggs ML, Bloom HL, Brandimarto J, Brown MR, Buckbinder L, Carey DJ, Chasman DI, Chen X, Chen X, Chung J, Chutkow W, Cook JP, Delgado GE, Denaxas S, Doney AS, Dörr M, Dudley SC, Dunn ME, Engström G, Esko T, Felix SB, Finan C, Ford I, Ghanbari M, Ghasemi S, Giedraitis V, Giulianini F, Gottdiener JS, Gross S, Guðbjartsson DF, Gutmann R, Haggerty CM, van der Harst P, Hyde CL, Ingelsson E, Jukema JW, Kavousi M, Khaw KT, Kleber ME, Køber L, Koekemoer A, Langenberg C, Lind L, Lindgren CM, London B, Lotta LA, Lovering RC, Luan J, Magnusson P, Mahajan A, Margulies KB, März W, Melander O, Mordi IR, Morgan T, Morris AD, Morris AP, Morrison AC, Nagle MW, Nelson CP, Niessner A, Niiranen T, O'Donoghue ML, Owens AT, Palmer CNA, Parry HM, Perola M, Portilla-Fernandez E, Psaty BM, Rice KM, Ridker PM, Romaine SPR, Rotter JI, Salo P, Salomaa V, van Setten J, Shalaby AA, Smelser DT, Smith NL, Stender S, Stott DJ, Svensson P, et alShah S, Henry A, Roselli C, Lin H, Sveinbjörnsson G, Fatemifar G, Hedman ÅK, Wilk JB, Morley MP, Chaffin MD, Helgadottir A, Verweij N, Dehghan A, Almgren P, Andersson C, Aragam KG, Ärnlöv J, Backman JD, Biggs ML, Bloom HL, Brandimarto J, Brown MR, Buckbinder L, Carey DJ, Chasman DI, Chen X, Chen X, Chung J, Chutkow W, Cook JP, Delgado GE, Denaxas S, Doney AS, Dörr M, Dudley SC, Dunn ME, Engström G, Esko T, Felix SB, Finan C, Ford I, Ghanbari M, Ghasemi S, Giedraitis V, Giulianini F, Gottdiener JS, Gross S, Guðbjartsson DF, Gutmann R, Haggerty CM, van der Harst P, Hyde CL, Ingelsson E, Jukema JW, Kavousi M, Khaw KT, Kleber ME, Køber L, Koekemoer A, Langenberg C, Lind L, Lindgren CM, London B, Lotta LA, Lovering RC, Luan J, Magnusson P, Mahajan A, Margulies KB, März W, Melander O, Mordi IR, Morgan T, Morris AD, Morris AP, Morrison AC, Nagle MW, Nelson CP, Niessner A, Niiranen T, O'Donoghue ML, Owens AT, Palmer CNA, Parry HM, Perola M, Portilla-Fernandez E, Psaty BM, Rice KM, Ridker PM, Romaine SPR, Rotter JI, Salo P, Salomaa V, van Setten J, Shalaby AA, Smelser DT, Smith NL, Stender S, Stott DJ, Svensson P, Tammesoo ML, Taylor KD, Teder-Laving M, Teumer A, Thorgeirsson G, Thorsteinsdottir U, Torp-Pedersen C, Trompet S, Tyl B, Uitterlinden AG, Veluchamy A, Völker U, Voors AA, Wang X, Wareham NJ, Waterworth D, Weeke PE, Weiss R, Wiggins KL, Xing H, Yerges-Armstrong LM, Yu B, Zannad F, Zhao JH, Hemingway H, Samani NJ, McMurray JJV, Yang J, Visscher PM, Newton-Cheh C, Malarstig A, Holm H, Lubitz SA, Sattar N, Holmes MV, Cappola TP, Asselbergs FW, Hingorani AD, Kuchenbaecker K, Ellinor PT, Lang CC, Stefansson K, Smith JG, Vasan RS, Swerdlow DI, Lumbers RT. Genome-wide association and Mendelian randomisation analysis provide insights into the pathogenesis of heart failure. Nat Commun 2020; 11:163. [PMID: 31919418 PMCID: PMC6952380 DOI: 10.1038/s41467-019-13690-5] [Show More Authors] [Citation(s) in RCA: 516] [Impact Index Per Article: 103.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 11/18/2019] [Indexed: 12/20/2022] Open
Abstract
Heart failure (HF) is a leading cause of morbidity and mortality worldwide. A small proportion of HF cases are attributable to monogenic cardiomyopathies and existing genome-wide association studies (GWAS) have yielded only limited insights, leaving the observed heritability of HF largely unexplained. We report results from a GWAS meta-analysis of HF comprising 47,309 cases and 930,014 controls. Twelve independent variants at 11 genomic loci are associated with HF, all of which demonstrate one or more associations with coronary artery disease (CAD), atrial fibrillation, or reduced left ventricular function, suggesting shared genetic aetiology. Functional analysis of non-CAD-associated loci implicate genes involved in cardiac development (MYOZ1, SYNPO2L), protein homoeostasis (BAG3), and cellular senescence (CDKN1A). Mendelian randomisation analysis supports causal roles for several HF risk factors, and demonstrates CAD-independent effects for atrial fibrillation, body mass index, and hypertension. These findings extend our knowledge of the pathways underlying HF and may inform new therapeutic strategies.
Collapse
Affiliation(s)
- Sonia Shah
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia
- Institute of Cardiovascular Science, University College London, London, UK
- British Heart Foundation Research Accelerator, University College London, London, UK
| | - Albert Henry
- Institute of Cardiovascular Science, University College London, London, UK
- British Heart Foundation Research Accelerator, University College London, London, UK
- Institute of Health Informatics, University College London, London, UK
| | - Carolina Roselli
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Honghuang Lin
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA
| | | | - Ghazaleh Fatemifar
- British Heart Foundation Research Accelerator, University College London, London, UK
- Institute of Health Informatics, University College London, London, UK
- Health Data Research UK London, University College London, London, UK
| | - Åsa K Hedman
- Cardiovascular Medicine unit, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
| | - Jemma B Wilk
- Pfizer Worldwide Research & Development, 1 Portland St, Cambridge, MA, USA
| | - Michael P Morley
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark D Chaffin
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Anna Helgadottir
- deCODE genetics/Amgen Inc., Sturlugata 8, 101, Reykjavik, Iceland
| | - Niek Verweij
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, Imperial College London, St Mary's Campus, London, W2 1PG, UK
- MRC-PHE Centre for Environment and Health, Department of Epidemiology and Biostatistics, Imperial College London, St Mary's Campus, London, W2 1PG, UK
| | - Peter Almgren
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Charlotte Andersson
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA
- Department of Cardiology, Herlev Gentofte Hospital, Herlev Ringvej 57, 2650, Herlev, Denmark
| | - Krishna G Aragam
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Johan Ärnlöv
- Department of Neurobiology, Care Sciences and Society/ Section of Family Medicine and Primary Care, Karolinska Institutet, Stockholm, Sweden
- School of Health and Social Sciences, Dalarna University, Falun, Sweden
| | - Joshua D Backman
- Regeneron Genetics Center, 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Mary L Biggs
- Department of Biostatistics, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Heather L Bloom
- Division of Cardiology, Department of Medicine, Emory University Medical Center, Atlanta, GA, USA
| | - Jeffrey Brandimarto
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael R Brown
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas School of Public Health, Houston, Texas, USA
| | - Leonard Buckbinder
- Pfizer Worldwide Research & Development, 1 Portland St, Cambridge, MA, USA
| | - David J Carey
- Department of Molecular and Functional Genomics, Geisinger, Danville, PA, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Xing Chen
- Pfizer Worldwide Research & Development, 1 Portland St, Cambridge, MA, USA
| | - Xu Chen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jonathan Chung
- Regeneron Genetics Center, 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - William Chutkow
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - James P Cook
- Department of Biostatistics, University of Liverpool, Liverpool, UK
| | - Graciela E Delgado
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty of Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Spiros Denaxas
- British Heart Foundation Research Accelerator, University College London, London, UK
- Institute of Health Informatics, University College London, London, UK
- Health Data Research UK London, University College London, London, UK
- The National Institute for Health Research University College London Hospitals Biomedical Research Centre, University College London, London, UK
- The Alan Turing Institute, London, United Kingdom
| | - Alexander S Doney
- Division of Molecular & Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Marcus Dörr
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Samuel C Dudley
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Michael E Dunn
- Regeneron Pharmaceuticals, Cardiovascular Research, 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Gunnar Engström
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Tõnu Esko
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, 51010, Estonia
| | - Stephan B Felix
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Chris Finan
- Institute of Cardiovascular Science, University College London, London, UK
- British Heart Foundation Research Accelerator, University College London, London, UK
| | - Ian Ford
- Robertson Center for Biostatistics, University of Glasgow, Glasgow, UK
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sahar Ghasemi
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Vilmantas Giedraitis
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, 75185, Sweden
| | - Franco Giulianini
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, 02215, USA
| | - John S Gottdiener
- Department of Medicine, Division of Cardiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Stefan Gross
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Daníel F Guðbjartsson
- deCODE genetics/Amgen Inc., Sturlugata 8, 101, Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, 101, Reykjavik, Iceland
| | - Rebecca Gutmann
- Division of Cardiovascular Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | | | - Pim van der Harst
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Durrer Center for Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, The Netherlands
| | - Craig L Hyde
- Pfizer Worldwide Research & Development, 1 Portland St, Cambridge, MA, USA
| | - Erik Ingelsson
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, 94305, USA
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, LUMC, Leiden, The Netherlands
