101
|
Zhang X, Brody JA, Graff M, Highland HM, Chami N, Xu H, Wang Z, Ferrier K, Chittoor G, Josyula NS, Li X, Li Z, Allison MA, Becker DM, Bielak LF, Bis JC, Boorgula MP, Bowden DW, Broome JG, Buth EJ, Carlson CS, Chang KM, Chavan S, Chiu YF, Chuang LM, Conomos MP, DeMeo DL, Du M, Duggirala R, Eng C, Fohner AE, Freedman BI, Garrett ME, Guo X, Haiman C, Heavner BD, Hidalgo B, Hixson JE, Ho YL, Hobbs BD, Hu D, Hui Q, Hwu CM, Jackson RD, Jain D, Kalyani RR, Kardia SL, Kelly TN, Lange EM, LeNoir M, Li C, Marchand LL, McDonald MLN, McHugh CP, Morrison AC, Naseri T, NHLBI Trans-Omics for Precision Medicine (TOPMed) Consortium, O’Connell J, O’Donnell CJ, Palmer ND, Pankow JS, Perry JA, Peters U, Preuss MH, Rao D, Regan EA, Reupena SM, Roden DM, Rodriguez-Santana J, Sitlani CM, Smith JA, Tiwari HK, Vasan RS, Wang Z, Weeks DE, Wessel J, Wiggins KL, Wilkens LR, Wilson PW, Yanek LR, Yoneda ZT, Zhao W, Zöllner S, Arnett DK, Ashley-Koch AE, Barnes KC, Blangero J, Boerwinkle E, Burchard EG, Carson AP, Chasman DI, Chen YDI, Curran JE, Fornage M, Gordeuk VR, He J, Heckbert SR, Hou L, Irvin MR, Kooperberg C, et alZhang X, Brody JA, Graff M, Highland HM, Chami N, Xu H, Wang Z, Ferrier K, Chittoor G, Josyula NS, Li X, Li Z, Allison MA, Becker DM, Bielak LF, Bis JC, Boorgula MP, Bowden DW, Broome JG, Buth EJ, Carlson CS, Chang KM, Chavan S, Chiu YF, Chuang LM, Conomos MP, DeMeo DL, Du M, Duggirala R, Eng C, Fohner AE, Freedman BI, Garrett ME, Guo X, Haiman C, Heavner BD, Hidalgo B, Hixson JE, Ho YL, Hobbs BD, Hu D, Hui Q, Hwu CM, Jackson RD, Jain D, Kalyani RR, Kardia SL, Kelly TN, Lange EM, LeNoir M, Li C, Marchand LL, McDonald MLN, McHugh CP, Morrison AC, Naseri T, NHLBI Trans-Omics for Precision Medicine (TOPMed) Consortium, O’Connell J, O’Donnell CJ, Palmer ND, Pankow JS, Perry JA, Peters U, Preuss MH, Rao D, Regan EA, Reupena SM, Roden DM, Rodriguez-Santana J, Sitlani CM, Smith JA, Tiwari HK, Vasan RS, Wang Z, Weeks DE, Wessel J, Wiggins KL, Wilkens LR, Wilson PW, Yanek LR, Yoneda ZT, Zhao W, Zöllner S, Arnett DK, Ashley-Koch AE, Barnes KC, Blangero J, Boerwinkle E, Burchard EG, Carson AP, Chasman DI, Chen YDI, Curran JE, Fornage M, Gordeuk VR, He J, Heckbert SR, Hou L, Irvin MR, Kooperberg C, Minster RL, Mitchell BD, Nouraie M, Psaty BM, Raffield LM, Reiner AP, Rich SS, Rotter JI, Shoemaker MB, Smith NL, Taylor KD, Telen MJ, Weiss ST, Zhang Y, Heard-Costa N, Sun YV, Lin X, Adrienne Cupples L, Lange LA, Liu CT, Loos RJ, North KE, Justice AE. WHOLE GENOME SEQUENCING ANALYSIS OF BODY MASS INDEX IDENTIFIES NOVEL AFRICAN ANCESTRY-SPECIFIC RISK ALLELE. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.08.21.23293271. [PMID: 37662265 PMCID: PMC10473809 DOI: 10.1101/2023.08.21.23293271] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Obesity is a major public health crisis associated with high mortality rates. Previous genome-wide association studies (GWAS) investigating body mass index (BMI) have largely relied on imputed data from European individuals. This study leveraged whole-genome sequencing (WGS) data from 88,873 participants from the Trans-Omics for Precision Medicine (TOPMed) Program, of which 51% were of non-European population groups. We discovered 18 BMI-associated signals (P < 5 × 10-9). Notably, we identified and replicated a novel low frequency single nucleotide polymorphism (SNP) in MTMR3 that was common in individuals of African descent. Using a diverse study population, we further identified two novel secondary signals in known BMI loci and pinpointed two likely causal variants in the POC5 and DMD loci. Our work demonstrates the benefits of combining WGS and diverse cohorts in expanding current catalog of variants and genes confer risk for obesity, bringing us one step closer to personalized medicine.
Collapse
Affiliation(s)
- Xinruo Zhang
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jennifer A. Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Mariaelisa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Heather M. Highland
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nathalie Chami
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hanfei Xu
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Zhe Wang
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kendra Ferrier
- Division of Biomedical Informatics and Personalized Medicine, School of Medicine University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | | | | | - Xihao Li
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Zilin Li
- Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Matthew A. Allison
- Department of Family Medicine, Division of Preventive Medicine, The University of California San Diego, La Jolla, CA, USA
| | - Diane M. Becker
- Department of Medicine, General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lawrence F. Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Joshua C. Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Donald W. Bowden
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jai G. Broome
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, USA
| | - Erin J. Buth
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Christopher S. Carlson
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Kyong-Mi Chang
- The Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sameer Chavan
- Department of Medicine, School of Medicine, University of Colorado, Aurora, CO, USA
| | - Yen-Feng Chiu
- Institute of Population Health Sciences, National Health Research Institutes, Taipei, Taiwan
| | - Lee-Ming Chuang
- Department of Internal Medicine, Division of Metabolism/Endocrinology, National Taiwan University Hospital, Taipei, Taiwan
| | - Matthew P. Conomos
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Dawn L. DeMeo
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Margaret Du
- Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ravindranath Duggirala
- Life Sciences, College of Arts and Sciences, Texas A&M University-San Antonio, San Antonio, TX, USA
| | - Celeste Eng
- Department of Medicine, Lung Biology Center, University of California, San Francisco, San Francisco, CA, USA
| | - Alison E. Fohner
- Epidemiology, Institute of Public Health Genetics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Barry I. Freedman
- Internal Medicine, Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Melanie E. Garrett
- Department of Medicine, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Xiuqing Guo
- Department of Pediatrics, Genomic Outcomes, 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
| | - Chris Haiman
- Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Benjamin D. Heavner
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Bertha Hidalgo
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA
| | - James E. Hixson
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yuk-Lam Ho
- Veterans Affairs Boston Healthcare System, Boston, MA, USA
| | - Brian D. Hobbs
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Donglei Hu
- Department of Medicine, Lung Biology Center, University of California, San Francisco, San Francisco, CA, USA
| | - Qin Hui
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
- Atlanta VA Health Care System, Decatur, GA, USA
| | - Chii-Min Hwu
- Department of Medicine, Division of Endocrinology and Metabolism, Taipei Veterans General Hospital, Taipei, Taiwan, Taiwan
| | | | - Deepti Jain
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Rita R. Kalyani
- Department of Medicine, Endocrinology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sharon L.R. Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Tanika N. Kelly
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Ethan M. Lange
- Division of Biomedical Informatics and Personalized Medicine, School of Medicine University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Michael LeNoir
- Department of Pediatrics, Bay Area Pediatrics, Oakland, CA, USA
| | - Changwei Li
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Loic Le. Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Merry-Lynn N. McDonald
- Department of Medicine, Pulmonary, Allergy and Critical Care, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Caitlin P. McHugh
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Alanna C. Morrison
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Take Naseri
- Ministry of Health, Government of Samoa, Apia, Samoa
| | | | - Jeffrey O’Connell
- Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland, Baltimore, MD, USA
| | - Christopher J. O’Donnell
- Veterans Affairs Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicholette D. Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - James S. Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - James A. Perry
- Department of Medicine, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michael H. Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - D.C. Rao
- Division of Biostatistics, Washington University in St. Louis, St. Louis, MO, USA
| | - Elizabeth A. Regan
- Department of Medicine, Rheumatology, National Jewish Health, Denver, CO, USA
| | | | - Dan M. Roden
- Medicine, Pharmacology, and Biomedical Informatics, Clinical Pharmacology and Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Colleen M. Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jennifer A. Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Hemant K. Tiwari
- Department of Biostatistics, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA
| | | | - Zeyuan Wang
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Daniel E. Weeks
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jennifer Wessel
- Department of Epidemiology, Indiana University, Indianapolis, IN, USA
- Department of Medicine, Indiana University, Indianapolis, IN, USA
- Diabaetes Translational Research Center, Indiana University, Indianapolis, IN, USA
| | - Kerri L. Wiggins
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Lynne R. Wilkens
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Peter W.F. Wilson
- Atlanta VA Health Care System, Decatur, GA, USA
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Lisa R. Yanek
- Department of Medicine, General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zachary T. Yoneda
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Sebastian Zöllner
- Department of Biostatistics, Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Donna K. Arnett
- Department of Epidemiology, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Allison E. Ashley-Koch
- Department of Medicine, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Kathleen C. Barnes
- Department of Medicine, School of Medicine, University of Colorado, Aurora, CO, USA
| | - John Blangero
- Human Genetics and South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Esteban G. Burchard
- Bioengineering and Therapeutic Sciences and Medicine, Lung Biology Center, University of California, San Francisco, San Francisco, CA, USA
| | - April P. Carson
- Department of Medicine, University of Mississippi, Jackson, MI, USA
| | - Daniel I. Chasman
- Division of Preventive Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yii-Der Ida Chen
- Department of Medical Genetics, Genomic Outcomes, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Joanne E. Curran
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Myriam Fornage
- 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
| | - Victor R. Gordeuk
- Department of Medicine, School of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jiang He
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Susan R. Heckbert
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Lifang Hou
- Northwestern University, Chicago, IL, USA
| | - Marguerite R. Irvin
- Department of Epidemiology, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ryan L. Minster
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Braxton D. Mitchell
- Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland, Baltimore, MD, USA
| | - Mehdi Nouraie
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Laura M. Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Stephen S. Rich
- Public Health Science, Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jerome I. Rotter
- Department of Pediatrics, Genomic Outcomes, 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
| | - M. Benjamin Shoemaker
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nicholas L. Smith
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA, USA
- Seattle Epidemiologic Research and Information Center, Office of Research and Development, Department of Veterans Affairs, Seattle, WA, USA
| | - Kent D. Taylor
- Department of Pediatrics, Genomic Outcomes, 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
| | - Marilyn J. Telen
- Department of Medicine, Hematology, Duke University Medical Center, Durham, NC, USA
| | - Scott T. Weiss
- Department of Medicine, Channing Division of Network Medicine, Harvard Medical School, Boston, MA, USA
| | - Yingze Zhang
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nancy Heard-Costa
- Framingham Heart Study, School of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Yan V. Sun
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
- Atlanta VA Health Care System, Decatur, GA, USA
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Statistics, Harvard University, Boston, MA, USA
| | - L. Adrienne Cupples
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Leslie A. Lange
- Division of Biomedical Informatics and Personalized Medicine, School of Medicine University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Ching-Ti Liu
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Ruth J.F. Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Kari E. North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | |
Collapse
|
102
|
Zanussi JT, Zhao J, Wei WQ, Karakoc G, Chung CP, Feng Q, Olsen NJ, Stein CM, Kawai VK. Clinical diagnoses associated with a positive antinuclear antibody test in patients with and without autoimmune disease. BMC Rheumatol 2023; 7:24. [PMID: 37550754 PMCID: PMC10405518 DOI: 10.1186/s41927-023-00349-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 07/27/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND Antinuclear antibodies (ANA) are antibodies present in several autoimmune disorders. However, a large proportion of the general population (20%) also have a positive test; very few of these individuals will develop an autoimmune disease, and the clinical impact of a positive ANA in them is not known. Thus, we test the hypothesis that ANA + test reflects a state of immune dysregulation that alters risk for some clinical disorders in individuals without an autoimmune disease. METHODS We performed high throughput association analyses in a case-control study using real world data from the de-identified electronic health record (EHR) system from Vanderbilt University Medical Center. The study population included individuals with an ANA titer ≥ 1:80 at any time (ANA +) and those with negative results (ANA-). The cohort was stratified into sub-cohorts of individuals with and without an autoimmune disease. A phenome-wide association study (PheWAS) adjusted by sex, year of birth, race, and length of follow-up was performed in the study cohort and in the sub-cohorts. As secondary analyses, only clinical diagnoses after ANA testing were included in the analyses. RESULTS The cohort included 70,043 individuals: 49,546 without and 20,497 with an autoimmune disease, 26,579 were ANA + and 43,464 ANA-. In the study cohort and the sub-cohort with autoimmune disease, ANA + was associated (P ≤ 5 × 10-5) with 88 and 136 clinical diagnoses respectively, including lupus (OR ≥ 5.4, P ≤ 7.8 × 10-202) and other autoimmune diseases and complications. In the sub-cohort without autoimmune diseases, ANA + was associated with increased risk of Raynaud's syndrome (OR ≥ 2.1) and alveolar/perialveolar-related pneumopathies (OR ≥ 1.4) and decreased risk of hepatitis C, tobacco use disorders, mood disorders, convulsions, fever of unknown origin, and substance abuse disorders (OR ≤ 0.8). Analyses including only diagnoses after ANA testing yielded similar results. CONCLUSION A positive ANA test, in addition to known associations with autoimmune diseases, Raynaud's phenomenon, and idiopathic fibrosing alveolitis related disorders, is associated with decreased prevalence of several non-autoimmune diseases.
Collapse
Affiliation(s)
- Jacy T Zanussi
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Juan Zhao
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Gul Karakoc
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cecilia P Chung
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
- Division of Rheumatology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Tennessee Valley Healthcare System - Nashville Campus, Nashville, TN, USA
| | - QiPing Feng
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Nancy J Olsen
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| | - C Michael Stein
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Vivian K Kawai
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
103
|
Zhang S, Strayer N, Vessels T, Choi K, Wang GW, Li Y, Bejan CA, Hsi RS, Bick AG, Velez Edwards DR, Savona MR, Philips EJ, Pulley J, Self WH, Hopkins WC, Roden DM, Smoller JW, Ruderfer DM, Xu Y. PheMIME: An Interactive Web App and Knowledge Base for Phenome-Wide, Multi-Institutional Multimorbidity Analysis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.07.23.23293047. [PMID: 37547012 PMCID: PMC10402210 DOI: 10.1101/2023.07.23.23293047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Motivation Multimorbidity, characterized by the simultaneous occurrence of multiple diseases in an individual, is an increasing global health concern, posing substantial challenges to healthcare systems. Comprehensive understanding of disease-disease interactions and intrinsic mechanisms behind multimorbidity can offer opportunities for innovative prevention strategies, targeted interventions, and personalized treatments. Yet, there exist limited tools and datasets that characterize multimorbidity patterns across different populations. To bridge this gap, we used large-scale electronic health record (EHR) systems to develop the Phenome-wide Multi-Institutional Multimorbidity Explorer (PheMIME), which facilitates research in exploring and comparing multimorbidity patterns among multiple institutions, potentially leading to the discovery of novel and robust disease associations and patterns that are interoperable across different systems and organizations. Results PheMIME integrates summary statistics from phenome-wide analyses of disease multimorbidities. These are currently derived from three major institutions: Vanderbilt University Medical Center, Mass General Brigham, and the UK Biobank. PheMIME offers interactive exploration of multimorbidity through multi-faceted visualization. Incorporating an enhanced version of associationSubgraphs, PheMIME enables dynamic analysis and inference of disease clusters, promoting the discovery of multimorbidity patterns. Once a disease of interest is selected, the tool generates interactive visualizations and tables that users can delve into multimorbidities or multimorbidity networks within a single system or compare across multiple systems. The utility of PheMIME is demonstrated through a case study on schizophrenia. Availability and implementation The PheMIME knowledge base and web application are accessible at https://prod.tbilab.org/PheMIME/. A comprehensive tutorial, including a use-case example, is available at https://prod.tbilab.org/PheMIME_supplementary_materials/. Furthermore, the source code for PheMIME can be freely downloaded from https://github.com/tbilab/PheMIME. Data availability statement The data underlying this article are available in the article and in its online web application or supplementary material.