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Kay-Tee Khaw
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Marcus E Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty of Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Lars Køber
- Department of Cardiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Andrea Koekemoer
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, CB2 0QQ, UK
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Cecilia M Lindgren
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Barry London
- Division of Cardiovascular Medicine and Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA, USA
| | - Luca A Lotta
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, CB2 0QQ, UK
| | - Ruth C Lovering
- Institute of Cardiovascular Science, University College London, London, UK
- British Heart Foundation Research Accelerator, University College London, London, UK
| | - Jian'an Luan
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, CB2 0QQ, UK
| | - Patrik Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Kenneth B Margulies
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Winfried März
- Department of Biostatistics, University of Liverpool, Liverpool, UK
- Synlab Academy, Synlab Holding Deutschland GmbH, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Olle Melander
- Department of Internal Medicine, Clinical Sciences, Lund University and Skåne University Hospital, Malmö, Sweden
| | - Ify R Mordi
- Division of Molecular & Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Thomas Morgan
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Andrew D Morris
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew P Morris
- Department of Biostatistics, University of Liverpool, Liverpool, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Alanna C Morrison
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas School of Public Health, Houston, Texas, USA
| | - Michael W Nagle
- Pfizer Worldwide Research & Development, 1 Portland St, Cambridge, MA, USA
| | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Alexander Niessner
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Teemu Niiranen
- National Institute for Health and Welfare, Helsinki, Finland
- Department of Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Michelle L O'Donoghue
- TIMI Study Group, Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA
| | - Anjali T Owens
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Colin N A Palmer
- Division of Molecular & Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Helen M Parry
- Division of Molecular & Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Markus Perola
- National Institute for Health and Welfare, Helsinki, Finland
| | - Eliana Portilla-Fernandez
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Bruce M Psaty
- Department of Medicine, Epidemiology, and Health Services, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA, USA
| | - Kenneth M Rice
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Paul M Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Simon P R Romaine
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Departments of Pediatrics and Medicine, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Perttu Salo
- National Institute for Health and Welfare, Helsinki, Finland
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki, Finland
| | - Jessica van Setten
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Alaa A Shalaby
- Division of Cardiology, Department of Medicine, University of Pittsburgh Medical Center and VA Pittsburgh HCS, Pittsburgh, PA, USA
| | - Diane T Smelser
- Department of Molecular and Functional Genomics, Geisinger, Danville, PA, USA
| | - Nicholas L Smith
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Seattle Epidemiologic Research and Information Center, Department of Veterans Affairs Office of Research & Development, Seattle, WA, USA
| | - Steen Stender
- Department of Clinical Biochemistry, Copenhagen University Hospital, Herlev and Gentofte, København, Denmark
| | - David J Stott
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Per Svensson
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
- Department of Cardiology, Södersjukhuset, Stockholm, Sweden
| | - Mari-Liis Tammesoo
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, 51010, Estonia
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences, LABiomed and Departments of Pediatrics at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Maris Teder-Laving
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, 51010, Estonia
| | - Alexander Teumer
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Guðmundur Thorgeirsson
- deCODE genetics/Amgen Inc., Sturlugata 8, 101, Reykjavik, Iceland
- Division of Cardiology, Department of Internal Medicine, Landspitali, National University Hospital of Iceland, Hringbraut, 101, Reykjavik, Iceland
| | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen Inc., Sturlugata 8, 101, Reykjavik, Iceland
- Faculty of Medicine, Department of Medicine, University of Iceland, Saemundargata 2, 101, Reykjavik, Iceland
| | - Christian Torp-Pedersen
- Department of Epidemiology and Biostatistics, Aalborg University Hospital, Aalborg, Denmark
- Department of Health, Science and Technology, Aalborg University Hospital, Aalborg, Denmark
- Departments of Cardiology, Aalborg University Hospital, Aalborg, Denmark
| | - 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
| | - Benoit Tyl
- Translational and Clinical Research, Servier Cardiovascular Center for Therapeutic Innovation, 50 rue Carnot, 92284, Suresnes, France
| | - Andre G Uitterlinden
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Abirami Veluchamy
- Division of Molecular & Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Uwe Völker
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Adriaan A Voors
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Xiaosong Wang
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, CB2 0QQ, UK
| | | | - Peter E Weeke
- Department of Cardiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Raul Weiss
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH, USA
| | - Kerri L Wiggins
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Heming Xing
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | | | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, The University of Texas School of Public Health, Houston, Texas, USA
| | - Faiez Zannad
- Université de Lorraine, CHU de Nancy, Inserm and INI-CRCT (F-CRIN), Institut Lorrain du Coeur et des Vaisseaux, 54500, Vandoeuvre Lès, Nancy, France
| | - Jing Hua Zhao
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, CB2 0QQ, UK
| | - Harry Hemingway
- British Heart Foundation Research Accelerator, University College London, London, UK
- Institute of Health Informatics, University College London, London, UK
- Health Data Research UK London, University College London, London, UK
- BHF Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - John J V McMurray
- BHF Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Jian Yang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Peter M Visscher
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Christopher Newton-Cheh
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA, USA
| | - Anders Malarstig
- Cardiovascular Medicine unit, Department of Medicine Solna, Karolinska Institute, Stockholm, Sweden
- Pfizer Worldwide Research & Development, 1 Portland St, Cambridge, MA, USA
| | - Hilma Holm
- deCODE genetics/Amgen Inc., Sturlugata 8, 101, Reykjavik, Iceland
| | - Steven A Lubitz
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiac Arrhythmia Service and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Naveed Sattar
- BHF Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Michael V Holmes
- Medical Research Council Population Health Research Unit at the University of Oxford, Oxford, UK
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, Big Data Institute, University of Oxford, Oxford, UK
- National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospital, Oxford, UK
| | - Thomas P Cappola
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Folkert W Asselbergs
- Institute of Cardiovascular Science, University College London, London, UK
- British Heart Foundation Research Accelerator, University College London, London, UK
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Aroon D Hingorani
- Institute of Cardiovascular Science, University College London, London, UK
- British Heart Foundation Research Accelerator, University College London, London, UK
| | - Karoline Kuchenbaecker
- Division of Psychiatry, University College of London, London, W1T 7NF, UK
- UCL Genetics Institute, University College London, London, WC1E 6BT, UK
| | - Patrick T Ellinor
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiac Arrhythmia Service and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Chim C Lang
- Division of Molecular & Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - Kari Stefansson
- deCODE genetics/Amgen Inc., Sturlugata 8, 101, Reykjavik, Iceland
- Faculty of Medicine, Department of Medicine, University of Iceland, Saemundargata 2, 101, Reykjavik, Iceland
| | - J Gustav Smith
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Cardiology, Clinical Sciences, Lund University and Skåne University Hospital, Lund, Sweden
- Wallenberg Center for Molecular Medicine and Lund University Diabetes Center, Lund University, Lund, Sweden
| | - Ramachandran S Vasan
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA
- Sections of Cardiology, Preventive Medicine and Epidemiology, Department of Medicine, Boston University Schools of Medicine and Public Health, Boston, MA, USA
| | - Daniel I Swerdlow
- Institute of Cardiovascular Science, University College London, London, UK
| | - R Thomas Lumbers
- British Heart Foundation Research Accelerator, University College London, London, UK.
- Institute of Health Informatics, University College London, London, UK.
- Health Data Research UK London, University College London, London, UK.
- Bart's Heart Centre, St. Bartholomew's Hospital, London, UK.
| |
Collapse
|
44
|
He S, Tian Y, Feng S, Wu Y, Shen X, Chen K, He Y, Sun Q, Li X, Xu J, Wen Z, Qu JY. In vivo single-cell lineage tracing in zebrafish using high-resolution infrared laser-mediated gene induction microscopy. eLife 2020; 9:e52024. [PMID: 31904340 PMCID: PMC7018510 DOI: 10.7554/elife.52024] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 01/04/2020] [Indexed: 12/15/2022] Open
Abstract
Heterogeneity broadly exists in various cell types both during development and at homeostasis. Investigating heterogeneity is crucial for comprehensively understanding the complexity of ontogeny, dynamics, and function of specific cell types. Traditional bulk-labeling techniques are incompetent to dissect heterogeneity within cell population, while the new single-cell lineage tracing methodologies invented in the last decade can hardly achieve high-fidelity single-cell labeling and long-term in-vivo observation simultaneously. In this work, we developed a high-precision infrared laser-evoked gene operator heat-shock system, which uses laser-induced CreERT2 combined with loxP-DsRedx-loxP-GFP reporter to achieve precise single-cell labeling and tracing. In vivo study indicated that this system can precisely label single cell in brain, muscle and hematopoietic system in zebrafish embryo. Using this system, we traced the hematopoietic potential of hemogenic endothelium (HE) in the posterior blood island (PBI) of zebrafish embryo and found that HEs in the PBI are heterogeneous, which contains at least myeloid unipotent and myeloid-lymphoid bipotent subtypes.
Collapse
Affiliation(s)
- Sicong He
- Department of Electronic and Computer EngineeringThe Hong Kong University of Science and TechnologyKowloonChina
- State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyKowloonChina
- Center of Systems Biology and Human HealthThe Hong Kong University of Science and TechnologyKowloonChina
| | - Ye Tian
- State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyKowloonChina
- Center of Systems Biology and Human HealthThe Hong Kong University of Science and TechnologyKowloonChina
- Division of Life ScienceThe Hong Kong University of Science and TechnologyKowloonChina
| | - Shachuan Feng
- State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyKowloonChina
- Center of Systems Biology and Human HealthThe Hong Kong University of Science and TechnologyKowloonChina
- Division of Life ScienceThe Hong Kong University of Science and TechnologyKowloonChina
| | - Yi Wu
- State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyKowloonChina
- Center of Systems Biology and Human HealthThe Hong Kong University of Science and TechnologyKowloonChina
- Division of Life ScienceThe Hong Kong University of Science and TechnologyKowloonChina
| | - Xinwei Shen
- Department of MathematicsThe Hong Kong University of Science and TechnologyKowloonChina
| | - Kani Chen
- Department of MathematicsThe Hong Kong University of Science and TechnologyKowloonChina
| | - Yingzhu He
- Department of Electronic and Computer EngineeringThe Hong Kong University of Science and TechnologyKowloonChina
- State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyKowloonChina
- Center of Systems Biology and Human HealthThe Hong Kong University of Science and TechnologyKowloonChina
| | - Qiqi Sun
- Department of Electronic and Computer EngineeringThe Hong Kong University of Science and TechnologyKowloonChina
- State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyKowloonChina
- Center of Systems Biology and Human HealthThe Hong Kong University of Science and TechnologyKowloonChina
| | - Xuesong Li
- Department of Electronic and Computer EngineeringThe Hong Kong University of Science and TechnologyKowloonChina
- State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyKowloonChina
- Center of Systems Biology and Human HealthThe Hong Kong University of Science and TechnologyKowloonChina
| | - Jin Xu
- Division of Cell, Developmental and Integrative Biology, School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Zilong Wen
- State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyKowloonChina
- Center of Systems Biology and Human HealthThe Hong Kong University of Science and TechnologyKowloonChina
- Division of Life ScienceThe Hong Kong University of Science and TechnologyKowloonChina
| | - Jianan Y Qu
- Department of Electronic and Computer EngineeringThe Hong Kong University of Science and TechnologyKowloonChina
- State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyKowloonChina
- Center of Systems Biology and Human HealthThe Hong Kong University of Science and TechnologyKowloonChina
| |
Collapse
|
45
|
The impact of disregarding family structure on genome-wide association analysis of complex diseases in cohorts with simple pedigrees. J Appl Genet 2019; 61:75-86. [PMID: 31755004 DOI: 10.1007/s13353-019-00526-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/19/2019] [Accepted: 10/10/2019] [Indexed: 12/12/2022]
Abstract
The generalized linear mixed models (GLMMs) methodology is the standard framework for genome-wide association studies (GWAS) of complex diseases in family-based cohorts. Fitting GLMMs in very large cohorts, however, can be computationally demanding. Also, the modified versions of GLMM using faster algorithms may underperform, for instance when a single nucleotide polymorphism (SNP) is correlated with fixed-effects covariates. We investigated the extent to which disregarding family structure may compromise GWAS in cohorts with simple pedigrees by contrasting logistic regression models (i.e., with no family structure) to three LMMs-based ones. Our analyses showed that the logistic regression models in general resulted in smaller P values compared with the LMMs-based models; however, the differences in P values were mostly minor. Disregarding family structure had little impact on determining disease-associated SNPs at genome-wide level of significance (i.e., P < 5E-08) as the four P values resulted from the tested methods for any SNP were all below or all above 5E-08. Nevertheless, larger discrepancies were detected between logistic regression and LMMs-based models at suggestive level of significance (i.e., of 5E-08 ≤ P < 5E-06). The SNP effects estimated by the logistic regression models were not statistically different from those estimated by GLMMs that implemented Wald's test. However, several SNP effects were significantly different from their counterparts in LMMs analyses. We suggest that fitting GLMMs with Wald's test on a pre-selected subset of SNPs obtained from logistic regression models can ensure the balance between the speed of analyses and the accuracy of parameters.