Collapse
Affiliation(s)
- Siwei Zhang
- Department of Biostatistics, Vanderbilt University, Nashville, TN, USA
| | | | - Tess Vessels
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Karmel Choi
- Psychiatric & Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston MA
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston MA
| | | | - Yajing Li
- Department of Biostatistics, Vanderbilt University, Nashville, TN, USA
| | - Cosmin A Bejan
- Department of Biomedical informatics, Vanderbilt University, Nashville, TN, USA
| | - Ryan S Hsi
- Department of Urology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexander G Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Digna R Velez Edwards
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael R Savona
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Elizabeth J Philips
- Center for Drug Safety and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, Australia
| | - Jill Pulley
- Vanderbilt Institute for Clinical and Translational Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wesley H Self
- Vanderbilt Institute for Clinical and Translational Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wilkins Consuelo Hopkins
- Vanderbilt Institute for Clinical and Translational Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dan M Roden
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jordan W Smoller
- Psychiatric & Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston MA
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston MA
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA
| | - Douglas M Ruderfer
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical informatics, Vanderbilt University, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yaomin Xu
- Department of Biostatistics, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical informatics, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
104
|
Allaire P, He J, Mayer J, Moat L, Gerstenberger P, Wilhorn R, Strutz S, Kim DS, Zeng C, Cox N, Shay JW, Denny J, Bastarache L, Hebbring S. Genetic and clinical determinants of telomere length. HGG ADVANCES 2023; 4:100201. [PMID: 37216007 PMCID: PMC10199259 DOI: 10.1016/j.xhgg.2023.100201] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Many epidemiologic studies have identified important relationships between leukocyte telomere length (LTL) with genetics and health. Most of these studies have been significantly limited in scope by focusing predominantly on individual diseases or restricted to GWAS analysis. Using two large patient populations derived from Vanderbilt University and Marshfield Clinic biobanks linked to genomic and phenomic data from medical records, we investigated the inter-relationship between LTL, genomics, and human health. Our GWAS confirmed 11 genetic loci previously associated with LTL and two novel loci in SCNN1D and PITPNM1. PheWAS of LTL identified 67 distinct clinical phenotypes associated with both short and long LTL. We demonstrated that several diseases associated with LTL were related to one another but were largely independent from LTL genetics. Age of death was correlated with LTL independent of age. Those with very short LTL (<-1.5 standard deviation [SD]) died 10.4 years (p < 0.0001) younger than those with average LTL (±0.5 SD; mean age of death = 74.2 years). Likewise, those with very long LTL (>1.5 SD) died 1.9 years (p = 0.0175) younger than those with average LTL. This is consistent with the PheWAS results showing diseases associating with both short and long LTL. Finally, we estimated that the genome (12.8%) and age (8.5%) explain the largest proportion of LTL variance, whereas the phenome (1.5%) and sex (0.9%) explained a smaller fraction. In total, 23.7% of LTL variance was explained. These observations provide the rationale for expanded research to understand the multifaceted correlations between TL biology and human health over time, leading to effective LTL usage in medical applications.
Collapse
Affiliation(s)
- Patrick Allaire
- Marshfield Clinic Research Institute, Center for Precision Medicine Research, Marshfield, WI, USA
| | - Jing He
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John Mayer
- Marshfield Clinic Research Institute, Office of Research Computing and Analytics, Marshfield, WI, USA
| | - Luke Moat
- Marshfield Clinic Research Institute, Center for Precision Medicine Research, Marshfield, WI, USA
| | - Peter Gerstenberger
- Marshfield Clinic Research Institute, Center for Precision Medicine Research, Marshfield, WI, USA
| | - Reynor Wilhorn
- Marshfield Clinic Research Institute, Center for Precision Medicine Research, Marshfield, WI, USA
| | - Sierra Strutz
- Marshfield Clinic Research Institute, Center for Precision Medicine Research, Marshfield, WI, USA
| | - David S.L. Kim
- Marshfield Clinic Health System, Pathology, Marshfield, WI, USA
| | - Chenjie Zeng
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nancy Cox
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jerry W. Shay
- University of Texas Southwestern Medical Center, Department of Cell Biology and the Simmons Comprehensive Cancer Center, Dallas, TX, USA
| | - Joshua Denny
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lisa Bastarache
- Center for Precision Medicine, Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Scott Hebbring
- Marshfield Clinic Research Institute, Center for Precision Medicine Research, Marshfield, WI, USA
| |
Collapse
|
105
|
Niarchou M, Miller-Fleming T, Malow BA, Davis LK. The physical and psychiatric health conditions related to autism genetic scores, across genetic ancestries, sexes and age-groups in electronic health records. J Neurodev Disord 2023; 15:18. [PMID: 37328826 PMCID: PMC10273739 DOI: 10.1186/s11689-023-09485-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 05/24/2023] [Indexed: 06/18/2023] Open
Abstract
BACKGROUND Although polygenic scores (PGS) for autism have been related to various psychiatric and medical conditions, most studies to date have been conducted in research ascertained populations. We aimed to identify the psychiatric and physical conditions associated with autism PGS in a health care setting. METHODS We computed PGS for 12,383 unrelated participants of African genetic ancestry (AF) and 65,363 unrelated participants of European genetic ancestry (EU) from Vanderbilt's de-identified biobank. Next, we performed phenome wide association studies of the autism PGS within these two genetic ancestries. RESULTS Seven associations surpassed the Bonferroni adjusted threshold for statistical significance (p = 0.05/1374 = 3.6 × 10-5) in the EU participants, including mood disorders (OR (95%CI) = 1.08(1.05 to 1.10), p = 1.0 × 10-10), autism (OR (95%CI) = 1.34(1.24 to 1.43), p = 1.2 × 10-9), and breast cancer (OR (95%CI) = 1.09(1.05 to 1.14), 2.6 × 10-5). There was no statistical evidence for PGS-phenotype associations in the AF participants. Conditioning on the diagnosis of autism or on median body mass index (BMI) did not impact the strength of the reported associations. Although we observed some sex differences in the pattern of associations, there was no significant interaction between sex and autism PGS. Finally, the associations between autism PGS and autism diagnosis were stronger in childhood and adolescence, while the associations with mood disorders and breast cancer were stronger in adulthood. DISCUSSION Our findings indicate that autism PGS is not only related to autism diagnosis but may also be related to adult-onset conditions, including mood disorders and some cancers. CONCLUSIONS Our study raises the hypothesis that genes associated with autism may also increase the risk for cancers later in life. Future studies are necessary to replicate and extend our findings.
Collapse
Affiliation(s)
- Maria Niarchou
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Tyne Miller-Fleming
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Beth A Malow
- Sleep Disorders Division, Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lea K Davis
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Division of Neurology, Pharmacology and Special Education, Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
106
|
Vessels T, Strayer N, Choi KW, Lee H, Zhang S, Han L, Morley TJ, Smoller JW, Xu Y, Ruderfer DM. Identifying modifiable comorbidities of schizophrenia by integrating electronic health records and polygenic risk. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.01.23290057. [PMID: 37333378 PMCID: PMC10274978 DOI: 10.1101/2023.06.01.23290057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Patients with schizophrenia have substantial comorbidity contributing to reduced life expectancy of 10-20 years. Identifying which comorbidities might be modifiable could improve rates of premature mortality in this population. We hypothesize that conditions that frequently co-occur but lack shared genetic risk with schizophrenia are more likely to be products of treatment, behavior, or environmental factors and therefore potentially modifiable. To test this hypothesis, we calculated phenome-wide comorbidity from electronic health records (EHR) in 250,000 patients in each of two independent health care institutions (Vanderbilt University Medical Center and Mass General Brigham) and association with schizophrenia polygenic risk scores (PRS) across the same phenotypes (phecodes) in linked biobanks. Comorbidity with schizophrenia was significantly correlated across institutions (r = 0.85) and consistent with prior literature. After multiple test correction, there were 77 significant phecodes comorbid with schizophrenia. Overall, comorbidity and PRS association were highly correlated (r = 0.55, p = 1.29×10-118), however, 36 of the EHR identified comorbidities had significantly equivalent schizophrenia PRS distributions between cases and controls. Fifteen of these lacked any PRS association and were enriched for phenotypes known to be side effects of antipsychotic medications (e.g., "movement disorders", "convulsions", "tachycardia") or other schizophrenia related factors such as from smoking ("bronchitis") or reduced hygiene (e.g., "diseases of the nail") highlighting the validity of this approach. Other phenotypes implicated by this approach where the contribution from shared common genetic risk with schizophrenia was minimal included tobacco use disorder, diabetes, and dementia. This work demonstrates the consistency and robustness of EHR-based schizophrenia comorbidities across independent institutions and with the existing literature. It identifies comorbidities with an absence of shared genetic risk indicating other causes that might be more modifiable and where further study of causal pathways could improve outcomes for patients.
Collapse
Affiliation(s)
- Tess Vessels
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville TN
| | - Nicholas Strayer
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville TN
| | - Karmel W. Choi
- Psychiatric & Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston MA
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston MA
| | - Hyunjoon Lee
- Psychiatric & Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston MA
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston MA
| | - Siwei Zhang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville TN
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville TN
| | - Lide Han
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville TN
| | - Theodore J. Morley
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville TN
| | - Jordan W. Smoller
- Psychiatric & Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston MA
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston MA
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA
| | - Yaomin Xu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville TN
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville TN
| | - Douglas M. Ruderfer
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville TN
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville TN
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
107
|
Shoaib M, Ye Q, IglayReger H, Tan MH, Boehnke M, Burant CF, Soleimanpour SA, Gagliano Taliun SA. Evaluation of polygenic risk scores to differentiate between type 1 and type 2 diabetes. Genet Epidemiol 2023; 47:303-313. [PMID: 36821788 PMCID: PMC10202843 DOI: 10.1002/gepi.22521] [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: 12/13/2022] [Revised: 02/11/2023] [Accepted: 02/11/2023] [Indexed: 02/25/2023]
Abstract
Polygenic risk scores (PRS) quantify the genetic liability to disease and are calculated using an individual's genotype profile and disease-specific genome-wide association study (GWAS) summary statistics. Type 1 (T1D) and type 2 (T2D) diabetes both are determined in part by genetic loci. Correctly differentiating between types of diabetes is crucial for accurate diagnosis and treatment. PRS have the potential to address possible misclassification of T1D and T2D. Here we evaluated PRS models for T1D and T2D in European genetic ancestry participants from the UK Biobank (UKB) and then in the Michigan Genomics Initiative (MGI). Specifically, we investigated the utility of T1D and T2D PRS to discriminate between T1D, T2D, and controls in unrelated UKB individuals of European ancestry. We derived PRS models using external non-UKB GWAS. The T1D PRS model with the best discrimination between T1D cases and controls (area under the receiver operator curve [AUC] = 0.805) also yielded the best discrimination of T1D from T2D cases in the UKB (AUC = 0.792) and separation in MGI (AUC = 0.686). In contrast, the best T2D model did not discriminate between T1D and T2D cases (AUC = 0.527). Our analysis suggests that a T1D PRS model based on independent single nucleotide polymorphisms may help differentiate between T1D, T2D, and controls in individuals of European genetic ancestry.
Collapse
Affiliation(s)
- Muhammad Shoaib
- Montreal Heart Institute Research Centre, Montréal, Québec, Canada
- Université de Montréal, Université de Montréal, Montréal, Québec, Canada
| | - Qiang Ye
- Montreal Heart Institute Research Centre, Montréal, Québec, Canada
- Université de Montréal, Université de Montréal, Montréal, Québec, Canada
| | - Heidi IglayReger
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Meng H. Tan
- Division of Metabolism, Endocrinology & Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Charles F. Burant
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Scott A. Soleimanpour
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sarah A. Gagliano Taliun
- Montreal Heart Institute Research Centre, Montréal, Québec, Canada
- Department of Medicine and Department of Neurosciences, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
108
|
Seeling KS, Hehl L, Vell MS, Rendel MD, Creasy KT, Trautwein C, Mehler DMA, Keszthelyi D, Schneider KM, Schneider CV. Comorbidities, biomarkers and cause specific mortality in patients with irritable bowel syndrome: A phenome-wide association study. United European Gastroenterol J 2023; 11:458-470. [PMID: 37151116 PMCID: PMC10256994 DOI: 10.1002/ueg2.12397] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 04/02/2023] [Indexed: 05/09/2023] Open
Abstract
BACKGROUND Irritable bowel syndrome (IBS) is one of the most common functional digestive disorders. Our understanding about its comorbidities, biomarkers, or long-term risks is still incomplete. OBJECTIVE To characterize comorbidities and biomarkers for IBS and establish the effect of IBS on overall- and cause specific mortality. METHODS We analyzed data from the population-based cohort of the UK Biobank (UKB) with 493,974 participants, including self-reported physician-diagnosed (n = 20,603) and ICD-10 diagnosed (n = 7656) IBS patients, with a mean follow-up of 11 years. We performed a phenome-wide association study (PheWAS) and competing risk analysis to characterize common clinical features in IBS patients. RESULTS In PheWAS analyses, 260 PheCodes were significantly overrepresented in self-reported physician-diagnosed IBS patients, 633 in patients with ICD-10 diagnosed IBS (ICD-10-IBS), with 221 (40%) overlapping. In addition to gastrointestinal diseases, psychiatric, musculoskeletal, and endocrine/metabolic disorders represented the most strongly associated PheCodes in IBS patients. Self-reported physician-diagnosed IBS was not associated with increased overall mortality and the risk of death from cancer was decreased (hazard ratio [HR] = 0.78 [95% CI = 0.7-0.9]). Lastly, we evaluated changes in serum metabolites in IBS patients and identified glycoprotein acetyls (GlycA) as a potential biomarker in IBS. One standard deviation increase in GlycA raised the risk of self-reported IBS/ICD-10 coded by 9%-20% (odds ratio [OR] = 1.09 [95% CI = 1.1-1.1]/OR = 1.20 [95% CI = 1.1-1.3]) and the risk of overall mortality in ICD-10-IBS patients by 28% (HR = 1.28 [95% CI = 1.1-1.5]). CONCLUSION Our large-scale association study determined IBS patients having an increased risk of several different comorbidities and that GlycA was increased in IBS patients.