Collapse
|
46
|
Noordam R, Bos MM, Wang H, Winkler TW, Bentley AR, Kilpeläinen TO, de Vries PS, Sung YJ, Schwander K, Cade BE, Manning A, Aschard H, Brown MR, Chen H, Franceschini N, Musani SK, Richard M, Vojinovic D, Aslibekyan S, Bartz TM, de las Fuentes L, Feitosa M, Horimoto AR, Ilkov M, Kho M, Kraja A, Li C, Lim E, Liu Y, Mook-Kanamori DO, Rankinen T, Tajuddin SM, van der Spek A, Wang Z, Marten J, Laville V, Alver M, Evangelou E, Graff ME, He M, Kühnel B, Lyytikäinen LP, Marques-Vidal P, Nolte IM, Palmer ND, Rauramaa R, Shu XO, Snieder H, Weiss S, Wen W, Yanek LR, Adolfo C, Ballantyne C, Bielak L, Biermasz NR, Boerwinkle E, Dimou N, Eiriksdottir G, Gao C, Gharib SA, Gottlieb DJ, Haba-Rubio J, Harris TB, Heikkinen S, Heinzer R, Hixson JE, Homuth G, Ikram MA, Komulainen P, Krieger JE, Lee J, Liu J, Lohman KK, Luik AI, Mägi R, Martin LW, Meitinger T, Metspalu A, Milaneschi Y, Nalls MA, O'Connell J, Peters A, Peyser P, Raitakari OT, Reiner AP, Rensen PCN, Rice TK, Rich SS, Roenneberg T, Rotter JI, Schreiner PJ, Shikany J, Sidney SS, Sims M, Sitlani CM, Sofer T, Strauch K, Swertz MA, Taylor KD, Uitterlinden AG, et alNoordam R, Bos MM, Wang H, Winkler TW, Bentley AR, Kilpeläinen TO, de Vries PS, Sung YJ, Schwander K, Cade BE, Manning A, Aschard H, Brown MR, Chen H, Franceschini N, Musani SK, Richard M, Vojinovic D, Aslibekyan S, Bartz TM, de las Fuentes L, Feitosa M, Horimoto AR, Ilkov M, Kho M, Kraja A, Li C, Lim E, Liu Y, Mook-Kanamori DO, Rankinen T, Tajuddin SM, van der Spek A, Wang Z, Marten J, Laville V, Alver M, Evangelou E, Graff ME, He M, Kühnel B, Lyytikäinen LP, Marques-Vidal P, Nolte IM, Palmer ND, Rauramaa R, Shu XO, Snieder H, Weiss S, Wen W, Yanek LR, Adolfo C, Ballantyne C, Bielak L, Biermasz NR, Boerwinkle E, Dimou N, Eiriksdottir G, Gao C, Gharib SA, Gottlieb DJ, Haba-Rubio J, Harris TB, Heikkinen S, Heinzer R, Hixson JE, Homuth G, Ikram MA, Komulainen P, Krieger JE, Lee J, Liu J, Lohman KK, Luik AI, Mägi R, Martin LW, Meitinger T, Metspalu A, Milaneschi Y, Nalls MA, O'Connell J, Peters A, Peyser P, Raitakari OT, Reiner AP, Rensen PCN, Rice TK, Rich SS, Roenneberg T, Rotter JI, Schreiner PJ, Shikany J, Sidney SS, Sims M, Sitlani CM, Sofer T, Strauch K, Swertz MA, Taylor KD, Uitterlinden AG, van Duijn CM, Völzke H, Waldenberger M, Wallance RB, van Dijk KW, Yu C, Zonderman AB, Becker DM, Elliott P, Esko T, Gieger C, Grabe HJ, Lakka TA, Lehtimäki T, North KE, Penninx BWJH, Vollenweider P, Wagenknecht LE, Wu T, Xiang YB, Zheng W, Arnett DK, Bouchard C, Evans MK, Gudnason V, Kardia S, Kelly TN, Kritchevsky SB, Loos RJF, Pereira AC, Province M, Psaty BM, Rotimi C, Zhu X, Amin N, Cupples LA, Fornage M, Fox EF, Guo X, Gauderman WJ, Rice K, Kooperberg C, Munroe PB, Liu CT, Morrison AC, Rao DC, van Heemst D, Redline S. Multi-ancestry sleep-by-SNP interaction analysis in 126,926 individuals reveals lipid loci stratified by sleep duration. Nat Commun 2019; 10:5121. [PMID: 31719535 PMCID: PMC6851116 DOI: 10.1038/s41467-019-12958-0] [Show More Authors] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 10/04/2019] [Indexed: 12/12/2022] Open
Abstract
Both short and long sleep are associated with an adverse lipid profile, likely through different biological pathways. To elucidate the biology of sleep-associated adverse lipid profile, we conduct multi-ancestry genome-wide sleep-SNP interaction analyses on three lipid traits (HDL-c, LDL-c and triglycerides). In the total study sample (discovery + replication) of 126,926 individuals from 5 different ancestry groups, when considering either long or short total sleep time interactions in joint analyses, we identify 49 previously unreported lipid loci, and 10 additional previously unreported lipid loci in a restricted sample of European-ancestry cohorts. In addition, we identify new gene-sleep interactions for known lipid loci such as LPL and PCSK9. The previously unreported lipid loci have a modest explained variance in lipid levels: most notable, gene-short-sleep interactions explain 4.25% of the variance in triglyceride level. Collectively, these findings contribute to our understanding of the biological mechanisms involved in sleep-associated adverse lipid profiles.
Collapse
Affiliation(s)
- Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands.
| | - Maxime M Bos
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Heming Wang
- Division of Sleep and Circadian Disorders, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Thomas W Winkler
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tuomas O Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
- Department of Environmental Medicine and Public Health, The Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yun Ju Sung
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Karen Schwander
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian E Cade
- Division of Sleep and Circadian Disorders, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Alisa Manning
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Hugues Aschard
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
- Centre de Bioinformatique, Biostatistique et Biologie Intégrative (C3BI), Institut Pasteur, Paris, France
| | - Michael R Brown
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Han Chen
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Precision Health, School of Public Health & School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Solomon K Musani
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Melissa Richard
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Dina Vojinovic
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Stella Aslibekyan
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Biostatistics and Medicine, University of Washington, Seattle, WA, USA
| | - Lisa de las Fuentes
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Mary Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrea R Horimoto
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, SP, Brazil
| | | | - Minjung Kho
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Aldi Kraja
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Changwei Li
- Epidemiology and Biostatistics, University of Georgia at Athens College of Public Health, Athens, GA, USA
| | - Elise Lim
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Yongmei Liu
- Public Health Sciences, Epidemiology and Prevention, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, Netherlands
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Salman M Tajuddin
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Ashley van der Spek
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Zhe Wang
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jonathan Marten
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Vincent Laville
- Centre de Bioinformatique, Biostatistique et Biologie Intégrative (C3BI), Institut Pasteur, Paris, France
| | - Maris Alver
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Maria E Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Meian He
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Brigitte Kühnel
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Pedro Marques-Vidal
- Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Ilja M Nolte
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, The Netherlands
| | | | - Rainer Rauramaa
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Harold Snieder
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, The Netherlands
| | - Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Lisa R Yanek
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Correa Adolfo
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Christie Ballantyne
- Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX, USA
- Houston Methodist Debakey Heart and Vascular Center, Houston, TX, USA
| | - Larry Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Nienke R Biermasz
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden, The Netherlands
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Niki Dimou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | | | - Chuan Gao
- Molecular Genetics and Genomics Program, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Sina A Gharib
- Computational Medicine Core, Center for Lung Biology, UW Medicine Sleep Center, Medicine, University of Washington, Seattle, WA, USA
| | - Daniel J Gottlieb
- Division of Sleep and Circadian Disorders, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - José Haba-Rubio
- Medicine, Sleep Laboratory, Lausanne University Hospital, Lausanne, Switzerland
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Sami Heikkinen
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus, Finland
| | - Raphaël Heinzer
- Medicine, Sleep Laboratory, Lausanne University Hospital, Lausanne, Switzerland
| | - James E Hixson
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Pirjo Komulainen
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Jose E Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Jiwon Lee
- Division of Sleep and Circadian Disorders, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Jingmin Liu
- Fred Hutchinson Cancer Research Center, University of Washington School of Public Health, Seattle, WA, USA
| | - Kurt K Lohman
- Public Health Sciences, Biostatistical Sciences, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Annemarie I Luik
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Reedik Mägi
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Lisa W Martin
- Cardiology, School of Medicine and Health Sciences, George Washington University, Washington, D.C., USA
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Andres Metspalu
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Yuri Milaneschi
- Cardiology, School of Medicine and Health Sciences, George Washington University, Washington, D.C., USA
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Data Tecnica International, Glen Echo, MD, USA
| | - Jeff O'Connell
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
- Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Neuherberg, Germany
| | - Patricia Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- University of Turku, Turku, Finland
| | - Alex P Reiner
- Fred Hutchinson Cancer Research Center, University of Washington School of Public Health, Seattle, WA, USA
| | - Patrick C N Rensen
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden, The Netherlands
| | - Treva K Rice
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Till Roenneberg
- Institute of Medical Psychology, Ludwig-Maximilians-Universitat Munchen, Munich, Germany
| | - Jerome I Rotter
- Genomic Outcomes, Department of Pediatrics, Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, CC, USA
| | - Pamela J Schreiner
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - James Shikany
- Division of Preventive Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stephen S Sidney
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Mario Sims
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Medicine, University of Washington, Seattle, WA, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
- Institute of Human Genetics, Technische Universität München, Munich, Germany
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute for Medical Informatics Biometry and Epidemiology, Ludwig-Maximilians-Universitat Munchen, Munich, Germany
| | - Morris A Swertz
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Kent D Taylor
- Genomic Outcomes, Department of Pediatrics, Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, CC, USA
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Neuherberg, Germany
| | - Robert B Wallance
- Department of Epidemiology, University of Iowa College of Public Health, Iowa City, IA, USA
| | - Ko Willems van Dijk
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden, The Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Caizheng Yu
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Alan B Zonderman
- Behavioral Epidemiology Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Diane M Becker
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Paul Elliott
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
- MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- National Institute of Health Research Imperial College London Biomedical Research Centre, London, UK
- UK-DRI Dementia Research Institute at Imperial College London, London, UK
| | - Tõnu Esko
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Boston, MA, USA
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Hans J Grabe
- Department Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Timo A Lakka
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus, Finland
- Department of Clinical Phsiology and Nuclear Medicine, Kuopia University Hospital, Kuopio, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Peter Vollenweider
- Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Lynne E Wagenknecht
- Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Tangchun Wu
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong-Bing Xiang
- SKLORG & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P. R. China
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Donna K Arnett
- Dean's Office, University of Kentucky College of Public Health, Lexington, KS, USA
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Michele K Evans
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Sharon Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Tanika N Kelly
- Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Stephen B Kritchevsky
- Sticht Center for Healthy Aging and Rehabilitation, Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health Development Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandre C Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Mike Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Epidemiology, Medicine and Health Services, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington, Health Research Institute, Seattle, WA, USA
| | - Charles Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xiaofeng Zhu
- Department of Population Quantitative and Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Najaf Amin
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - L Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- NHLBI Framingham Heart Study, Framingham, MA, USA
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ervin F Fox
- Cardiology, Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Xiuqing Guo
- Genomic Outcomes, Department of Pediatrics, Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, CC, USA
| | - W James Gauderman
- Biostatistics, Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kenneth Rice
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Charles Kooperberg
- Fred Hutchinson Cancer Research Center, University of Washington School of Public Health, Seattle, WA, USA
| | - Patricia B Munroe
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- NIHR Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, London, UK
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Dabeeru C Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Diana van Heemst
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA.
- Division of Pulmonary Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
47
|
Fang S, Xu T, Xiong M, Zhou X, Wang Y, Haydu LE, Ross MI, Gershenwald JE, Prieto VG, Cormier JN, Wargo J, Sui D, Wei Q, Amos CI, Lee JE. Role of Immune Response, Inflammation, and Tumor Immune Response-Related Cytokines/Chemokines in Melanoma Progression. J Invest Dermatol 2019; 139:2352-2358.e3. [PMID: 31176707 PMCID: PMC6814532 DOI: 10.1016/j.jid.2019.03.1158] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/07/2019] [Accepted: 03/24/2019] [Indexed: 01/12/2023]
Abstract
To investigate the role of tumor cytokines/chemokines in melanoma immune response, we estimated the proportions of immune cell subsets in melanoma tumors from The Cancer Genome Atlas, followed by evaluation of the association between cytokine/chemokine expression and these subsets. We then investigated the association of immune cell subsets, chemokines, and cytokines with patient survival. Finally, we evaluated the immune cell tumor-infiltrating lymphocyte (TIL) score for correlation with melanoma patient outcome in a separate cohort. There was good agreement between RNA sequencing estimation of T-cell subset and pathologist-determined TIL score. Expression levels of cytokines IL-12A, IFNG, and IL-10, and chemokines CXCL9 and CXCL10 were positively correlated with PDCD1, CTLA-4, and CD8+ T-cell subset, but negatively correlated with tumor purity (Bonferroni-corrected P < 0.05). In multivariable analysis, higher expression levels of cytokines IFN-γ and TGFB1, but not chemokines, were associated with improved overall survival. A higher expression level of CD8+ T-cell subset was also associated with improved overall survival (hazard ratio [HR] = 0.06, 95% confidence interval [CI] = 0.01-0.35, P = 0.002). Finally, multivariable analysis showed that patients with a brisk TIL score had improved melanoma-specific survival than those with a nonbrisk score (HR = 0.51, 95% CI = 0.27-0.98, P = 0.0423). These results suggest that the expression of specific tumor cytokines represents important biomarkers of melanoma immune response.
Collapse
Affiliation(s)
- Shenying Fang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| | - Tao Xu
- Department of Biostatistics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Momiao Xiong
- Department of Biostatistics, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xinke Zhou
- The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yuling Wang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lauren E Haydu
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Merrick I Ross
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Victor G Prieto
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Janice N Cormier
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jennifer Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Dawen Sui
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Qingyi Wei
- Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina, USA; Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA
| | | | - Jeffrey E Lee
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
48
|
Veluchamy A, Ballerini L, Vitart V, Schraut KE, Kirin M, Campbell H, Joshi PK, Relan D, Harris S, Brown E, Vaidya SS, Dhillon B, Zhou K, Pearson ER, Hayward C, Polasek O, Deary IJ, MacGillivray T, Wilson JF, Trucco E, Palmer CNA, Doney ASF. Novel Genetic Locus Influencing Retinal Venular Tortuosity Is Also Associated With Risk of Coronary Artery Disease. Arterioscler Thromb Vasc Biol 2019; 39:2542-2552. [PMID: 31597446 PMCID: PMC6882544 DOI: 10.1161/atvbaha.119.312552] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Supplemental Digital Content is available in the text. The retina may provide readily accessible imaging biomarkers of global cardiovascular health. Increasing evidence suggests variation in retinal vascular traits is highly heritable. This study aimed to identify the genetic determinants of retinal vascular traits.
Collapse
Affiliation(s)
- Abirami Veluchamy
- From the Division of Population Health and Genomics (A.V., E.R.P., C.N.A.P., A.S.F.D.), University of Dundee, United Kingdom
| | - Lucia Ballerini
- Ninewells Hospital and Medical School and VAMPIRE project, Computer Vision and Image Processing Group, School of Science and Engineering (Computing) (L.B., E.T.), University of Dundee, United Kingdom.,VAMPIRE project, Centre for Clinical Brain Sciences, Chancellor's Building, Royal Infirmary of Edinburgh, Scotland, United Kingdom (L.B., D.R., B.D., T.M.)
| | - Veronique Vitart
- MRC Human Genetics Unit (V.V., C.H., J.F.W.), MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, United Kingdom
| | - Katharina E Schraut
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Scotland, United Kingdom (K.E.S., M.K., H.C., P.K.J., J.F.W.).,Centre for Cardiovascular Science (K.E.S.), Queen's Medical Research Institute, University of Edinburgh, Royal Infirmary of Edinburgh, Scotland, United Kingdom
| | - Mirna Kirin
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Scotland, United Kingdom (K.E.S., M.K., H.C., P.K.J., J.F.W.).,Department of Public Health, University of Split, School of Medicine, Croatia (M.K., O.P.)
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Scotland, United Kingdom (K.E.S., M.K., H.C., P.K.J., J.F.W.)
| | - Peter K Joshi
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Scotland, United Kingdom (K.E.S., M.K., H.C., P.K.J., J.F.W.)
| | - Devanjali Relan
- VAMPIRE project, Centre for Clinical Brain Sciences, Chancellor's Building, Royal Infirmary of Edinburgh, Scotland, United Kingdom (L.B., D.R., B.D., T.M.).,Department of Computer Science, BML Munjal University, Gurgaon, Haryana, India (D.R.)
| | - Sarah Harris
- Medical Genetics Section, Centre for Genomic and Experimental Medicine (S.H.), MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, United Kingdom.,Centre for Cognitive Ageing and Cognitive Epidemiology (S.H., I.J.D.), University of Edinburgh, United Kingdom.,Department of Psychology (S.H.), University of Edinburgh, United Kingdom
| | - Ellie Brown
- Clinical Research Imaging Centre (E.B., S.S.V.), Queen's Medical Research Institute, University of Edinburgh, Royal Infirmary of Edinburgh, Scotland, United Kingdom
| | - Suraj S Vaidya
- Clinical Research Imaging Centre (E.B., S.S.V.), Queen's Medical Research Institute, University of Edinburgh, Royal Infirmary of Edinburgh, Scotland, United Kingdom
| | - Baljean Dhillon
- VAMPIRE project, Centre for Clinical Brain Sciences, Chancellor's Building, Royal Infirmary of Edinburgh, Scotland, United Kingdom (L.B., D.R., B.D., T.M.)
| | - Kaixin Zhou
- Renji Hospital, University of Chinese Academy of Sciences, Chongqing, China (K.Z.)