Collapse
Affiliation(s)
- Katharina Sophie Seeling
- Medical Clinic IIIGastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
| | - Leonida Hehl
- Medical Clinic IIIGastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
| | - Mara Sophie Vell
- Medical Clinic IIIGastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
| | - Miriam Daphne Rendel
- Medical Clinic IIIGastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
| | - Kate Townsend Creasy
- Department of Biobehavioral Health SciencesPerelman School of NursingUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Christian Trautwein
- Medical Clinic IIIGastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
| | - David Marc Anton Mehler
- Department of Psychiatry, Psychotherapy and PsychosomaticsUniversity Hospital RWTH AachenAachenGermany
- Institute for Translational PsychiatryUniversity of MünsterMünsterGermany
| | - Daniel Keszthelyi
- Division of Gastroenterology‐HepatologyDepartment of Internal MedicineMaastricht University Medical Center+Maastrichtthe Netherlands
| | - Kai Markus Schneider
- Medical Clinic IIIGastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
| | - Carolin Victoria Schneider
- Medical Clinic IIIGastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
- The Institute for Translational Medicine and TherapeuticsThe Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
109
|
Honigberg MC, Truong B, Khan RR, Xiao B, Bhatta L, Vy HMT, Guerrero RF, Schuermans A, Selvaraj MS, Patel AP, Koyama S, Cho SMJ, Vellarikkal SK, Trinder M, Urbut SM, Gray KJ, Brumpton BM, Patil S, Zöllner S, Antopia MC, Saxena R, Nadkarni GN, Do R, Yan Q, Pe'er I, Verma SS, Gupta RM, Haas DM, Martin HC, van Heel DA, Laisk T, Natarajan P. Polygenic prediction of preeclampsia and gestational hypertension. Nat Med 2023; 29:1540-1549. [PMID: 37248299 PMCID: PMC10330886 DOI: 10.1038/s41591-023-02374-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 04/26/2023] [Indexed: 05/31/2023]
Abstract
Preeclampsia and gestational hypertension are common pregnancy complications associated with adverse maternal and child outcomes. Current tools for prediction, prevention and treatment are limited. Here we tested the association of maternal DNA sequence variants with preeclampsia in 20,064 cases and 703,117 control individuals and with gestational hypertension in 11,027 cases and 412,788 control individuals across discovery and follow-up cohorts using multi-ancestry meta-analysis. Altogether, we identified 18 independent loci associated with preeclampsia/eclampsia and/or gestational hypertension, 12 of which are new (for example, MTHFR-CLCN6, WNT3A, NPR3, PGR and RGL3), including two loci (PLCE1 and FURIN) identified in the multitrait analysis. Identified loci highlight the role of natriuretic peptide signaling, angiogenesis, renal glomerular function, trophoblast development and immune dysregulation. We derived genome-wide polygenic risk scores that predicted preeclampsia/eclampsia and gestational hypertension in external cohorts, independent of clinical risk factors, and reclassified eligibility for low-dose aspirin to prevent preeclampsia. Collectively, these findings provide mechanistic insights into the hypertensive disorders of pregnancy and have the potential to advance pregnancy risk stratification.
Collapse
Affiliation(s)
- Michael C Honigberg
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| | - Buu Truong
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Raiyan R Khan
- Department of Computer Science, Columbia University, New York, NY, USA
| | - Brenda Xiao
- University of Pennsylvania, Philadelphia, PA, USA
| | - Laxmi Bhatta
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Levanger, Norway
| | - Ha My T Vy
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rafael F Guerrero
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Art Schuermans
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Margaret Sunitha Selvaraj
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Aniruddh P Patel
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Satoshi Koyama
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - So Mi Jemma Cho
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Integrative Research Center for Cerebrovascular and Cardiovascular Diseases, Seoul, Republic of Korea
| | - Shamsudheen Karuthedath Vellarikkal
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Mark Trinder
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sarah M Urbut
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Kathryn J Gray
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Ben M Brumpton
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Levanger, Norway
| | - Snehal Patil
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Sebastian Zöllner
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Mariah C Antopia
- Department of Integrative Biology, University of Texas at San Antonio, San Antonio, TX, USA
| | - Richa Saxena
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Girish N Nadkarni
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ron Do
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qi Yan
- Department of Obstetrics and Gynecology, Columbia University, New York, NY, USA
| | - Itsik Pe'er
- Department of Computer Science, Columbia University, New York, NY, USA
| | | | - Rajat M Gupta
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - David M Haas
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hilary C Martin
- Department of Human Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - David A van Heel
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Triin Laisk
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Pradeep Natarajan
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
110
|
Chung CP, Karakoc G, Dickson A, Liu G, Gamboa JL, Mosley JD, Cox NJ, Kawai VK. APOL1 and the risk of adverse renal outcomes in patients of African ancestry with systemic lupus erythematosus. Lupus 2023; 32:763-770. [PMID: 37105192 PMCID: PMC10189827 DOI: 10.1177/09612033231172660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) disproportionately affects individuals of African ancestry (AA) compared to European ancestry (EA). In the general population, high risk (HR) variants in the apolipoprotein L1 (APOL1) gene increase the risk of renal and hypertensive disorders in individuals of AA. Since SLE is characterized by an interferon signature and APOL1 expression is driven by interferon, we examined the hypothesis that APOL1 HR genotypes predominantly drive higher rates of renal and hypertensive-related comorbidities observed in SLE patients of AA versus those of EA. METHODS We performed a retrospective cohort study in patients with SLE of EA and AA using a genetic biobank linked to de-identified electronic health records. APOL1 HR genotypes were defined as G1/G1, G2/G2, or G1/G2 and low risk (LR) genotypes as 1 or 0 copies of the G1 and G2 alleles. To identify renal and hypertensive-related disorders that differed in prevalence by ancestry, we used a phenome-wide association approach. We then used logistic regression to compare the prevalence of renal and hypertensive-related disorders in EA and AA patients, both including and excluding patients with the APOL1 HR genotype. In a sensitivity analysis, we examined the association of end stage renal disease secondary to lupus nephritis (LN-related ESRD) with ancestry and the APOL1 genotype. RESULTS We studied 784 patients with SLE; 195 (24.9%) were of AA, of whom 27 (13.8%) had APOL1 HR genotypes. Eighteen renal and hypertensive-related phenotypes were more common in AA than EA patients (p-value ≤ 1.4E-4). All phenotypes remained significantly different after exclusion of patients with APOL1 HR genotypes, and most point odds ratios (ORs) decreased only slightly. Even among ORs with the greatest decrease, risk for AA patients without the APOL1 HR genotype remained significantly elevated compared to EA patients. In the sensitivity analysis, LN-related ESRD was more prevalent in SLE patients of AA versus EA and AA patients with the APOL1 HR genotype versus LR (p-value < .05 for both). CONCLUSION The higher prevalence of renal and hypertensive disorders in SLE patients of AA compared to those of EA is not fully explained by the presence of APOL1 high risk variants.
Collapse
Affiliation(s)
- Cecilia P Chung
- Division of Rheumatology and
Immunology, Department of Medicine Vanderbilt
University Medical Center, Nashville, TN, USA
- Tennessee Valley Healthcare System -
Nashville Campus, Nashville, TN, USA
- Division of Clinical Pharmacology,
Department of Medicine, Vanderbilt University Medical
Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical
Center, Nashville, TN, USA
- Division of Rheumatology, University of Miami, Miami, FL, USA
- Miami VA Healthcare
System, Miami, FL, USA
| | - Gul Karakoc
- Division of Clinical Pharmacology,
Department of Medicine, Vanderbilt University Medical
Center, Nashville, TN, USA
| | - Alyson Dickson
- Division of Rheumatology and
Immunology, Department of Medicine Vanderbilt
University Medical Center, Nashville, TN, USA
| | - Ge Liu
- Division of Clinical Pharmacology,
Department of Medicine, Vanderbilt University Medical
Center, Nashville, TN, USA
| | - Jorge L Gamboa
- Division of Clinical Pharmacology,
Department of Medicine, Vanderbilt University Medical
Center, Nashville, TN, USA
| | - Jonathan D Mosley
- Division of Clinical Pharmacology,
Department of Medicine, Vanderbilt University Medical
Center, Nashville, TN, USA
- Department of Biomedical
Informatics, Vanderbilt University School of
Medicine, Nashville, TN, USA
| | - Nancy J Cox
- Vanderbilt Genetics Institute, Vanderbilt University Medical
Center, Nashville, TN, USA
| | - Vivian K Kawai
- Division of Clinical Pharmacology,
Department of Medicine, Vanderbilt University Medical
Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical
Center, Nashville, TN, USA
| |
Collapse
|
111
|
Nagar SD, Jordan IK, Mariño-Ramírez L. The landscape of health disparities in the UK Biobank. Database (Oxford) 2023; 2023:7143539. [PMID: 37114803 PMCID: PMC10132819 DOI: 10.1093/database/baad026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 03/01/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023]
Abstract
The UK Biobank (UKB), a large-scale biomedical database that includes demographic and electronic health record data for more than half a million ethnically diverse participants, is a potentially valuable resource for the study of health disparities. However, publicly accessible databases that catalog health disparities in the UKB do not exist. We developed the UKB Health Disparities Browser with the aims of (i) facilitating the exploration of the landscape of health disparities in the UK and (ii) directing the attention to areas of disparities research that might have the greatest public health impact. Health disparities were characterized for UKB participant groups defined by age, country of residence, ethnic group, sex and socioeconomic deprivation. We defined disease cohorts for UKB participants by mapping participant International Classification of Diseases, Tenth Revision (ICD-10) diagnosis codes to phenotype codes (phecodes). For each of the population attributes used to define population groups, disease percent prevalence values were computed for all groups from phecode case-control cohorts, and the magnitude of the disparities was calculated by both the difference and ratio of the range of disease prevalence values among groups to identify high- and low-prevalence disparities. We identified numerous diseases and health conditions with disparate prevalence values across population attributes, and we deployed an interactive web browser to visualize the results of our analysis: https://ukbatlas.health-disparities.org. The interactive browser includes overall and group-specific prevalence data for 1513 diseases based on a cohort of >500 000 participants from the UKB. Researchers can browse and sort by disease prevalence and prevalence differences to visualize health disparities for each of the five population attributes, and users can search for diseases of interest by disease names or codes. Database URL https://ukbatlas.health-disparities.org/.
Collapse
Affiliation(s)
| | - I King Jordan
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332 USA
- IHRC-Georgia Tech Applied Bioinformatics Laboratory, Atlanta, GA 30332 USA
| | - Leonardo Mariño-Ramírez
- National Institute on Minority Health and Health Disparities, National Institutes of Health, Rockville, MD 20818 USA
| |
Collapse
|
112
|
Hicks EM, Seah C, Cote A, Marchese S, Brennand KJ, Nestler EJ, Girgenti MJ, Huckins LM. Integrating genetics and transcriptomics to study major depressive disorder: a conceptual framework, bioinformatic approaches, and recent findings. Transl Psychiatry 2023; 13:129. [PMID: 37076454 PMCID: PMC10115809 DOI: 10.1038/s41398-023-02412-7] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 03/17/2023] [Accepted: 03/24/2023] [Indexed: 04/21/2023] Open
Abstract
Major depressive disorder (MDD) is a complex and heterogeneous psychiatric syndrome with genetic and environmental influences. In addition to neuroanatomical and circuit-level disturbances, dysregulation of the brain transcriptome is a key phenotypic signature of MDD. Postmortem brain gene expression data are uniquely valuable resources for identifying this signature and key genomic drivers in human depression; however, the scarcity of brain tissue limits our capacity to observe the dynamic transcriptional landscape of MDD. It is therefore crucial to explore and integrate depression and stress transcriptomic data from numerous, complementary perspectives to construct a richer understanding of the pathophysiology of depression. In this review, we discuss multiple approaches for exploring the brain transcriptome reflecting dynamic stages of MDD: predisposition, onset, and illness. We next highlight bioinformatic approaches for hypothesis-free, genome-wide analyses of genomic and transcriptomic data and their integration. Last, we summarize the findings of recent genetic and transcriptomic studies within this conceptual framework.
Collapse
Affiliation(s)
- Emily M Hicks
- Pamela Sklar Division of Psychiatric Genomics, Departments of Psychiatry and of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Carina Seah
- Pamela Sklar Division of Psychiatric Genomics, Departments of Psychiatry and of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Alanna Cote
- Pamela Sklar Division of Psychiatric Genomics, Departments of Psychiatry and of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Shelby Marchese
- Pamela Sklar Division of Psychiatric Genomics, Departments of Psychiatry and of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Kristen J Brennand
- Pamela Sklar Division of Psychiatric Genomics, Departments of Psychiatry and of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, 06511, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Matthew J Girgenti
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06511, USA.
| | - Laura M Huckins
- Pamela Sklar Division of Psychiatric Genomics, Departments of Psychiatry and of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA.
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06511, USA.
| |
Collapse
|
113
|
Johnson JS, Cote AC, Dobbyn A, Sloofman LG, Xu J, Cotter L, Charney AW, Eating Disorders Working Group of the Psychiatric Genomics Consortium, Birgegård A, Jordan J, Kennedy M, Landén M, Maguire SL, Martin NG, Mortensen PB, Thornton LM, Bulik CM, Huckins LM. Mapping anorexia nervosa genes to clinical phenotypes. Psychol Med 2023; 53:2619-2633. [PMID: 35379376 PMCID: PMC10123844 DOI: 10.1017/s0033291721004554] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/23/2021] [Accepted: 10/20/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Anorexia nervosa (AN) is a psychiatric disorder with complex etiology, with a significant portion of disease risk imparted by genetics. Traditional genome-wide association studies (GWAS) produce principal evidence for the association of genetic variants with disease. Transcriptomic imputation (TI) allows for the translation of those variants into regulatory mechanisms, which can then be used to assess the functional outcome of genetically regulated gene expression (GReX) in a broader setting through the use of phenome-wide association studies (pheWASs) in large and diverse clinical biobank populations with electronic health record phenotypes. METHODS Here, we applied TI using S-PrediXcan to translate the most recent PGC-ED AN GWAS findings into AN-GReX. For significant genes, we imputed AN-GReX in the Mount Sinai BioMe™ Biobank and performed pheWASs on over 2000 outcomes to test the clinical consequences of aberrant expression of these genes. We performed a secondary analysis to assess the impact of body mass index (BMI) and sex on AN-GReX clinical associations. RESULTS Our S-PrediXcan analysis identified 53 genes associated with AN, including what is, to our knowledge, the first-genetic association of AN with the major histocompatibility complex. AN-GReX was associated with autoimmune, metabolic, and gastrointestinal diagnoses in our biobank cohort, as well as measures of cholesterol, medications, substance use, and pain. Additionally, our analyses showed moderation of AN-GReX associations with measures of cholesterol and substance use by BMI, and moderation of AN-GReX associations with celiac disease by sex. CONCLUSIONS Our BMI-stratified results provide potential avenues of functional mechanism for AN-genes to investigate further.
Collapse
Affiliation(s)
- Jessica S. Johnson
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alanna C. Cote
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Amanda Dobbyn
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Laura G. Sloofman
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jiayi Xu
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Liam Cotter
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alexander W. Charney
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- James J. Peters Department of Veterans Affairs Medical Center, Mental Illness Research, Education and Clinical Centers, Bronx, NY 14068, USA
| | | | - Andreas Birgegård
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jennifer Jordan
- Department of Psychological Medicine, Christchurch School of Medicine & Health Sciences, University of Otago, 2 Riccarton Avenue, PO Box 4345, 8140 Christchurch, New Zealand
| | - Martin Kennedy
- Department of Psychological Medicine, Christchurch School of Medicine & Health Sciences, University of Otago, 2 Riccarton Avenue, PO Box 4345, 8140 Christchurch, New Zealand
| | - Mikaél Landén
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Institute of Neuroscience and Physiology, Sahlgrenska Academy at Gothenburg University, SE-413 45 Gothenburg, Sweden
| | - Sarah L. Maguire
- InsideOut Institute, University of Sydney, New South Wales 2006, Australia
| | - Nicholas G. Martin
- QIMR Berghofer Medical Research Institute, Locked Bag 2000, Royal Brisbane Hospital, Herston, QLD 4029, Australia
| | - Preben Bo Mortensen
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
| | - Laura M. Thornton
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27517, USA
| | - Cynthia M. Bulik
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27517, USA
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27517, USA
| | - Laura M. Huckins
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- James J. Peters Department of Veterans Affairs Medical Center, Mental Illness Research, Education and Clinical Centers, Bronx, NY 14068, USA
| |
Collapse
|
114
|
Wang Q, Qi H, Wu Y, Yu L, Bouchareb R, Li S, Lassén E, Casalena G, Stadler K, Ebefors K, Yi Z, Shi S, Salem F, Gordon R, Lu L, Williams RW, Duffield J, Zhang W, Itan Y, Böttinger E, Daehn I. Genetic susceptibility to diabetic kidney disease is linked to promoter variants of XOR. Nat Metab 2023; 5:607-625. [PMID: 37024752 PMCID: PMC10821741 DOI: 10.1038/s42255-023-00776-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/07/2023] [Indexed: 04/08/2023]
Abstract
The lifetime risk of kidney disease in people with diabetes is 10-30%, implicating genetic predisposition in the cause of diabetic kidney disease (DKD). Here we identify an expression quantitative trait loci (QTLs) in the cis-acting regulatory region of the xanthine dehydrogenase, or xanthine oxidoreductase (Xor), a binding site for C/EBPβ, to be associated with diabetes-induced podocyte loss in DKD in male mice. We examine mouse inbred strains that are susceptible (DBA/2J) and resistant (C57BL/6J) to DKD, as well as a panel of recombinant inbred BXD mice, to map QTLs. We also uncover promoter XOR orthologue variants in humans associated with high risk of DKD. We introduced the risk variant into the 5'-regulatory region of XOR in DKD-resistant mice, which resulted in increased Xor activity associated with podocyte depletion, albuminuria, oxidative stress and damage restricted to the glomerular endothelium, which increase further with type 1 diabetes, high-fat diet and ageing. Therefore, differential regulation of Xor contributes to phenotypic consequences with diabetes and ageing.