| | - Ewan R Pearson
- From the Division of Population Health and Genomics (A.V., E.R.P., C.N.A.P., A.S.F.D.), University of Dundee, United Kingdom
| | - Caroline Hayward
- MRC Human Genetics Unit (V.V., C.H., J.F.W.), MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, United Kingdom
| | - Ozren Polasek
- Department of Public Health, University of Split, School of Medicine, Croatia (M.K., O.P.)
| | - Ian J Deary
- Department of Psychology (I.J.D.), University of Edinburgh, United Kingdom.,Centre for Cognitive Ageing and Cognitive Epidemiology (S.H., I.J.D.), University of Edinburgh, United Kingdom
| | - Thomas MacGillivray
- VAMPIRE project, Centre for Clinical Brain Sciences, Chancellor's Building, Royal Infirmary of Edinburgh, Scotland, United Kingdom (L.B., D.R., B.D., T.M.)
| | - James F Wilson
- MRC Human Genetics Unit (V.V., C.H., J.F.W.), MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, United Kingdom.,Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Scotland, United Kingdom (K.E.S., M.K., H.C., P.K.J., J.F.W.)
| | - Emanuele Trucco
- Ninewells Hospital and Medical School and VAMPIRE project, Computer Vision and Image Processing Group, School of Science and Engineering (Computing) (L.B., E.T.), University of Dundee, United Kingdom
| | - Colin N A Palmer
- From the Division of Population Health and Genomics (A.V., E.R.P., C.N.A.P., A.S.F.D.), University of Dundee, United Kingdom
| | - Alexander S F Doney
- From the Division of Population Health and Genomics (A.V., E.R.P., C.N.A.P., A.S.F.D.), University of Dundee, United Kingdom
| |
Collapse
|
49
|
Tin A, Marten J, Halperin Kuhns VL, Li Y, Wuttke M, Kirsten H, Sieber KB, Qiu C, Gorski M, Yu Z, Giri A, Sveinbjornsson G, Li M, Chu AY, Hoppmann A, O'Connor LJ, Prins B, Nutile T, Noce D, Akiyama M, Cocca M, Ghasemi S, van der Most PJ, Horn K, Xu Y, Fuchsberger C, Sedaghat S, Afaq S, Amin N, Ärnlöv J, Bakker SJL, Bansal N, Baptista D, Bergmann S, Biggs ML, Biino G, Boerwinkle E, Bottinger EP, Boutin TS, Brumat M, Burkhardt R, Campana E, Campbell A, Campbell H, Carroll RJ, Catamo E, Chambers JC, Ciullo M, Concas MP, Coresh J, Corre T, Cusi D, Felicita SC, de Borst MH, De Grandi A, de Mutsert R, de Vries APJ, Delgado G, Demirkan A, Devuyst O, Dittrich K, Eckardt KU, Ehret G, Endlich K, Evans MK, Gansevoort RT, Gasparini P, Giedraitis V, Gieger C, Girotto G, Gögele M, Gordon SD, Gudbjartsson DF, Gudnason V, Haller T, Hamet P, Harris TB, Hayward C, Hicks AA, Hofer E, Holm H, Huang W, Hutri-Kähönen N, Hwang SJ, Ikram MA, Lewis RM, Ingelsson E, Jakobsdottir J, Jonsdottir I, Jonsson H, Joshi PK, Josyula NS, Jung B, Kähönen M, Kamatani Y, Kanai M, Kerr SM, Kiess W, Kleber ME, Koenig W, et alTin A, Marten J, Halperin Kuhns VL, Li Y, Wuttke M, Kirsten H, Sieber KB, Qiu C, Gorski M, Yu Z, Giri A, Sveinbjornsson G, Li M, Chu AY, Hoppmann A, O'Connor LJ, Prins B, Nutile T, Noce D, Akiyama M, Cocca M, Ghasemi S, van der Most PJ, Horn K, Xu Y, Fuchsberger C, Sedaghat S, Afaq S, Amin N, Ärnlöv J, Bakker SJL, Bansal N, Baptista D, Bergmann S, Biggs ML, Biino G, Boerwinkle E, Bottinger EP, Boutin TS, Brumat M, Burkhardt R, Campana E, Campbell A, Campbell H, Carroll RJ, Catamo E, Chambers JC, Ciullo M, Concas MP, Coresh J, Corre T, Cusi D, Felicita SC, de Borst MH, De Grandi A, de Mutsert R, de Vries APJ, Delgado G, Demirkan A, Devuyst O, Dittrich K, Eckardt KU, Ehret G, Endlich K, Evans MK, Gansevoort RT, Gasparini P, Giedraitis V, Gieger C, Girotto G, Gögele M, Gordon SD, Gudbjartsson DF, Gudnason V, Haller T, Hamet P, Harris TB, Hayward C, Hicks AA, Hofer E, Holm H, Huang W, Hutri-Kähönen N, Hwang SJ, Ikram MA, Lewis RM, Ingelsson E, Jakobsdottir J, Jonsdottir I, Jonsson H, Joshi PK, Josyula NS, Jung B, Kähönen M, Kamatani Y, Kanai M, Kerr SM, Kiess W, Kleber ME, Koenig W, Kooner JS, Körner A, Kovacs P, Krämer BK, Kronenberg F, Kubo M, Kühnel B, La Bianca M, Lange LA, Lehne B, Lehtimäki T, Liu J, Loeffler M, Loos RJF, Lyytikäinen LP, Magi R, Mahajan A, Martin NG, März W, Mascalzoni D, Matsuda K, Meisinger C, Meitinger T, Metspalu A, Milaneschi Y, O'Donnell CJ, Wilson OD, Gaziano JM, Mishra PP, Mohlke KL, Mononen N, Montgomery GW, Mook-Kanamori DO, Müller-Nurasyid M, Nadkarni GN, Nalls MA, Nauck M, Nikus K, Ning B, Nolte IM, Noordam R, O'Connell JR, Olafsson I, Padmanabhan S, Penninx BWJH, Perls T, Peters A, Pirastu M, Pirastu N, Pistis G, Polasek O, Ponte B, Porteous DJ, Poulain T, Preuss MH, Rabelink TJ, Raffield LM, Raitakari OT, Rettig R, Rheinberger M, Rice KM, Rizzi F, Robino A, Rudan I, Krajcoviechova A, Cifkova R, Rueedi R, Ruggiero D, Ryan KA, Saba Y, Salvi E, Schmidt H, Schmidt R, Shaffer CM, Smith AV, Smith BH, Spracklen CN, Strauch K, Stumvoll M, Sulem P, Tajuddin SM, Teren A, Thiery J, Thio CHL, Thorsteinsdottir U, Toniolo D, Tönjes A, Tremblay J, Uitterlinden AG, Vaccargiu S, van der Harst P, van Duijn CM, Verweij N, Völker U, Vollenweider P, Waeber G, Waldenberger M, Whitfield JB, Wild SH, Wilson JF, Yang Q, Zhang W, Zonderman AB, Bochud M, Wilson JG, Pendergrass SA, Ho K, Parsa A, Pramstaller PP, Psaty BM, Böger CA, Snieder H, Butterworth AS, Okada Y, Edwards TL, Stefansson K, Susztak K, Scholz M, Heid IM, Hung AM, Teumer A, Pattaro C, Woodward OM, Vitart V, Köttgen A. Target genes, variants, tissues and transcriptional pathways influencing human serum urate levels. Nat Genet 2019; 51:1459-1474. [PMID: 31578528 PMCID: PMC6858555 DOI: 10.1038/s41588-019-0504-x] [Show More Authors] [Citation(s) in RCA: 252] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 08/27/2019] [Indexed: 12/22/2022]
Abstract
Elevated serum urate levels cause gout and correlate with cardiometabolic diseases via poorly understood mechanisms. We performed a trans-ancestry genome-wide association study of serum urate in 457,690 individuals, identifying 183 loci (147 previously unknown) that improve the prediction of gout in an independent cohort of 334,880 individuals. Serum urate showed significant genetic correlations with many cardiometabolic traits, with genetic causality analyses supporting a substantial role for pleiotropy. Enrichment analysis, fine-mapping of urate-associated loci and colocalization with gene expression in 47 tissues implicated the kidney and liver as the main target organs and prioritized potentially causal genes and variants, including the transcriptional master regulators in the liver and kidney, HNF1A and HNF4A. Experimental validation showed that HNF4A transactivated the promoter of ABCG2, encoding a major urate transporter, in kidney cells, and that HNF4A p.Thr139Ile is a functional variant. Transcriptional coregulation within and across organs may be a general mechanism underlying the observed pleiotropy between urate and cardiometabolic traits.