Collapse
Affiliation(s)
- Qin Wang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pharmacy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Haiying Qi
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yiming Wu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Liping Yu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rihab Bouchareb
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shuyu Li
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emelie Lassén
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gabriella Casalena
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Krisztian Stadler
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Kerstin Ebefors
- Department of Neuroscience and Physiology, Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Zhengzi Yi
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shaolin Shi
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fadi Salem
- Pathology, Molecular and Cell based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ronald Gordon
- Pathology, Molecular and Cell based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Robert W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | - Weijia Zhang
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yuval Itan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Erwin Böttinger
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Hasso Plattner Institute for Digital Heath at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Digital Health Center, Hasso Plattner Institut, University of Potsdam, Potsdam, Germany
| | - Ilse Daehn
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
115
|
Verbitsky M, Krishnamurthy S, Krithivasan P, Hughes D, Khan A, Marasà M, Vena N, Khosla P, Zhang J, Lim TY, Glessner JT, Weng C, Shang N, Shen Y, Hripcsak G, Hakonarson H, Ionita-Laza I, Levy B, Kenny EE, Loos RJ, Kiryluk K, Sanna-Cherchi S, Crosslin DR, Furth S, Warady BA, Igo RP, Iyengar SK, Wong CS, Parsa A, Feldman HI, Gharavi AG. Genomic Disorders in CKD across the Lifespan. J Am Soc Nephrol 2023; 34:607-618. [PMID: 36302597 PMCID: PMC10103259 DOI: 10.1681/asn.2022060725] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/15/2022] [Indexed: 01/24/2023] Open
Abstract
SIGNIFICANCE STATEMENT Pathogenic structural genetic variants, also known as genomic disorders, have been associated with pediatric CKD. This study extends those results across the lifespan, with genomic disorders enriched in both pediatric and adult patients compared with controls. In the Chronic Renal Insufficiency Cohort study, genomic disorders were also associated with lower serum Mg, lower educational performance, and a higher risk of death. A phenome-wide association study confirmed the link between kidney disease and genomic disorders in an unbiased way. Systematic detection of genomic disorders can provide a molecular diagnosis and refine prediction of risk and prognosis. BACKGROUND Genomic disorders (GDs) are associated with many comorbid outcomes, including CKD. Identification of GDs has diagnostic utility. METHODS We examined the prevalence of GDs among participants in the Chronic Kidney Disease in Children (CKiD) cohort II ( n =248), Chronic Renal Insufficiency Cohort (CRIC) study ( n =3375), Columbia University CKD Biobank (CU-CKD; n =1986), and the Family Investigation of Nephropathy and Diabetes (FIND; n =1318) compared with 30,746 controls. We also performed a phenome-wide association analysis (PheWAS) of GDs in the electronic MEdical Records and GEnomics (eMERGE; n =11,146) cohort. RESULTS We found nine out of 248 (3.6%) CKiD II participants carried a GD, replicating prior findings in pediatric CKD. We also identified GDs in 72 out of 6679 (1.1%) adult patients with CKD in the CRIC, CU-CKD, and FIND cohorts, compared with 199 out of 30,746 (0.65%) GDs in controls (OR, 1.7; 95% CI, 1.3 to 2.2). Among adults with CKD, we found recurrent GDs at the 1q21.1, 16p11.2, 17q12, and 22q11.2 loci. The 17q12 GD (diagnostic of renal cyst and diabetes syndrome) was most frequent, present in 1:252 patients with CKD and diabetes. In the PheWAS, dialysis and neuropsychiatric phenotypes were the top associations with GDs. In CRIC participants, GDs were associated with lower serum magnesium, lower educational achievement, and higher mortality risk. CONCLUSION Undiagnosed GDs are detected both in children and adults with CKD. Identification of GDs in these patients can enable a precise genetic diagnosis, inform prognosis, and help stratify risk in clinical studies. GDs could also provide a molecular explanation for nephropathy and comorbidities, such as poorer neurocognition for a subset of patients.
Collapse
Affiliation(s)
- Miguel Verbitsky
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York
| | | | - Priya Krithivasan
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York
| | - Daniel Hughes
- Institute for Genomic Medicine, Columbia University Medical Center, New York, New York
| | - Atlas Khan
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York
| | - Maddalena Marasà
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York
| | - Natalie Vena
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York
| | - Pavan Khosla
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York
| | - Junying Zhang
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York
| | - Tze Y. Lim
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York
| | - Joseph T. Glessner
- Center for Applied Genomics and Department of Pediatrics, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Chunhua Weng
- Department of Biomedical Informatics, Columbia University, New York, New York
| | - Ning Shang
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York
- Department of Biomedical Informatics, Columbia University, New York, New York
| | - Yufeng Shen
- Department of Systems Biology and Columbia Genome Center, Columbia University, New York, New York
| | - George Hripcsak
- Department of Biomedical Informatics, Columbia University, New York, New York
| | - Hakon Hakonarson
- Center for Applied Genomics and Department of Pediatrics, Perelman School of Medicine, Philadelphia, Pennsylvania
| | | | - Brynn Levy
- Department of Pathology and Cell Biology, Columbia University, New York, New York
| | - Eimear E. Kenny
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ruth J.F. Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York
| | - Simone Sanna-Cherchi
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York
| | - David R. Crosslin
- Division of Biomedical Informatics and Genomics, Tulane University School of Medicine, New Orleans, Louisiana
| | - Susan Furth
- Departments of Pediatrics and Epidemiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bradley A. Warady
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Robert P. Igo
- Department of Population and Quantitative Health Sciences, Case Western Reserve University and Louis Stoke, Cleveland, Ohio
| | - Sudha K. Iyengar
- Department of Population and Quantitative Health Sciences, Case Western Reserve University and Louis Stoke, Cleveland, Ohio
| | - Craig S. Wong
- Division of Pediatric Nephrology, University of New Mexico Children’s Hospital, Albuquerque, New Mexico
| | - Afshin Parsa
- Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | - Harold I. Feldman
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, Philadelphia, Pennsylvania
- Department of Medicine, Perelman School of Medicine, Philadelphia, Pennsylvania
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Ali G. Gharavi
- Division of Nephrology, Department of Medicine, Columbia University, New York, New York
| |
Collapse
|
116
|
Abedi V, Lambert C, Chaudhary D, Rieder E, Avula V, Hwang W, Li J, Zand R. Defining the Age of Young Ischemic Stroke Using Data-Driven Approaches. J Clin Med 2023; 12:jcm12072600. [PMID: 37048683 PMCID: PMC10095415 DOI: 10.3390/jcm12072600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/15/2023] [Accepted: 03/21/2023] [Indexed: 04/03/2023] Open
Abstract
Introduction: The cut-point for defining the age of young ischemic stroke (IS) is clinically and epidemiologically important, yet it is arbitrary and differs across studies. In this study, we leveraged electronic health records (EHRs) and data science techniques to estimate an optimal cut-point for defining the age of young IS. Methods: Patient-level EHRs were extracted from 13 hospitals in Pennsylvania, and used in two parallel approaches. The first approach included ICD9/10, from IS patients to group comorbidities, and computed similarity scores between every patient pair. We determined the optimal age of young IS by analyzing the trend of patient similarity with respect to their clinical profile for different ages of index IS. The second approach used the IS cohort and control (without IS), and built three sets of machine-learning models—generalized linear regression (GLM), random forest (RF), and XGBoost (XGB)—to classify patients for seventeen age groups. After extracting feature importance from the models, we determined the optimal age of young IS by analyzing the pattern of comorbidity with respect to the age of index IS. Both approaches were completed separately for male and female patients. Results: The stroke cohort contained 7555 ISs, and the control included 31,067 patients. In the first approach, the optimal age of young stroke was 53.7 and 51.0 years in female and male patients, respectively. In the second approach, we created 102 models, based on three algorithms, 17 age brackets, and two sexes. The optimal age was 53 (GLM), 52 (RF), and 54 (XGB) for female, and 52 (GLM and RF) and 53 (RF) for male patients. Different age and sex groups exhibited different comorbidity patterns. Discussion: Using a data-driven approach, we determined the age of young stroke to be 54 years for women and 52 years for men in our mainly rural population, in central Pennsylvania. Future validation studies should include more diverse populations.
Collapse
Affiliation(s)
- Vida Abedi
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Health System, Danville, PA 17822, USA
- Department of Public Health Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Clare Lambert
- Department of Neurology, Yale New Haven Hospital, New Haven, CT 06510, USA
| | - Durgesh Chaudhary
- Geisinger Neuroscience Institute, Geisinger Health System, Danville, PA 17822, USA
- Department of Neurology, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Emily Rieder
- Geisinger Commonwealth, School of Medicine, Scranton, PA 18509, USA
| | - Venkatesh Avula
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Health System, Danville, PA 17822, USA
| | - Wenke Hwang
- Department of Public Health Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Jiang Li
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Health System, Danville, PA 17822, USA
| | - Ramin Zand
- Geisinger Neuroscience Institute, Geisinger Health System, Danville, PA 17822, USA
- Department of Neurology, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
- Correspondence: ; Tel.: +1-(717)-531-1804; Fax: +1-(717)-531-0384
| |
Collapse
|
117
|
Xu P, Li H, Xu K, Cui X, Liu Z, Wang X. Genetic variation in BnGRP1 contributes to low phosphorus tolerance in Brassica napus. JOURNAL OF EXPERIMENTAL BOTANY 2023:erad114. [PMID: 36964902 DOI: 10.1093/jxb/erad114] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Indexed: 06/18/2023]
Abstract
Lack of phosphorus (P) is a major environmental factor affecting rapeseed (Brassica napus. L) root growth and development. For breeding purposes, it is crucial to identify the molecular mechanisms of root system architecture (RSA) traits underlying low P tolerance in rapeseed. The natural variations in the glycine-rich protein gene, BnGRP1, were analyzed in the natural population of 400 rapeseed cultivars under low P stress through genome-wide association study (GWAS) and transcriptome analyses. Based on 11 SNP mutations in BnGRP1 sequence, ten types of haplotypes (Hap) were formed. Compared with the other types, the cultivar of BnGRP1Hap1 type in the panel demonstrated the longest root length and heaviest root weight. BnGRP1Hap1 overexpression in rapeseed depicted the ability to enhance its resistance in response to low P tolerance. CRISPR/Cas9-derived BnGRP1Hap4 knockout mutations in rapeseed can lead to sensitivity to low P stress. Furthermore, BnGRP1Hap1 influences the expression of phosphate transporter 1 (PHT1) genes associated with P absorption. Overall, the findings of this study highlight new mechanisms of GRP1 genes in enhancing low P tolerance in rapeseed.
Collapse
Affiliation(s)
- Ping Xu
- College of Agriculture and Forestry Science, Linyi University, Middle of Shuangling Road, Lanshan District, Linyi, 276000, China
| | - Haiyuan Li
- College of Agriculture and Forestry Science, Linyi University, Middle of Shuangling Road, Lanshan District, Linyi, 276000, China
| | - Ke Xu
- College of Agriculture and Forestry Science, Linyi University, Middle of Shuangling Road, Lanshan District, Linyi, 276000, China
| | - Xiaoyu Cui
- College of Agriculture and Forestry Science, Linyi University, Middle of Shuangling Road, Lanshan District, Linyi, 276000, China
| | - Zhenning Liu
- College of Agriculture and Forestry Science, Linyi University, Middle of Shuangling Road, Lanshan District, Linyi, 276000, China
| | - Xiaohua Wang
- College of Agriculture and Forestry Science, Linyi University, Middle of Shuangling Road, Lanshan District, Linyi, 276000, China
| |
Collapse
|
118
|
Truong B, Hull LE, Ruan Y, Huang QQ, Hornsby W, Martin H, van Heel DA, Wang Y, Martin AR, Lee SH, Natarajan P. Integrative polygenic risk score improves the prediction accuracy of complex traits and diseases. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.21.23286110. [PMID: 36865265 PMCID: PMC9980241 DOI: 10.1101/2023.02.21.23286110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Polygenic risk scores (PRS) are an emerging tool to predict the clinical phenotypes and outcomes of individuals. Validation and transferability of existing PRS across independent datasets and diverse ancestries are limited, which hinders the practical utility and exacerbates health disparities. We propose PRSmix, a framework that evaluates and leverages the PRS corpus of a target trait to improve prediction accuracy, and PRSmix+, which incorporates genetically correlated traits to better capture the human genetic architecture. We applied PRSmix to 47 and 32 diseases/traits in European and South Asian ancestries, respectively. PRSmix demonstrated a mean prediction accuracy improvement of 1.20-fold (95% CI: [1.10; 1.3]; P-value = 9.17 × 10-5) and 1.19-fold (95% CI: [1.11; 1.27]; P-value = 1.92 × 10-6), and PRSmix+ improved the prediction accuracy by 1.72-fold (95% CI: [1.40; 2.04]; P-value = 7.58 × 10-6) and 1.42-fold (95% CI: [1.25; 1.59]; P-value = 8.01 × 10-7) in European and South Asian ancestries, respectively. Compared to the previously established cross-trait-combination method with scores from pre-defined correlated traits, we demonstrated that our method can improve prediction accuracy for coronary artery disease up to 3.27-fold (95% CI: [2.1; 4.44]; P-value after FDR correction = 2.6 × 10-4). Our method provides a comprehensive framework to benchmark and leverage the combined power of PRS for maximal performance in a desired target population.