Collapse
Affiliation(s)
- Adrienne Tin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Welch Centre for Prevention, Epidemiology and Clinical Research, Baltimore, MD, USA.
| | - Jonathan Marten
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | | | - Yong Li
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Matthias Wuttke
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Karsten B Sieber
- Target Sciences-Genetics, GlaxoSmithKline, Collegeville, PA, USA
| | - Chengxiang Qiu
- Department of Medicine and Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Mathias Gorski
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Zhi Yu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ayush Giri
- Division of Quantitative Sciences, Department of Obstetrics & Gynecology, Vanderbilt Genetics Institute, Vanderbilt Epidemiology Center, Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, TN, USA
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
| | | | - Man Li
- Department of Medicine, Division of Nephrology and Hypertension, University of Utah, Salt Lake City, UT, USA
| | | | - Anselm Hoppmann
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Luke J O'Connor
- Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Bram Prins
- Strangeways Research Laboratory, University of Cambridge, Cambridge, UK
| | - Teresa Nutile
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso-CNR, Naples, Italy
| | - Damia Noce
- Eurac Research, Institute for Biomedicine, Bolzano, Italy
| | - Masato Akiyama
- Laboratory for Statistical Analysis, RIKEN Centre for Integrative Medical Sciences, Yokohama (Kanagawa), Japan
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Massimiliano Cocca
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - Sahar Ghasemi
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Peter J van der Most
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Katrin Horn
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Yizhe Xu
- Department of Medicine, Division of Nephrology and Hypertension, University of Utah, Salt Lake City, UT, USA
| | | | - Sanaz Sedaghat
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Saima Afaq
- Department of Epidemiology and Biostatistics, Faculty of Medicine, School of Public Health, Imperial College London, London, UK
- Institute of Public Health & Social Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Najaf Amin
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Johan Ärnlöv
- Department of Neurobiology, Care Sciences and Society, Division of Family Medicine and Primary Care, Karolinska Institutet, Stockholm, Sweden
- School of Health and Social Studies, Dalarna University, Falun, Sweden
| | - Stephan J L Bakker
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Nisha Bansal
- Division of Nephrology, University of Washington, Seattle, WA, USA
- Kidney Research Institute, University of Washington, Seattle, WA, USA
| | | | - Sven Bergmann
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Mary L Biggs
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Ginevra Biino
- Institute of Molecular Genetics, National Research Council of Italy, Pavia, Italy
| | - Eric Boerwinkle
- Human Genetics Centre, University of Texas Health Science Centre, Houston, TX, USA
| | - Erwin P Bottinger
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thibaud S Boutin
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Marco Brumat
- University of Trieste, Department of Medicine, Surgery and Health Sciences, Trieste, Italy
| | - Ralph Burkhardt
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig, Leipzig, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Eric Campana
- University of Trieste, Department of Medicine, Surgery and Health Sciences, Trieste, Italy
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Robert J Carroll
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eulalia Catamo
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - John C Chambers
- Department of Epidemiology and Biostatistics, Faculty of Medicine, School of Public Health, Imperial College London, London, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Cardiology, Ealing Hospital, London, UK
- Imperial College Healthcare NHS Trust, Imperial College London, London, UK
- MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | - Marina Ciullo
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso-CNR, Naples, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Maria Pina Concas
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Tanguy Corre
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - Daniele Cusi
- Institute of Biomedical Technologies, Italy National Research Council, Milano, Italy
- Bio4Dreams, Milano, Italy
| | | | - Martin H de Borst
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Aiko P J de Vries
- Section of Nephrology, Department of Internal Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| | - Graciela Delgado
- Fifth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Ayşe Demirkan
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Genetics, University Medical Center Groningen, Groningen, the Netherlands
| | - Olivier Devuyst
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Katalin Dittrich
- Department of Women and Child Health, Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany
- Centre for Pediatric Research, University of Leipzig, Leipzig, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Georg Ehret
- Cardiology, Geneva University Hospitals, Geneva, Switzerland
| | - Karlhans Endlich
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Ron T Gansevoort
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Paolo Gasparini
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
- University of Trieste, Department of Medicine, Surgery and Health Sciences, Trieste, Italy
| | - Vilmantas Giedraitis
- Department of Public Health and Caring Sciences, Molecular Geriatrics, Uppsala University, Uppsala, Sweden
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Giorgia Girotto
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
- University of Trieste, Department of Medicine, Surgery and Health Sciences, Trieste, Italy
| | - Martin Gögele
- Eurac Research, Institute for Biomedicine, Bolzano, Italy
| | - Scott D Gordon
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Vilmundur Gudnason
- Icelandic Heart Association, Kópavogur, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Toomas Haller
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Pavel Hamet
- Montreal University Hospital Research Centre, Centre Hospitalier de lUniversité de Montréal, Montreal, Quebec, Canada
- Medpharmgene, Montreal, Quebec, Canada
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Andrew A Hicks
- Eurac Research, Institute for Biomedicine, Bolzano, Italy
| | - Edith Hofer
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria
| | - Hilma Holm
- deCODE Genetics, Amgen Inc., Reykjavik, Iceland
| | - Wei Huang
- Department of Genetics, Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Centre, Shanghai, China
- Shanghai Industrial Technology Institute, Shanghai, China
| | - Nina Hutri-Kähönen
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Pediatrics, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Shih-Jen Hwang
- National Heart, Lung, and Blood Institute Framingham Heart Study, Framingham, MA, USA
- The Centre for Population Studies, National Heart, Lung, and Blood Institute, Framingham, MA, USA
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Raychel M Lewis
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Erik Ingelsson
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Molecular Epidemiology and Science for Life Laboratory, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Stanford Diabetes Research Center, Stanford University, Stanford, CA, USA
| | - Johanna Jakobsdottir
- Icelandic Heart Association, Kópavogur, Iceland
- The Centre of Public Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | - Helgi Jonsson
- Landspitalinn University Hospital, Reykjavík, Iceland
- University of Iceland, Reykjavík, Iceland
| | - Peter K Joshi
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Navya Shilpa Josyula
- Geisinger Research, Biomedical and Translational Informatics Institute, Rockville, MD, USA
| | - Bettina Jung
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, and Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Centre for Integrative Medical Sciences, Yokohama (Kanagawa), Japan
- Kyoto-McGill International Collaborative School in Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masahiro Kanai
- Laboratory for Statistical Analysis, RIKEN Centre for Integrative Medical Sciences, Yokohama (Kanagawa), Japan
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Shona M Kerr
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Wieland Kiess
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Department of Women and Child Health, Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany
- Centre for Pediatric Research, University of Leipzig, Leipzig, Germany
| | - Marcus E Kleber
- Fifth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Wolfgang Koenig
- Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Institute of Epidemiology and Biostatistics, University of Ulm, Ulm, Germany
| | - Jaspal S Kooner
- Department of Cardiology, Ealing Hospital, London, UK
- Imperial College Healthcare NHS Trust, Imperial College London, London, UK
- MRC-PHE Centre for Environment and Health, 323 School of Public Health, Imperial College London, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Antje Körner
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Department of Women and Child Health, Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany
- Centre for Pediatric Research, University of Leipzig, Leipzig, Germany
| | - Peter Kovacs
- Integrated Research and Treatment Centre Adiposity Diseases, University of Leipzig, Leipzig, Germany
| | - Bernhard K Krämer
- Fifth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Florian Kronenberg
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Michiaki Kubo
- RIKEN Centre for Integrative Medical Sciences, Yokohama (Kanagawa), Japan
| | - Brigitte Kühnel
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
| | - Martina La Bianca
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - Leslie A Lange
- Division of Biomedical Informatics and Personalized Medicine, School of Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, CO, USA
| | - Benjamin Lehne
- Department of Epidemiology and Biostatistics, Faculty of Medicine, School of Public Health, Imperial College London, London, UK
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jun Liu
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Reedik Magi
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Nicholas G Martin
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Winfried März