Collapse
Affiliation(s)
- Buu Truong
- Program in Medical and Population Genetics and the Cardiovascular
Disease Initiative, Broad Institute of MIT and Harvard, 415 Main St, Cambridge, MA
02142
- Center for Genomic Medicine and Cardiovascular Research Center,
Massachusetts General Hospital, 185 Cambridge Street, Boston, MA, 02114
| | - Leland E. Hull
- Division of General Internal Medicine, 100 Cambridge Street,
Massachusetts General Hospital, Boston, MA, 02114
- Department of Medicine, Harvard Medical School, 25 Shattuck
Street, Boston, MA 02115
| | - Yunfeng Ruan
- Program in Medical and Population Genetics and the Cardiovascular
Disease Initiative, Broad Institute of MIT and Harvard, 415 Main St, Cambridge, MA
02142
- Center for Genomic Medicine and Cardiovascular Research Center,
Massachusetts General Hospital, 185 Cambridge Street, Boston, MA, 02114
| | - Qin Qin Huang
- Department of Human Genetics, Wellcome Sanger Institute,
Cambridge, UK
| | - Whitney Hornsby
- Program in Medical and Population Genetics and the Cardiovascular
Disease Initiative, Broad Institute of MIT and Harvard, 415 Main St, Cambridge, MA
02142
- Center for Genomic Medicine and Cardiovascular Research Center,
Massachusetts General Hospital, 185 Cambridge Street, Boston, MA, 02114
| | - Hilary Martin
- Department of Human Genetics, Wellcome Sanger Institute,
Cambridge, UK
| | - David A. van Heel
- Blizard Institute, Barts and the London School of Medicine and
Dentistry, Queen Mary University of London, London, UK
| | - Ying Wang
- Program in Medical and Population Genetics and the Cardiovascular
Disease Initiative, Broad Institute of MIT and Harvard, 415 Main St, Cambridge, MA
02142
- Stanley Center for Psychiatric Research, Broad Institute of
Harvard and MIT, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General
Hospital, Boston, MA, USA
| | - Alicia R. Martin
- Stanley Center for Psychiatric Research, Broad Institute of
Harvard and MIT, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General
Hospital, Boston, MA, USA
| | - S. Hong Lee
- Australian Centre for Precision Health, University of South
Australia Cancer Research Institute, University of South Australia, Adelaide, SA, 5000,
Australia
| | - Pradeep Natarajan
- Program in Medical and Population Genetics and the Cardiovascular
Disease Initiative, Broad Institute of MIT and Harvard, 415 Main St, Cambridge, MA
02142
- Center for Genomic Medicine and Cardiovascular Research Center,
Massachusetts General Hospital, 185 Cambridge Street, Boston, MA, 02114
- Department of Medicine, Harvard Medical School, 25 Shattuck
Street, Boston, MA 02115
| |
Collapse
|
119
|
Hemerich D, Smit RAJ, Preuss M, Stalbow L, van der Laan SW, Asselbergs FW, van Setten J, Tragante V. Effect of tissue-grouped regulatory variants associated to type 2 diabetes in related secondary outcomes. Sci Rep 2023; 13:3579. [PMID: 36864090 PMCID: PMC9981672 DOI: 10.1038/s41598-023-30369-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 02/21/2023] [Indexed: 03/04/2023] Open
Abstract
Genome-wide association studies have identified over five hundred loci that contribute to variation in type 2 diabetes (T2D), an established risk factor for many diseases. However, the mechanisms and extent through which these loci contribute to subsequent outcomes remain elusive. We hypothesized that combinations of T2D-associated variants acting on tissue-specific regulatory elements might account for greater risk for tissue-specific outcomes, leading to diversity in T2D disease progression. We searched for T2D-associated variants acting on regulatory elements and expression quantitative trait loci (eQTLs) in nine tissues. We used T2D tissue-grouped variant sets as genetic instruments to conduct 2-Sample Mendelian Randomization (MR) in ten related outcomes whose risk is increased by T2D using the FinnGen cohort. We performed PheWAS analysis to investigate whether the T2D tissue-grouped variant sets had specific predicted disease signatures. We identified an average of 176 variants acting in nine tissues implicated in T2D, and an average of 30 variants acting on regulatory elements that are unique to the nine tissues of interest. In 2-Sample MR analyses, all subsets of regulatory variants acting in different tissues were associated with increased risk of the ten secondary outcomes studied on similar levels. No tissue-grouped variant set was associated with an outcome significantly more than other tissue-grouped variant sets. We did not identify different disease progression profiles based on tissue-specific regulatory and transcriptome information. Bigger sample sizes and other layers of regulatory information in critical tissues may help identify subsets of T2D variants that are implicated in certain secondary outcomes, uncovering system-specific disease progression.
Collapse
Affiliation(s)
- Daiane Hemerich
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roelof A J Smit
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lauren Stalbow
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sander W van der Laan
- Central Diagnostics Laboratory, Division Laboratories, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Folkert W Asselbergs
- Department of Cardiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Health Data Research UK and Institute of Health Informatics, University College London, London, UK
| | - Jessica van Setten
- Department of Cardiology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Vinicius Tragante
- Department of Cardiology, UMC Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
120
|
Wang L, Li X, Montazeri A, MacFarlane AJ, Momoli F, Duthie S, Senekal M, Eguiagaray IM, Munger R, Bennett D, Campbell H, Rubini M, McNulty H, Little J, Theodoratou E. Phenome-wide association study of genetically predicted B vitamins and homocysteine biomarkers with multiple health and disease outcomes: analysis of the UK Biobank. Am J Clin Nutr 2023; 117:564-575. [PMID: 36811473 PMCID: PMC7614280 DOI: 10.1016/j.ajcnut.2023.01.005] [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: 07/12/2022] [Revised: 01/04/2023] [Accepted: 01/09/2023] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Although a number of health outcomes such as CVDs, metabolic-related outcomes, neurological disorders, pregnancy outcomes, and cancers have been identified in relation to B vitamins, evidence is of uneven quality and volume, and there is uncertainty about putative causal relationships. OBJECTIVES To explore the effects of B vitamins and homocysteine on a wide range of health outcomes based on a large biorepository linking biological samples and electronic medical records. METHODS First, we performed a phenome-wide association study (PheWAS) to investigate the associations of genetically predicted plasma concentrations (genetic component of the circulating concentrations) of folate, vitamin B6, vitamin B12, and their metabolite homocysteine with a wide range of disease outcomes (including both prevalent and incident events) among 385,917 individuals in the UK Biobank. Second, 2-sample Mendelian randomization (MR) analysis was used to replicate any observed associations and detect causality. We considered MR P <0.05 as significant for replication. Third, dose-response, mediation, and bioinformatics analyses were carried out to examine any nonlinear trends and to disentangle the underlying mediating biological mechanisms for the identified associations. RESULTS In total, 1117 phenotypes were tested in each PheWAS analysis. After multiple corrections, 32 phenotypic associations of B vitamins and homocysteine were identified. Two-sample MR analysis supported that 3 of them were causal, including associations of higher plasma vitamin B6 with lower risk of calculus of kidney (OR: 0.64; 95% CI: 0.42, 0.97; P = 0.033), higher homocysteine concentration with higher risk of hypercholesterolemia (OR: 1.28, 95% CI: 1.04, 1.56; P = 0.018), and chronic kidney disease (OR: 1.32, 95% CI: 1.06, 1.63; P = 0.012). Significant nonlinear dose-response relationships were observed for the associations of folate with anemia, vitamin B12 with vitamin B-complex deficiencies, anemia and cholelithiasis, and homocysteine with cerebrovascular disease. CONCLUSIONS This study provides strong evidence for the associations of B vitamins and homocysteine with endocrine/metabolic and genitourinary disorders.
Collapse
Affiliation(s)
- Lijuan Wang
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Centre for Global Health, Usher Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Xue Li
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Azita Montazeri
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Franco Momoli
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Susan Duthie
- School of Pharmacy and Life Sciences, Robert Gordon University, Aberdeen, United Kingdom
| | - Marjanne Senekal
- Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Ines Mesa Eguiagaray
- Centre for Global Health, Usher Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Ron Munger
- Department of Nutrition and Food Sciences and the Center for Epidemiologic Studies, Utah State University, Logan, UT, USA
| | - Derrick Bennett
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Harry Campbell
- Centre for Global Health, Usher Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Michele Rubini
- Department of Neuroscience and rehabilitation, University of Ferrara, Ferrara, Italy
| | - Helene McNulty
- Nutrition Innovation Centre for Food and Health, Ulster University, Coleraine, Northern Ireland, United Kingdom
| | - Julian Little
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Evropi Theodoratou
- Centre for Global Health, Usher Institute, The University of Edinburgh, Edinburgh, United Kingdom; Cancer Research UK Edinburgh Centre, The University of Edinburgh MRC Institute of Genetics and Cancer, Edinburgh, United Kingdom.
| |
Collapse
|
121
|
Hatoum AS, Colbert SM, Johnson EC, Huggett SB, Deak JD, Pathak G, Jennings MV, Paul SE, Karcher NR, Hansen I, Baranger DA, Edwards A, Grotzinger A, Substance Use Disorder Working Group of the Psychiatric Genomics
Consortium, Tucker-Drob EM, Kranzler HR, Davis LK, Sanchez-Roige S, Polimanti R, Gelernter J, Edenberg HJ, Bogdan R, Agrawal A. Multivariate genome-wide association meta-analysis of over 1 million subjects identifies loci underlying multiple substance use disorders. NATURE. MENTAL HEALTH 2023; 1:210-223. [PMID: 37250466 PMCID: PMC10217792 DOI: 10.1038/s44220-023-00034-y] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 02/10/2023] [Indexed: 05/31/2023]
Abstract
Genetic liability to substance use disorders can be parsed into loci that confer general or substance-specific addiction risk. We report a multivariate genome-wide association meta-analysis that disaggregates general and substance-specific loci for published summary statistics of problematic alcohol use, problematic tobacco use, cannabis use disorder, and opioid use disorder in a sample of 1,025,550 individuals of European descent and 92,630 individuals of African descent. Nineteen independent SNPs were genome-wide significant (P < 5e-8) for the general addiction risk factor (addiction-rf), which showed high polygenicity. Across ancestries, PDE4B was significant (among other genes), suggesting dopamine regulation as a cross-substance vulnerability. An addiction-rf polygenic risk score was associated with substance use disorders, psychopathologies, somatic conditions, and environments associated with the onset of addictions. Substance-specific loci (9 for alcohol, 32 for tobacco, 5 for cannabis, 1 for opioids) included metabolic and receptor genes. These findings provide insight into genetic risk loci for substance use disorders that could be leveraged as treatment targets.
Collapse
Affiliation(s)
- Alexander S. Hatoum
- Washington University School of Medicine, Department of
Psychiatry, Saint Louis, USA
| | - Sarah M.C. Colbert
- Washington University School of Medicine, Department of
Psychiatry, Saint Louis, USA
| | - Emma C. Johnson
- Washington University School of Medicine, Department of
Psychiatry, Saint Louis, USA
| | | | - Joseph D. Deak
- Department of Psychiatry, Division of Human Genetics, Yale
School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven,
CT, USA
| | - Gita Pathak
- Department of Psychiatry, Division of Human Genetics, Yale
School of Medicine, New Haven, CT, USA
| | - Mariela V. Jennings
- UC San Diego School of Medicine, Department of Psychiatry,
San Diego, CA, USA
| | - Sarah E. Paul
- Department of Psychological & Brain Sciences,
Washington University in St. Louis
| | - Nicole R. Karcher
- Washington University School of Medicine, Department of
Psychiatry, Saint Louis, USA
| | - Isabella Hansen
- Department of Psychological & Brain Sciences,
Washington University in St. Louis
| | - David A.A. Baranger
- Washington University School of Medicine, Department of
Psychiatry, Saint Louis, USA
| | - Alexis Edwards
- Virginia Institute of Psychiatric and Behavioral Genetics,
Virginia Commonwealth University, Richmond, VA, USA
| | - Andrew Grotzinger
- University of Colorado-Boulder, Institute for Behavioral
Genetics, Boulder, CO, USA
| | | | - Elliot M. Tucker-Drob
- University of Texas at Austin, Department of Psychology and
Population Research Center, Austin, TX, USA
| | - Henry R. Kranzler
- Center for Studies of Addiction, Department of
Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia,
PA, USA
- VISN 4 MIRECC, Crescenz VAMC, Philadelphia, PA, USA
| | - Lea K. Davis
- Department of Medicine, Division of Genetic Medicine,
Vanderbilt University, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences,
Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Informatics, Vanderbilt
University Medical Center, Nashville, TN, USA
| | - Sandra Sanchez-Roige
- UC San Diego School of Medicine, Department of Psychiatry,
San Diego, CA, USA
- Department of Medicine, Division of Genetic Medicine,
Vanderbilt University, Nashville, TN, USA
| | - Renato Polimanti
- Department of Psychiatry, Division of Human Genetics, Yale
School of Medicine, New Haven, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven,
CT, USA
| | - Joel Gelernter
- Department of Psychiatry, Division of Human Genetics, Yale
School of Medicine, New Haven, CT, USA
- University of Texas at Austin, Department of Psychology and
Population Research Center, Austin, TX, USA
- Department of Genetics, Yale School of Medicine, New
Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New
Haven, CT, USA
| | - Howard J. Edenberg
- Department of Medical and Molecular Genetics, Indiana
University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana
University School of Medicine, Indianapolis, IN, USA
| | - Ryan Bogdan
- Department of Psychological & Brain Sciences,
Washington University in St. Louis
| | - Arpana Agrawal
- Washington University School of Medicine, Department of
Psychiatry, Saint Louis, USA
| |
Collapse
|
122
|
Miller-Fleming TW, Allos A, Gantz E, Yu D, Isaacs DA, Mathews CA, Scharf JM, Davis LK. Developing a Phenotype Risk Score for Tic Disorders in a Large, Clinical Biobank. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.21.23286253. [PMID: 36865201 PMCID: PMC9980249 DOI: 10.1101/2023.02.21.23286253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Importance Tics are a common feature of early-onset neurodevelopmental disorders, characterized by involuntary and repetitive movements or sounds. Despite affecting up to 2% of young children and having a genetic contribution, the underlying causes remain poorly understood, likely due to the complex phenotypic and genetic heterogeneity among affected individuals. Objective In this study, we leverage dense phenotype information from electronic health records to identify the disease features associated with tic disorders within the context of a clinical biobank. These disease features are then used to generate a phenotype risk score for tic disorder. Design Using de-identified electronic health records from a tertiary care center, we extracted individuals with tic disorder diagnosis codes. We performed a phenome-wide association study to identify the features enriched in tic cases versus controls (N=1,406 and 7,030; respectively). These disease features were then used to generate a phenotype risk score for tic disorder, which was applied across an independent set of 90,051 individuals. A previously curated set of tic disorder cases from an electronic health record algorithm followed by clinician chart review was used to validate the tic disorder phenotype risk score. Main Outcomes and Measures Phenotypic patterns associated with a tic disorder diagnosis in the electronic health record. Results Our tic disorder phenome-wide association study revealed 69 significantly associated phenotypes, predominantly neuropsychiatric conditions, including obsessive compulsive disorder, attention-deficit hyperactivity disorder, autism, and anxiety. The phenotype risk score constructed from these 69 phenotypes in an independent population was significantly higher among clinician-validated tic cases versus non-cases. Conclusions and Relevance Our findings provide support for the use of large-scale medical databases to better understand phenotypically complex diseases, such as tic disorders. The tic disorder phenotype risk score provides a quantitative measure of disease risk that can be leveraged for the assignment of individuals in case-control studies or for additional downstream analyses.