- Fifth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Synlab Academy, Synlab Holding Deutschland GmbH, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | | | - Koichi Matsuda
- Laboratory of Clinical Genome Sequencing, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Christa Meisinger
- Independent Research Group Clinical Epidemiology, Helmholtz Zentrum München, German Research Centre for Environmental Health, Neuherberg, Germany
- Ludwig-Maximilians-Universität München at UNIKA-T Augsburg, Augsburg, Germany
| | - Thomas Meitinger
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Human Genetics, Technische Universität München, Munich, Germany
| | - Andres Metspalu
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Christopher J O'Donnell
- VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Otis D Wilson
- Vanderbilt University Medical Centre, Division of Nephrology & Hypertension, Nashville, TN, USA
| | - J Michael Gaziano
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Massachusetts Veterans Epidemiology Research and Information Center, VA Cooperative Studies Program, VA Boston Healthcare System, Boston, MA, USA
| | - Pashupati P Mishra
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Nina Mononen
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | | | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Centre, Leiden, the Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Centre, Leiden, the Netherlands
| | - Martina Müller-Nurasyid
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
- Chair of Genetic Epidemiology, IBE, Faculty of Medicine, LMU Munich, Munich, Germany
- Department of Internal Medicine I (Cardiology), Hospital of the Ludwig-Maximilians-University Munich, Munich, Germany
| | - Girish N Nadkarni
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Data Tecnica International, Glen Echo, MD, USA
| | - Matthias Nauck
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Kjell Nikus
- Department of Cardiology, Heart Center, Tampere University Hospital, Tampere, Finland
- Department of Cardiology, Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Boting Ning
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Ilja M Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Raymond Noordam
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Centre, Leiden, the Netherlands
| | | | - Isleifur Olafsson
- Department of Clinical Biochemistry, Landspitali University Hospital, Reykjavik, Iceland
| | - Sandosh Padmanabhan
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Thomas Perls
- Department of Medicine, Geriatrics Section, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Mario Pirastu
- Institute of Genetic and Biomedical Research, National Research Council of Italy, UOS of Sassari, Sassari, Italy
| | - Nicola Pirastu
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Giorgio Pistis
- Department of Psychiatry, University Hospital of Lausanne, Lausanne, Switzerland
| | - Ozren Polasek
- Faculty of Medicine, University of Split, Split, Croatia
- Gen-info Ltd, Zagreb, Croatia
| | - Belen Ponte
- Nephrology Service, Department of Specialties in Internal Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - David J Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Tanja Poulain
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Michael H Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ton J Rabelink
- Section of Nephrology, Department of Internal Medicine, Leiden University Medical Centre, Leiden, the Netherlands
- Einthoven Laboratory of Experimental Vascular Research, Leiden University Medical Centre, Leiden, the Netherlands
| | - Laura M Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Rainer Rettig
- Institute of Physiology, University Medicine Greifswald, Karlsburg, Germany
| | - Myriam Rheinberger
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Kenneth M Rice
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Federica Rizzi
- Department of Health Sciences, University of Milan, Milano, Italy
- ePhood Scientific Unit, ePhood SRL, Milano, Italy
| | - Antonietta Robino
- Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Alena Krajcoviechova
- Center for Cardiovascular Prevention, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
- Thomayer Hospital, Prague, Czech Republic
| | - Renata Cifkova
- Center for Cardiovascular Prevention, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
- Department of Medicine II, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Rico Rueedi
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Daniela Ruggiero
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso-CNR, Naples, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Kathleen A Ryan
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yasaman Saba
- Molecular Biology and Biochemistry, Gottfried Schatz Research Centre for Cell Signaling, Metabolism and Aging, Medical University of Graz, Graz, Austria
| | - Erika Salvi
- Department of Health Sciences, University of Milan, Milano, Italy
- Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Helena Schmidt
- Institute of Molecular Biology and Biochemistry, Centre for Molecular Medicine, Medical University of Graz, Graz, Austria
| | - Reinhold Schmidt
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, Graz, Austria
| | - Christian M Shaffer
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Albert V Smith
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Blair H Smith
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | | | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
- Chair of Genetic Epidemiology, IBE, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Michael Stumvoll
- Division of Endocrinology, Nephrology and Rheumatology, University of Leipzig, Leipzig, Germany
| | | | - Salman M Tajuddin
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Andrej Teren
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Heart Centre Leipzig, Leipzig, Germany
| | - Joachim Thiery
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University of Leipzig, Leipzig, Germany
| | - Chris H L Thio
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | | | - Anke Tönjes
- Department of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
| | - Johanne Tremblay
- Montreal University Hospital Research Centre, Centre Hospitalier de lUniversité de Montréal, Montreal, Quebec, Canada
- Centre de Recherche du CHUM, Montreal, Quebec, Canada
| | - André G Uitterlinden
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Simona Vaccargiu
- Institute of Genetic and Biomedical Research, National Research Council of Italy, UOS of Sassari, Sassari, Italy
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Durrer Centre for Cardiovascular Research, the Netherlands Heart Institute, Utrecht, the Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Niek Verweij
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Genomics plc, Oxford, UK
| | - Uwe Völker
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Peter Vollenweider
- Internal Medicine, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Gerard Waeber
- Internal Medicine, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Centre for Environmental Health, Neuherberg, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - John B Whitfield
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Sarah H Wild
- Centre for Population Health Sciences, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - James F Wilson
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Faculty of Medicine, School of Public Health, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, London, UK
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Murielle Bochud
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Centre, Jackson, MS, USA
| | - Sarah A Pendergrass
- Geisinger Research, Biomedical and Translational Informatics Institute, Danville, PA, USA
| | - Kevin Ho
- Kidney Health Research Institute, Geisinger, Danville, PA, USA
- Department of Nephrology, Geisinger, Danville, PA, USA
| | - Afshin Parsa
- Division of Kidney, Urologic and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, Department of Epidemiology, Department of Health Service, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Carsten A Böger
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
- Department of Nephrology and Rheumatology, Kliniken Südostbayern AG, Traunstein, Germany
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Adam S Butterworth
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Yukinori Okada
- Laboratory for Statistical Analysis, RIKEN Centre for Integrative Medical Sciences, Osaka, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Centre, Nashville, TN, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
| | | | - Katalin Susztak
- Department of Medicine and Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - Iris M Heid
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Adriana M Hung
- Vanderbilt University Medical Centre, Division of Nephrology & Hypertension, Nashville, TN, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | | | - Owen M Woodward
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Veronique Vitart
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Anna Köttgen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany.
| |
Collapse
|
50
|
Seddighi S, Houck AL, Rowe JB, Pharoah PDP. Evidence of a Causal Association Between Cancer and Alzheimer's Disease: a Mendelian Randomization Analysis. Sci Rep 2019; 9:13548. [PMID: 31537833 PMCID: PMC6753207 DOI: 10.1038/s41598-019-49795-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/29/2019] [Indexed: 01/08/2023] Open
Abstract
While limited observational evidence suggests that cancer survivors have a decreased risk of developing Alzheimer's disease (AD), and vice versa, it is not clear whether this relationship is causal. Using a Mendelian randomization approach that provides evidence of causality, we found that genetically predicted lung cancer (OR 0.91, 95% CI 0.84-0.99, p = 0.019), leukemia (OR 0.98, 95% CI 0.96-0.995, p = 0.012), and breast cancer (OR 0.94, 95% CI 0.89-0.99, p = 0.028) were associated with 9.0%, 2.4%, and 5.9% lower odds of AD, respectively, per 1-unit higher log odds of cancer. When genetic predictors of all cancers were pooled, cancer was associated with 2.5% lower odds of AD (OR 0.98, 95% CI 0.96-0.988, p = 0.00027) per 1-unit higher log odds of cancer. Finally, genetically predicted smoking-related cancers showed a more robust inverse association with AD than non-smoking related cancers (OR 0.95, 95% CI 0.92-0.98, p = 0.0026, vs. OR 0.98, 95% CI 0.97-0.995, p = 0.0091).
Collapse
Affiliation(s)
- Sahba Seddighi
- University of Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
- Johns Hopkins University School of Medicine, Baltimore, USA
| | - Alexander L Houck
- University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Paul D P Pharoah
- Department of Oncology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| |
Collapse
|