Collapse
Affiliation(s)
- Tyne W. Miller-Fleming
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, TN, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Annmarie Allos
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, TN, USA
- Department of Cognitive Science, Dartmouth College, Hanover, NH, USA
| | - Emily Gantz
- Department of Pediatric Neurology, Children’s Hospital of Alabama, Birmingham, AL, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Monroe Carell Jr. Children’s Hospital at Vanderbilt, Nashville, TN, USA
| | - Dongmei Yu
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David A. Isaacs
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Monroe Carell Jr. Children’s Hospital at Vanderbilt, Nashville, TN, USA
| | - Carol A. Mathews
- Department of Psychiatry, Genetics Institute, Center for OCD, Anxiety and Related Disorders, University of Florida, Gainesville, FL, USA
| | - Jeremiah M. Scharf
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lea K. Davis
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, TN, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, TN, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, TN, USA
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, TN, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, TN, USA
| |
Collapse
|
123
|
Integrating oculomics with genomics reveals imaging biomarkers for preventive and personalized prediction of arterial aneurysms. EPMA J 2023; 14:73-86. [PMID: 36866161 PMCID: PMC9971392 DOI: 10.1007/s13167-023-00315-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/15/2023] [Indexed: 02/15/2023]
Abstract
Objective Arterial aneurysms are life-threatening but usually asymptomatic before requiring hospitalization. Oculomics of retinal vascular features (RVFs) extracted from retinal fundus images can reflect systemic vascular properties and therefore were hypothesized to provide valuable information on detecting the risk of aneurysms. By integrating oculomics with genomics, this study aimed to (i) identify predictive RVFs as imaging biomarkers for aneurysms and (ii) evaluate the value of these RVFs in supporting early detection of aneurysms in the context of predictive, preventive and personalized medicine (PPPM). Methods This study involved 51,597 UK Biobank participants who had retinal images available to extract oculomics of RVFs. Phenome-wide association analyses (PheWASs) were conducted to identify RVFs associated with the genetic risks of the main types of aneurysms, including abdominal aortic aneurysm (AAA), thoracic aneurysm (TAA), intracranial aneurysm (ICA) and Marfan syndrome (MFS). An aneurysm-RVF model was then developed to predict future aneurysms. The performance of the model was assessed in both derivation and validation cohorts and was compared with other models employing clinical risk factors. An RVF risk score was derived from our aneurysm-RVF model to identify patients with an increased risk of aneurysms. Results PheWAS identified a total of 32 RVFs that were significantly associated with the genetic risks of aneurysms. Of these, the number of vessels in the optic disc ('ntreeA') was associated with both AAA (β = -0.36, P = 6.75e-10) and ICA (β = -0.11, P = 5.51e-06). In addition, the mean angles between each artery branch ('curveangle_mean_a') were commonly associated with 4 MFS genes (FBN1: β = -0.10, P = 1.63e-12; COL16A1: β = -0.07, P = 3.14e-09; LOC105373592: β = -0.06, P = 1.89e-05; C8orf81/LOC441376: β = 0.07, P = 1.02e-05). The developed aneurysm-RVF model showed good discrimination ability in predicting the risks of aneurysms. In the derivation cohort, the C-index of the aneurysm-RVF model was 0.809 [95% CI: 0.780-0.838], which was similar to the clinical risk model (0.806 [0.778-0.834]) but higher than the baseline model (0.739 [0.733-0.746]). Similar performance was observed in the validation cohort, with a C-index of 0.798 (0.727-0.869) for the aneurysm-RVF model, 0.795 (0.718-0.871) for the clinical risk model and 0.719 (0.620-0.816) for the baseline model. An aneurysm risk score was derived from the aneurysm-RVF model for each study participant. The individuals in the upper tertile of the aneurysm risk score had a significantly higher risk of aneurysm compared to those in the lower tertile (hazard ratio = 17.8 [6.5-48.8], P = 1.02e-05). Conclusion We identified a significant association between certain RVFs and the risk of aneurysms and revealed the impressive capability of using RVFs to predict the future risk of aneurysms by a PPPM approach. Our finds have great potential to support not only the predictive diagnosis of aneurysms but also a preventive and more personalized screening plan which may benefit both patients and the healthcare system. Graphical abstract Supplementary Information The online version contains supplementary material available at 10.1007/s13167-023-00315-7.
Collapse
|
124
|
Zawistowski M, Fritsche LG, Pandit A, Vanderwerff B, Patil S, Schmidt EM, VandeHaar P, Willer CJ, Brummett CM, Kheterpal S, Zhou X, Boehnke M, Abecasis GR, Zöllner S. The Michigan Genomics Initiative: A biobank linking genotypes and electronic clinical records in Michigan Medicine patients. CELL GENOMICS 2023; 3:100257. [PMID: 36819667 PMCID: PMC9932985 DOI: 10.1016/j.xgen.2023.100257] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 06/07/2022] [Accepted: 01/05/2023] [Indexed: 02/04/2023]
Abstract
Biobanks of linked clinical patient histories and biological samples are an efficient strategy to generate large cohorts for modern genetics research. Biobank recruitment varies by factors such as geographic catchment and sampling strategy, which affect biobank demographics and research utility. Here, we describe the Michigan Genomics Initiative (MGI), a single-health-system biobank currently consisting of >91,000 participants recruited primarily during surgical encounters at Michigan Medicine. The surgical enrollment results in a biobank enriched for many diseases and ideally suited for a disease genetics cohort. Compared with the much larger population-based UK Biobank, MGI has higher prevalence for nearly all diagnosis-code-based phenotypes and larger absolute case counts for many phenotypes. Genome-wide association study (GWAS) results replicate known findings, thereby validating the genetic and clinical data. Our results illustrate that opportunistic biobank sampling within single health systems provides a unique and complementary resource for exploring the genetics of complex diseases.
Collapse
Affiliation(s)
- Matthew Zawistowski
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
| | - Lars G. Fritsche
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
| | - Anita Pandit
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
| | - Brett Vanderwerff
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
| | - Snehal Patil
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
| | - Ellen M. Schmidt
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
| | - Peter VandeHaar
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
| | - Cristen J. Willer
- Department of Internal Medicine, Division of Cardiovascular Medicine, Department of Human Genetics, University of Michigan, Ann Arbor, MI 48103, USA
| | - Chad M. Brummett
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI 48103, USA
| | - Sachin Kheterpal
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI 48103, USA
| | - Xiang Zhou
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
| | - Gonçalo R. Abecasis
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
- Regeneron Genetics Center, Tarrytown, NY 10591, USA
| | - Sebastian Zöllner
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48103, USA
- Department of Psychiatry, University of Michigan, Ann Arbor, MI 48103, USA
| |
Collapse
|
125
|
Fritsche LG, Jin W, Admon AJ, Mukherjee B. Characterizing and Predicting Post-Acute Sequelae of SARS CoV-2 Infection (PASC) in a Large Academic Medical Center in the US. J Clin Med 2023; 12:1328. [PMID: 36835863 PMCID: PMC9967320 DOI: 10.3390/jcm12041328] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/30/2023] [Accepted: 02/04/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND A growing number of Coronavirus Disease-2019 (COVID-19) survivors are affected by post-acute sequelae of SARS CoV-2 infection (PACS). Using electronic health record data, we aimed to characterize PASC-associated diagnoses and develop risk prediction models. METHODS In our cohort of 63,675 patients with a history of COVID-19, 1724 (2.7%) had a recorded PASC diagnosis. We used a case-control study design and phenome-wide scans to characterize PASC-associated phenotypes of the pre-, acute-, and post-COVID-19 periods. We also integrated PASC-associated phenotypes into phenotype risk scores (PheRSs) and evaluated their predictive performance. RESULTS In the post-COVID-19 period, known PASC symptoms (e.g., shortness of breath, malaise/fatigue) and musculoskeletal, infectious, and digestive disorders were enriched among PASC cases. We found seven phenotypes in the pre-COVID-19 period (e.g., irritable bowel syndrome, concussion, nausea/vomiting) and sixty-nine phenotypes in the acute-COVID-19 period (predominantly respiratory, circulatory, neurological) associated with PASC. The derived pre- and acute-COVID-19 PheRSs stratified risk well, e.g., the combined PheRSs identified a quarter of the cohort with a history of COVID-19 with a 3.5-fold increased risk (95% CI: 2.19, 5.55) for PASC compared to the bottom 50%. CONCLUSIONS The uncovered PASC-associated diagnoses across categories highlighted a complex arrangement of presenting and likely predisposing features, some with potential for risk stratification approaches.
Collapse
Affiliation(s)
- Lars G. Fritsche
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Center for Precision Health Data Science, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Weijia Jin
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Center for Precision Health Data Science, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Andrew J. Admon
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- VA Center for Clinical Management Research, LTC Charles S. Kettles VA Medical Center, Ann Arbor, MI 48109, USA
| | - Bhramar Mukherjee
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Center for Precision Health Data Science, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Michigan Institute for Data Science, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
126
|
Wu Y, Bayrak CS, Dong B, He S, Stenson PD, Cooper DN, Itan Y, Chen L. Identifying shared genetic factors underlying epilepsy and congenital heart disease in Europeans. Hum Genet 2023; 142:275-288. [PMID: 36352240 DOI: 10.1007/s00439-022-02502-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022]
Abstract
Epilepsy (EP) and congenital heart disease (CHD) are two apparently unrelated diseases that nevertheless display substantial mutual comorbidity. Thus, while congenital heart defects are associated with an elevated risk of developing epilepsy, the incidence of epilepsy in CHD patients correlates with CHD severity. Although genetic determinants have been postulated to underlie the comorbidity of EP and CHD, the precise genetic etiology is unknown. We performed variant and gene association analyses on EP and CHD patients separately, using whole exomes of genetically identified Europeans from the UK Biobank and Mount Sinai BioMe Biobank. We prioritized biologically plausible candidate genes and investigated the enriched pathways and other identified comorbidities by biological proximity calculation, pathway analyses, and gene-level phenome-wide association studies. Our variant- and gene-level results point to the Voltage-Gated Calcium Channels (VGCC) pathway as being a unifying framework for EP and CHD comorbidity. Additionally, pathway-level analyses indicated that the functions of disease-associated genes partially overlap between the two disease entities. Finally, phenome-wide association analyses of prioritized candidate genes revealed that cerebral blood flow and ulcerative colitis constitute the two main traits associated with both EP and CHD.
Collapse
Affiliation(s)
- Yiming Wu
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Cigdem Sevim Bayrak
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bosi Dong
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Shixu He
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Peter D Stenson
- Institute of Medical Genetics, Cardiff University, Cardiff, UK
| | - David N Cooper
- Institute of Medical Genetics, Cardiff University, Cardiff, UK
| | - Yuval Itan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY, USA.
| | - Lei Chen
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
127
|
Landstrom AP, Yang Q, Sun B, Perelli RM, Bidzimou MT, Zhang Z, Aguilar-Sanchez Y, Alsina KM, Cao S, Reynolds JO, Word TA, van der Sangen NM, Wells Q, Kannankeril PJ, Ludwig A, Kim JJ, Wehrens XH. Reduction in Junctophilin 2 Expression in Cardiac Nodal Tissue Results in Intracellular Calcium-Driven Increase in Nodal Cell Automaticity. Circ Arrhythm Electrophysiol 2023; 16:e010858. [PMID: 36706317 PMCID: PMC9974897 DOI: 10.1161/circep.122.010858] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 01/06/2023] [Indexed: 01/29/2023]
Abstract
BACKGROUND Spontaneously depolarizing nodal cells comprise the pacemaker of the heart. Intracellular calcium (Ca2+) plays a critical role in mediating nodal cell automaticity and understanding this so-called Ca2+ clock is critical to understanding nodal arrhythmias. We previously demonstrated a role for Jph2 (junctophilin 2) in regulating Ca2+-signaling through inhibition of RyR2 (ryanodine receptor 2) Ca2+ leak in cardiac myocytes; however, its role in pacemaker function and nodal arrhythmias remains unknown. We sought to determine whether nodal Jph2 expression silencing causes increased sinoatrial and atrioventricular nodal cell automaticity due to aberrant RyR2 Ca2+ leak. METHODS A tamoxifen-inducible, nodal tissue-specific, knockdown mouse of Jph2 was achieved using a Cre-recombinase-triggered short RNA hairpin directed against Jph2 (Hcn4:shJph2). In vivo cardiac rhythm was monitored by surface ECG, implantable cardiac telemetry, and intracardiac electrophysiology studies. Intracellular Ca2+ imaging was performed using confocal-based line scans of isolated nodal cells loaded with fluorescent Ca2+ reporter Cal-520. Whole cell patch clamp was conducted on isolated nodal cells to determine action potential kinetics and sodium-calcium exchanger function. RESULTS Hcn4:shJph2 mice demonstrated a 40% reduction in nodal Jph2 expression, resting sinus tachycardia, and impaired heart rate response to pharmacologic stress. In vivo intracardiac electrophysiology studies and ex vivo optical mapping demonstrated accelerated junctional rhythm originating from the atrioventricular node. Hcn4:shJph2 nodal cells demonstrated increased and irregular Ca2+ transient generation with increased Ca2+ spark frequency and Ca2+ leak from the sarcoplasmic reticulum. This was associated with increased nodal cell AP firing rate, faster diastolic repolarization rate, and reduced sodium-calcium exchanger activity during repolarized states compared to control. Phenome-wide association studies of the JPH2 locus identified an association with sinoatrial nodal disease and atrioventricular nodal block. CONCLUSIONS Nodal-specific Jph2 knockdown causes increased nodal automaticity through increased Ca2+ leak from intracellular stores. Dysregulated intracellular Ca2+ underlies nodal arrhythmogenesis in this mouse model.
Collapse
Affiliation(s)
- Andrew P. Landstrom
- Dept of Pediatrics, Division of Cardiology, Duke Univ School of Medicine, Durham, NC
- Dept of Cell Biology, Duke Univ School of Medicine, Durham, NC
| | - Qixin Yang
- Dept of Pediatrics, Division of Cardiology, Duke Univ School of Medicine, Durham, NC
- Dept of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang Univ, Hangzhou, China
| | - Bo Sun
- Dept of Pediatrics, Division of Cardiology, Duke Univ School of Medicine, Durham, NC
| | | | | | - Zhushan Zhang
- Dept of Cell Biology, Duke Univ School of Medicine, Durham, NC
| | - Yuriana Aguilar-Sanchez
- Integrative Molecular & Biomedical Sciences Program, Baylor College of Medicine, Houston, TX
| | - Katherina M. Alsina
- Integrative Molecular & Biomedical Sciences Program, Baylor College of Medicine, Houston, TX
| | - Shuyi Cao
- Dept of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX
| | - Julia O. Reynolds
- Dept of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX
| | - Tarah A. Word
- Dept of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX
| | | | - Quinn Wells
- Depts of Medicine, Pharmacology, and Biomedical Informatics, Vanderbilt Univ School of Medicine, Nashville, TN
| | - Prince J. Kannankeril
- Center for Pediatric Precision Medicine, Dept of Pediatrics, Vanderbilt Univ School of Medicine, Nashville, TN
| | - Andreas Ludwig
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jeffrey J. Kim
- Dept of Pediatrics, Section of Cardiology, Baylor College of Medicine, Houston, TX
| | - Xander H.T. Wehrens
- Dept of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX
- Dept of Pediatrics, Section of Cardiology, Baylor College of Medicine, Houston, TX
- Depts of Neuroscience & Center for Space Medicine and the Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX
| |
Collapse
|
128
|
Ung CY, Warwick A, Onoufriadis A, Barker JN, Parsons M, McGrath JA, Shaw TJ, Dand N. Comorbidities of Keloid and Hypertrophic Scars Among Participants in UK Biobank. JAMA Dermatol 2023; 159:172-181. [PMID: 36598763 PMCID: PMC9857738 DOI: 10.1001/jamadermatol.2022.5607] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/26/2022] [Indexed: 01/05/2023]
Abstract
Importance Keloids and hypertrophic scars (excessive scarring) are relatively understudied disfiguring chronic skin conditions with high treatment resistance. Objective To evaluate established comorbidities of excessive scarring in European individuals, with comparisons across ethnic groups, and to identify novel comorbidities via a phenome-wide association study (PheWAS). Design, Setting, and Participants This multicenter cross-sectional population-based cohort study used UK Biobank (UKB) data and fitted logistic regression models for testing associations between excessive scarring and a variety of outcomes, including previously studied comorbidities and 1518 systematically defined disease categories. Additional modeling was performed within subgroups of participants defined by self-reported ethnicity (as defined in UK Biobank). Of 502 701 UKB participants, analyses were restricted to 230078 individuals with linked primary care records. Exposures Keloid or hypertrophic scar diagnoses. Main Outcomes and Measures Previously studied disease associations (hypertension, uterine leiomyoma, vitamin D deficiency, atopic eczema) and phenotypes defined in the PheWAS Catalog. Results Of the 972 people with excessive scarring, there was a higher proportion of female participants compared with the 229 106 controls (65% vs 55%) and a lower proportion of White ethnicity (86% vs 95%); mean (SD) age of the total cohort was 64 (8) years. Associations were identified with hypertension and atopic eczema in models accounting for age, sex, and ethnicity, and the association with atopic eczema (odds ratio [OR], 1.68; 95% CI, 1.36-2.07; P < .001) remained statistically significant after accounting for additional potential confounders. Fully adjusted analyses within ethnic groups revealed associations with hypertension in Black participants (OR, 2.05; 95% CI, 1.13-3.72; P = .02) and with vitamin D deficiency in Asian participants (OR, 2.24; 95% CI, 1.26-3.97; P = .006). The association with uterine leiomyoma was borderline significant in Black women (OR, 1.93; 95% CI, 1.00-3.71; P = .05), whereas the association with atopic eczema was significant in White participants (OR, 1.68; 95% CI, 1.34-2.12; P < .001) and showed a similar trend in Asian (OR, 2.17; 95% CI, 1.01-4.67; P = .048) and Black participants (OR, 1.89; 95% CI, 0.83-4.28; P = .13). The PheWAS identified 110 significant associations across disease systems; of the nondermatological, musculoskeletal disease and pain symptoms were prominent. Conclusions and Relevance This cross-sectional study validated comorbidities of excessive scarring in UKB with comprehensive coverage of health outcomes. It also documented additional phenome-wide associations that will serve as a reference for future studies to investigate common underlying pathophysiologic mechanisms.
Collapse
Affiliation(s)
- Chuin Y. Ung
- St John’s Institute of Dermatology, School of Basic and Medical Biosciences, King’s College London, London, United Kingdom
- Centre for Inflammation Biology & Cancer Immunology, King’s College London, London, United Kingdom
| | - Alasdair Warwick
- University College London Institute of Cardiovascular Science, London, United Kingdom
| | - Alexandros Onoufriadis
- St John’s Institute of Dermatology, School of Basic and Medical Biosciences, King’s College London, London, United Kingdom
| | - Jonathan N. Barker
- St John’s Institute of Dermatology, School of Basic and Medical Biosciences, King’s College London, London, United Kingdom
| | - Maddy Parsons
- Randall Division of Cell and Molecular Biophysics, King’s College London, London, United Kingdom
| | - John A. McGrath
- St John’s Institute of Dermatology, School of Basic and Medical Biosciences, King’s College London, London, United Kingdom
| | - Tanya J. Shaw
- Centre for Inflammation Biology & Cancer Immunology, King’s College London, London, United Kingdom
| | - Nick Dand
- Department of Medical and Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
129
|
Brice AN, Vanderlinden LA, Marker KM, Mayer D, Lin M, Rafaels N, Shortt JA, Romero A, Lowery JT, Gignoux CR, Johnson RK. COVID-19 Mortality in the Colorado Center for Personalized Medicine Biobank. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:2368. [PMID: 36767733 PMCID: PMC9916246 DOI: 10.3390/ijerph20032368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 06/18/2023]
Abstract
Over 6.37 million people have died from COVID-19 worldwide, but factors influencing COVID-19-related mortality remain understudied. We aimed to describe and identify risk factors for COVID-19 mortality in the Colorado Center for Personalized Medicine (CCPM) Biobank using integrated data sources, including Electronic Health Records (EHRs). We calculated cause-specific mortality and case-fatality rates for COVID-19 and common pre-existing health conditions defined by diagnostic phecodes and encounters in EHRs. We performed multivariable logistic regression analyses of the association between each pre-existing condition and COVID-19 mortality. Of the 155,859 Biobank participants enrolled as of July 2022, 20,797 had been diagnosed with COVID-19. Of 5334 Biobank participants who had died, 190 were attributed to COVID-19. The case-fatality rate was 0.91% and the COVID-19 mortality rate was 122 per 100,000 persons. The odds of dying from COVID-19 were significantly increased among older men, and those with 14 of the 61 pre-existing conditions tested, including hypertensive chronic kidney disease (OR: 10.14, 95% CI: 5.48, 19.16) and type 2 diabetes with renal manifestations (OR: 5.59, 95% CI: 3.42, 8.97). Male patients who are older and have pre-existing kidney diseases may be at higher risk for death from COVID-19 and may require special care.
Collapse
Affiliation(s)
- Amanda N. Brice
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO 80045, USA
| | | | - Katie M. Marker
- Human Medical Genetics and Genomics Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - David Mayer
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Colorado Center for Personalized Medicine, Aurora, CO 80045, USA
| | - Meng Lin
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Colorado Center for Personalized Medicine, Aurora, CO 80045, USA
| | - Nicholas Rafaels
- Colorado Center for Personalized Medicine, Aurora, CO 80045, USA
| | - Jonathan A. Shortt
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Colorado Center for Personalized Medicine, Aurora, CO 80045, USA
| | - Alex Romero
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jan T. Lowery
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO 80045, USA
- Colorado Center for Personalized Medicine, Aurora, CO 80045, USA
| | - Christopher R. Gignoux
- Human Medical Genetics and Genomics Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Colorado Center for Personalized Medicine, Aurora, CO 80045, USA
| | - Randi K. Johnson
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO 80045, USA
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Colorado Center for Personalized Medicine, Aurora, CO 80045, USA
| |
Collapse
|
130
|
Petty LE, Silva R, de Souza LC, Vieira AR, Shaw DM, Below JE, Letra A. Genome-wide association study identifies novel risk loci for apical periodontitis. RESEARCH SQUARE 2023:rs.3.rs-2515434. [PMID: 36747740 PMCID: PMC9901028 DOI: 10.21203/rs.3.rs-2515434/v1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Apical periodontitis (AP) is a common consequence of root canal infection leading to periapical bone resorption. Microbial and host genetic factors, and their interactions, have been shown to play a role in AP development and progression. Variations in a few genes have been reported in association with AP, however, the lack of genome-wide studies has hindered progress in understanding the mechanisms involved in AP. Here, we report the first genome-wide association study of AP in a well-characterized population. Male and female adults (n=932) presenting with deep caries with AP (cases) or without AP (controls) were included. Genotyping was performed using the Illumina Expanded Multi-Ethnic Genotyping Array. Single-variant association testing was performed adjusting for sex and five principal components. Subphenotype association testing, analyses of genetically regulated gene expression, polygenic risk score and phenome-wide association (PheWAS) analyses were also performed. Eight loci reached near-genome-wide significant association with AP (p < 5 x 10-6); gene-focused analyses replicated three previously reported associations (p < 8.9 x 10-5). Sex-specific and subphenotype analyses revealed additional significant associations with variants genome-wide. Functionally oriented gene-based analyses revealed eight genes significantly associated with AP (p < 5 x 10-5), and PheWAS analysis revealed 33 phecodes associated with AP risk score (p < 3.08 x 10-5). This study identified novel genes/loci contributing to AP and revealed specific contributions to AP risk in males and females. Importantly, we identified additional systemic conditions significantly associated with AP risk. Our findings provide strong evidence for host-mediated effects on AP susceptibility.
Collapse
Affiliation(s)
- L E Petty
- Vanderbilt University Medical Center
| | - R Silva
- University of Pittsburgh School of Dental Medicine
| | - L Chaves de Souza
- University of Texas Health Science Center at Houston School of Dentistry: The University of Texas Health Science Center at Houston School of Dentistry
| | - A R Vieira
- University of Pittsburgh School of Dental Medicine
| | - D M Shaw
- Vanderbilt University Medical Center
| | - J E Below
- Vanderbilt University Medical Center
| | | |
Collapse
|
131
|
Kerchberger VE, Peterson JF, Wei WQ. Scanning the medical phenome to identify new diagnoses after recovery from COVID-19 in a US cohort. J Am Med Inform Assoc 2023; 30:233-244. [PMID: 36005898 PMCID: PMC9452157 DOI: 10.1093/jamia/ocac159] [Citation(s) in RCA: 2] [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: 04/07/2022] [Revised: 06/29/2022] [Accepted: 08/23/2022] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE COVID-19 survivors are at risk for long-term health effects, but assessing the sequelae of COVID-19 at large scales is challenging. High-throughput methods to efficiently identify new medical problems arising after acute medical events using the electronic health record (EHR) could improve surveillance for long-term consequences of acute medical problems like COVID-19. MATERIALS AND METHODS We augmented an existing high-throughput phenotyping method (PheWAS) to identify new diagnoses occurring after an acute temporal event in the EHR. We then used the temporal-informed phenotypes to assess development of new medical problems among COVID-19 survivors enrolled in an EHR cohort of adults tested for COVID-19 at Vanderbilt University Medical Center. RESULTS The study cohort included 186 105 adults tested for COVID-19 from March 5, 2020 to November 1, 2021; of which 30 088 (16.2%) tested positive. Median follow-up after testing was 412 days (IQR 274-528). Our temporal-informed phenotyping was able to distinguish phenotype chapters based on chronicity of their constituent diagnoses. PheWAS with temporal-informed phenotypes identified increased risk for 43 diagnoses among COVID-19 survivors during outpatient follow-up, including multiple new respiratory, cardiovascular, neurological, and pregnancy-related conditions. Findings were robust to sensitivity analyses, and several phenotypic associations were supported by changes in outpatient vital signs or laboratory tests from the pretesting to postrecovery period. CONCLUSION Temporal-informed PheWAS identified new diagnoses affecting multiple organ systems among COVID-19 survivors. These findings can inform future efforts to enable longitudinal health surveillance for survivors of COVID-19 and other acute medical conditions using the EHR.
Collapse
Affiliation(s)
- Vern Eric Kerchberger
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Josh F Peterson
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
132
|
Pillalamarri V, Shi W, Say C, Yang S, Lane J, Guallar E, Pankratz N, Arking DE. Whole-exome sequencing in 415,422 individuals identifies rare variants associated with mitochondrial DNA copy number. HGG ADVANCES 2023; 4:100147. [PMID: 36311265 PMCID: PMC9615038 DOI: 10.1016/j.xhgg.2022.100147] [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: 07/06/2022] [Accepted: 09/23/2022] [Indexed: 10/14/2022] Open
Abstract
Inter-individual variation in the number of copies of the mitochondrial genome, called mitochondrial DNA copy number (mtDNA-CN), reflects mitochondrial function and has been associated with various aging-related diseases. We examined 415,422 exomes of self-reported White ancestry individuals from the UK Biobank and tested the impact of rare variants, at the level of single variants and through aggregate variant-set tests, on mtDNA-CN. A survey across nine variant sets tested enrichment of putatively causal variants and identified 14 genes at experiment-wide significance and three genes at marginal significance. These included associations at known mtDNA depletion syndrome genes (mtDNA helicase TWNK, p = 1.1 × 10-30; mitochondrial transcription factor TFAM, p = 4.3 × 10-15; mtDNA maintenance exonuclease MGME1, p = 2.0 × 10-6) and the V617F dominant gain-of-function mutation in the tyrosine kinase JAK2 (p = 2.7 × 10-17), associated with myeloproliferative disease. Novel genes included the ATP-dependent protease CLPX (p = 8.4 × 10-9), involved in mitochondrial proteome quality, and the mitochondrial adenylate kinase AK2 (p = 4.7 × 10-8), involved in hematopoiesis. The most significant association was a missense variant in SAMHD1 (p = 4.2 × 10-28), found on a rare, 1.2-Mb shared ancestral haplotype on chromosome 20. SAMHD1 encodes a cytoplasmic host restriction factor involved in viral defense response and the mitochondrial nucleotide salvage pathway, and is associated with Aicardi-Goutières syndrome 5, a childhood encephalopathy and chronic inflammatory response disorder. Rare variants were enriched in Mendelian mtDNA depletion syndrome loci, and these variants implicated core processes in mtDNA replication, nucleoid structure formation, and maintenance. These data indicate that strong-effect mutations from the nuclear genome contribute to the genetic architecture of mtDNA-CN.
Collapse
Affiliation(s)
- Vamsee Pillalamarri
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Predoctoral Program in Human Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Maryland Genetics Epidemiology and Medicine Training Program, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Wen Shi
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Conrad Say
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Stephanie Yang
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Vertex Pharmaceuticals, Inc., Boston, MA 02210, USA
| | - John Lane
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Eliseo Guallar
- Departments of Epidemiology and Medicine and Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Dan E. Arking
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
133
|
Campos LA, Baltatu OC, Senar S, Ghimouz R, Alefishat E, Cipolla-Neto J. Multiplatform-Integrated Identification of Melatonin Targets for a Triad of Psychosocial-Sleep/Circadian-Cardiometabolic Disorders. Int J Mol Sci 2023; 24:ijms24010860. [PMID: 36614302 PMCID: PMC9821171 DOI: 10.3390/ijms24010860] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/10/2022] [Accepted: 12/30/2022] [Indexed: 01/05/2023] Open
Abstract
Several psychosocial, sleep/circadian, and cardiometabolic disorders have intricately interconnected pathologies involving melatonin disruption. Therefore, we hypothesize that melatonin could be a therapeutic target for treating potential comorbid diseases associated with this triad of psychosocial-sleep/circadian-cardiometabolic disorders. We investigated melatonin's target prediction and tractability for this triad of disorders. The melatonin's target prediction for the proposed psychosocial-sleep/circadian-cardiometabolic disorder triad was investigated using databases from Europe PMC, ChEMBL, Open Targets Genetics, Phenodigm, and PheWAS. The association scores for melatonin receptors MT1 and MT2 with this disorder triad were explored for evidence of target-disease predictions. The potential of melatonin as a tractable target in managing the disorder triad was investigated using supervised machine learning to identify melatonin activities in cardiovascular, neuronal, and metabolic assays at the cell, tissue, and organism levels in a curated ChEMBL database. Target-disease visualization was done by graphs created using "igraph" library-based scripts and displayed using the Gephi ForceAtlas algorithm. The combined Europe PMC (data type: text mining), ChEMBL (data type: drugs), Open Targets Genetics Portal (data type: genetic associations), PhenoDigm (data type: animal models), and PheWAS (data type: genetic associations) databases yielded types and varying levels of evidence for melatonin-disease triad correlations. Of the investigated databases, 235 association scores of melatonin receptors with the targeted diseases were greater than 0.2; to classify the evidence per disease class: 37% listed psychosocial disorders, 9% sleep/circadian disorders, and 54% cardiometabolic disorders. Using supervised machine learning, 546 cardiovascular, neuronal, or metabolic experimental assays with predicted or measured melatonin activity scores were identified in the ChEMBL curated database. Of 248 registered trials, 144 phase I to IV trials for melatonin or agonists have been completed, of which 33.3% were for psychosocial disorders, 59.7% were for sleep/circadian disorders, and 6.9% were for cardiometabolic disorders. Melatonin's druggability was evidenced by evaluating target prediction and tractability for the triad of psychosocial-sleep/circadian-cardiometabolic disorders. While melatonin research and development in sleep/circadian and psychosocial disorders is more advanced, as evidenced by melatonin association scores, substantial evidence on melatonin discovery in cardiovascular and metabolic disorders supports continued R&D in cardiometabolic disorders, as evidenced by melatonin activity scores. A multiplatform analysis provided an integrative assessment of the target-disease investigations that may justify further translational research.
Collapse
Affiliation(s)
- Luciana Aparecida Campos
- Center of Innovation, Technology, and Education (CITE) at Anhembi Morumbi University—Anima Institute, Sao Jose dos Campos Technology Park, Sao Jose dos Campos 12247-016, Brazil
- Department of Public Health and Epidemiology, College of Medicine and Health Science, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Correspondence: (L.A.C.); (O.C.B.)
| | - Ovidiu Constantin Baltatu
- Center of Innovation, Technology, and Education (CITE) at Anhembi Morumbi University—Anima Institute, Sao Jose dos Campos Technology Park, Sao Jose dos Campos 12247-016, Brazil
- Department of Public Health and Epidemiology, College of Medicine and Health Science, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Correspondence: (L.A.C.); (O.C.B.)
| | | | - Rym Ghimouz
- Fatima College of Health Sciences, Abu Dhabi P.O. Box 3798, United Arab Emirates
| | - Eman Alefishat
- Department of Pharmacology, College of Medicine and Health Science, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, The University of Jordan, Amman 11942, Jordan
- Center for Biotechnology, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - José Cipolla-Neto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
134
|
Lake AM, Goleva SB, Samuels LR, Carpenter LM, Davis LK. Sex Differences in Health Conditions Associated with Sexual Assault in a Large Hospital Population. Complex Psychiatry 2023; 8:80-89. [PMID: 36660008 PMCID: PMC10288064 DOI: 10.1159/000527363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/25/2022] [Indexed: 01/21/2023] Open
Abstract
Introduction Sexual assault is an urgent public health concern with both immediate and long-lasting health consequences, affecting 44% of women and 25% of men during their lifetimes. Large studies are needed to understand the unique healthcare needs of this patient population. Methods We mined clinical notes to identify patients with a history of sexual assault in the electronic health record (EHR) at Vanderbilt University Medical Center (VUMC), a large university hospital in the Southeastern USA, from 1989 to 2021 (N = 3,376,424). Using a phenome-wide case-control study, we identified diagnoses co-occurring with disclosures of sexual assault. We performed interaction tests to examine whether sex modified any of these associations. Association analyses were restricted to a subset of patients receiving regular care at VUMC (N = 833,185). Results The phenotyping approach identified 14,496 individuals (0.43%) across the VUMC-EHR with documentation of sexual assault and achieved a positive predictive value of 93.0% (95% confidence interval = 85.6-97.0%), determined by manual patient chart review. Out of 1,703 clinical diagnoses tested across all subgroup analyses, 465 were associated with sexual assault. Sex-by-trauma interaction analysis revealed 55 sex-differential associations and demonstrated increased odds of psychiatric diagnoses in male survivors. Discussion This case-control study identified associations between disclosures of sexual assault and hundreds of health conditions, many of which demonstrated sex-differential effects. The findings of this study suggest that patients who have experienced sexual assault are at risk for developing wide-ranging medical and psychiatric comorbidities and that male survivors may be particularly vulnerable to developing mental illness.
Collapse
Affiliation(s)
- Allison M. Lake
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Slavina B. Goleva
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Lauren R. Samuels
- Department of Biostatistics, Vanderbilt University, Nashville, Tennessee, USA
| | - Laura M. Carpenter
- Department of Sociology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Lea K. Davis
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
135
|
|
136
|
Hellwege JN, Dorn C, Irvin MR, Limdi NA, Cimino J, Beasley TM, Tsao PS, Damrauer SM, Roden DM, Velez Edwards DR, Wei WQ, Edwards TL. Predictive models for abdominal aortic aneurysms using polygenic scores and PheWAS-derived risk factors. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2023; 28:425-436. [PMID: 36540997 PMCID: PMC9782709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Abdominal aortic aneurysms (AAA) are common enlargements of the abdominal aorta which can grow larger until rupture, often leading to death. Detection of AAA is often by ultrasonography and screening recommendations are mostly directed at men over 65 with a smoking history. Recent large-scale genome-wide association studies have identified genetic loci associated with AAA risk. We combined known risk factors, polygenic risk scores (PRS) and precedent clinical diagnoses from electronic health records (EHR) to develop predictive models for AAA, and compared performance against screening recommendations. The PRS included genome-wide summary statistics from the Million Veteran Program and FinnGen (10,467 cases, 378,713 controls of European ancestry), with optimization in Vanderbilt's BioVU and validated in the eMERGE Network, separately across both White and Black participants. Candidate diagnoses were identified through a temporally-oriented Phenome-wide association study in independent EHR data from Vanderbilt, and features were selected via elastic net. We calculated C-statistics in eMERGE for models including PRS, phecodes, and covariates using regression weights from BioVU. The AUC for the full model in the test set was 0.883 (95% CI 0.873-0.892), 0.844 (0.836-0.851) for covariates only, 0.613 (95% CI 0.604-0.622) when using primary USPSTF screening criteria, and 0.632 (95% CI 0.623-0.642) using primary and secondary criteria. Brier scores were between 0.003 and 0.023 for our models indicating good calibration, and net reclassification improvement over combined primary and secondary USPSTF criteria was 0.36-0.60. We provide PRS for AAA which are strongly associated with AAA risk and add to predictive model performance. These models substantially improve identification of people at risk of a AAA diagnosis compared with existing guidelines, with evidence of potential applicability in minority populations.
Collapse
Affiliation(s)
- Jacklyn N Hellwege
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute Vanderbilt University Medical Center 2525 West End Ave. Ste 700, Nashville, TN, 37203, USA,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Joo YY, Pacheco JA, Thompson WK, Rasmussen-Torvik LJ, Rasmussen LV, Lin FTJ, Andrade MD, Borthwick KM, Bottinger E, Cagan A, Carrell DS, Denny JC, Ellis SB, Gottesman O, Linneman JG, Pathak J, Peissig PL, Shang N, Tromp G, Veerappan A, Smith ME, Chisholm RL, Gawron AJ, Hayes MG, Kho AN. Multi-ancestry genome- and phenome-wide association studies of diverticular disease in electronic health records with natural language processing enriched phenotyping algorithm. PLoS One 2023; 18:e0283553. [PMID: 37196047 DOI: 10.1371/journal.pone.0283553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 03/09/2023] [Indexed: 05/19/2023] Open
Abstract
OBJECTIVE Diverticular disease (DD) is one of the most prevalent conditions encountered by gastroenterologists, affecting ~50% of Americans before the age of 60. Our aim was to identify genetic risk variants and clinical phenotypes associated with DD, leveraging multiple electronic health record (EHR) data sources of 91,166 multi-ancestry participants with a Natural Language Processing (NLP) technique. MATERIALS AND METHODS We developed a NLP-enriched phenotyping algorithm that incorporated colonoscopy or abdominal imaging reports to identify patients with diverticulosis and diverticulitis from multicenter EHRs. We performed genome-wide association studies (GWAS) of DD in European, African and multi-ancestry participants, followed by phenome-wide association studies (PheWAS) of the risk variants to identify their potential comorbid/pleiotropic effects in clinical phenotypes. RESULTS Our developed algorithm showed a significant improvement in patient classification performance for DD analysis (algorithm PPVs ≥ 0.94), with up to a 3.5 fold increase in terms of the number of identified patients than the traditional method. Ancestry-stratified analyses of diverticulosis and diverticulitis of the identified subjects replicated the well-established associations between ARHGAP15 loci with DD, showing overall intensified GWAS signals in diverticulitis patients compared to diverticulosis patients. Our PheWAS analyses identified significant associations between the DD GWAS variants and circulatory system, genitourinary, and neoplastic EHR phenotypes. DISCUSSION As the first multi-ancestry GWAS-PheWAS study, we showcased that heterogenous EHR data can be mapped through an integrative analytical pipeline and reveal significant genotype-phenotype associations with clinical interpretation. CONCLUSION A systematic framework to process unstructured EHR data with NLP could advance a deep and scalable phenotyping for better patient identification and facilitate etiological investigation of a disease with multilayered data.
Collapse
Affiliation(s)
- Yoonjung Yoonie Joo
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Jennifer A Pacheco
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - William K Thompson
- Center for Health Information Partnerships, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Laura J Rasmussen-Torvik
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Luke V Rasmussen
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Frederick T J Lin
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Mariza de Andrade
- College of Medicine, Mayo Clinic, Rochester, MN, United States of America
| | | | - Erwin Bottinger
- Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Andrew Cagan
- Partners Healthcare, Charlestown, MA, United States of America
| | - David S Carrell
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington, United States of America
| | - Joshua C Denny
- Departments of Biomedical Informatics and Medicine, Vanderbilt University, Nashville, TN, United States of America
| | - Stephen B Ellis
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Omri Gottesman
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - James G Linneman
- Office of Research Computing and Analytics, Marshfield Clinic Research Institute, Marshfield, WI, United States of America
| | - Jyotishman Pathak
- Department of Healthcare Policy and Research, Weill Cornell Medical College, New York, NY, United States of America
| | - Peggy L Peissig
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, Marshfield, WI, United States of America
| | - Ning Shang
- Department of Biomedical Informatics, Columbia University, New York, NY, United States of America
| | - Gerard Tromp
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Annapoorani Veerappan
- Department of Medicine, Gastroenterology, Duke University, Durham, NC, United States of America
| | - Maureen E Smith
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Rex L Chisholm
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| | - Andrew J Gawron
- Division of Gastroenterology, Hepatology & Nutrition, University of Utah, Salt Lake City, UT, United States of America
| | - M Geoffrey Hayes
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
- Department of Anthropology, Northwestern University, Evanston, IL, United States of America
| | - Abel N Kho
- Center for Health Information Partnerships, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
- Division of General Internal Medicine and Geriatrics, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America
| |
Collapse
|
138
|
Bai H, Zhang X, Bush WS. Pharmacogenomic and Statistical Analysis. Methods Mol Biol 2023; 2629:305-330. [PMID: 36929083 DOI: 10.1007/978-1-0716-2986-4_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Genetic variants can alter response to drugs and other therapeutic interventions. The study of this phenomenon, called pharmacogenomics, is similar in many ways to other types of genetic studies but has distinct methodological and statistical considerations. Genetic variants involved in the processing of exogenous compounds exhibit great diversity and complexity, and the phenotypes studied in pharmacogenomics are also more complex than typical genetic studies. In this chapter, we review basic concepts in pharmacogenomic study designs, data generation techniques, statistical analysis approaches, and commonly used methods and briefly discuss the ultimate translation of findings to clinical care.
Collapse
Affiliation(s)
- Haimeng Bai
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
- Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Xueyi Zhang
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - William S Bush
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
139
|
Park J, MacLean MT, Lucas AM, Torigian DA, Schneider CV, Cherlin T, Xiao B, Miller JE, Bradford Y, Judy RL, Verma A, Damrauer SM, Ritchie MD, Witschey WR, Rader DJ. Exome-wide association analysis of CT imaging-derived hepatic fat in a medical biobank. Cell Rep Med 2022; 3:100855. [PMID: 36513072 PMCID: PMC9798024 DOI: 10.1016/j.xcrm.2022.100855] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 08/22/2022] [Accepted: 11/17/2022] [Indexed: 12/14/2022]
Abstract
Nonalcoholic fatty liver disease is common and highly heritable. Genetic studies of hepatic fat have not sufficiently addressed non-European and rare variants. In a medical biobank, we quantitate hepatic fat from clinical computed tomography (CT) scans via deep learning in 10,283 participants with whole-exome sequences available. We conduct exome-wide associations of single variants and rare predicted loss-of-function (pLOF) variants with CT-based hepatic fat and perform cross-modality replication in the UK Biobank (UKB) by linking whole-exome sequences to MRI-based hepatic fat. We confirm single variants previously associated with hepatic fat and identify several additional variants, including two (FGD5 H600Y and CITED2 S198_G199del) that replicated in UKB. A burden of rare pLOF variants in LMF2 is associated with increased hepatic fat and replicates in UKB. Quantitative phenotypes generated from clinical imaging studies and intersected with genomic data in medical biobanks have the potential to identify molecular pathways associated with human traits and disease.
Collapse
Affiliation(s)
- Joseph Park
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew T MacLean
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anastasia M Lucas
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Drew A Torigian
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carolin V Schneider
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tess Cherlin
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Brenda Xiao
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jason E Miller
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuki Bradford
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Renae L Judy
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anurag Verma
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Scott M Damrauer
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Surgery, Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Marylyn D Ritchie
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Walter R Witschey
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel J Rader
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
140
|
Fang Y, Fritsche LG, Mukherjee B, Sen S, Richmond-Rakerd LS. Polygenic Liability to Depression Is Associated With Multiple Medical Conditions in the Electronic Health Record: Phenome-wide Association Study of 46,782 Individuals. Biol Psychiatry 2022; 92:923-931. [PMID: 35965108 PMCID: PMC10712651 DOI: 10.1016/j.biopsych.2022.06.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 04/01/2022] [Accepted: 06/02/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Major depressive disorder (MDD) is a leading cause of disease-associated disability, with much of the increased burden due to psychiatric and medical comorbidity. This comorbidity partly reflects common genetic influences across conditions. Integrating molecular-genetic tools with health records enables tests of association with the broad range of physiological and clinical phenotypes. However, standard phenome-wide association studies analyze associations with individual genetic variants. For polygenic traits such as MDD, aggregate measures of genetic risk may yield greater insight into associations across the clinical phenome. METHODS We tested for associations between a genome-wide polygenic risk score for MDD and medical and psychiatric traits in a phenome-wide association study of 46,782 unrelated, European-ancestry participants from the Michigan Genomics Initiative. RESULTS The MDD polygenic risk score was associated with 211 traits from 15 medical and psychiatric disease categories at the phenome-wide significance threshold. After excluding patients with depression, continued associations were observed with respiratory, digestive, neurological, and genitourinary conditions; neoplasms; and mental disorders. Associations with tobacco use disorder, respiratory conditions, and genitourinary conditions persisted after accounting for genetic overlap between depression and other psychiatric traits. Temporal analyses of time-at-first-diagnosis indicated that depression disproportionately preceded chronic pain and substance-related disorders, while asthma disproportionately preceded depression. CONCLUSIONS The present results can inform the biological links between depression and both mental and systemic diseases. Although MDD polygenic risk scores cannot currently forecast health outcomes with precision at the individual level, as molecular-genetic discoveries for depression increase, these tools may augment risk prediction for medical and psychiatric conditions.
Collapse
Affiliation(s)
- Yu Fang
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, Michigan.
| | - Lars G Fritsche
- Department of Biostatistics, School of Public Health, University of Michigan Medicine, Ann Arbor, Michigan; Rogel Cancer Center, University of Michigan Medicine, Ann Arbor, Michigan; Center for Statistical Genetics, School of Public Health, University of Michigan Medicine, Ann Arbor, Michigan
| | - Bhramar Mukherjee
- Department of Biostatistics, School of Public Health, University of Michigan Medicine, Ann Arbor, Michigan; Rogel Cancer Center, University of Michigan Medicine, Ann Arbor, Michigan; Center for Statistical Genetics, School of Public Health, University of Michigan Medicine, Ann Arbor, Michigan; Department of Epidemiology, School of Public Health, University of Michigan Medicine, Ann Arbor, Michigan
| | - Srijan Sen
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, Michigan; Department of Psychiatry, University of Michigan Medicine, Ann Arbor, Michigan
| | | |
Collapse
|
141
|
Yuan S, Wang L, Sun J, Yu L, Zhou X, Yang J, Zhu Y, Gill D, Burgess S, Denny JC, Larsson SC, Theodoratou E, Li X. Genetically predicted sex hormone levels and health outcomes: phenome-wide Mendelian randomization investigation. Int J Epidemiol 2022; 51:1931-1942. [PMID: 35218343 PMCID: PMC9749729 DOI: 10.1093/ije/dyac036] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 02/10/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Sex hormone-binding globulin (SHBG), testosterone and oestradiol have been associated with many diseases in observational studies; however, the causality of associations remains unestablished. METHODS A phenome-wide Mendelian randomization (MR) association study was performed to explore disease outcomes associated with genetically proxied circulating SHBG, testosterone and oestradiol levels by using updated genetic instruments in 339 197 unrelated White British individuals (54% female) in the UK Biobank. Two-sample MR analyses with data from large genetic studies were conducted to replicate identified associations in phenome-wide MR analyses. Multivariable MR analyses were performed to investigate mediation effects of hormone-related biomarkers in observed associations with diseases. RESULTS Phenome-wide MR analyses examined associations of genetically predicted SHBG, testosterone and oestradiol levels with 1211 disease outcomes, and identified 28 and 13 distinct phenotypes associated with genetically predicted SHBG and testosterone, respectively; 22 out of 28 associations for SHBG and 10 out of 13 associations for testosterone were replicated in two-sample MR analyses. Higher genetically predicted SHBG levels were associated with a reduced risk of hypertension, type 2 diabetes, diabetic complications, coronary atherosclerotic outcomes, gout and benign and malignant neoplasm of uterus, but an increased risk of varicose veins and fracture (mainly in females). Higher genetically predicted testosterone levels were associated with a lower risk of type 2 diabetes, coronary atherosclerotic outcomes, gout and coeliac disease mainly in males, but an increased risk of cholelithiasis in females. CONCLUSIONS These findings suggest that sex hormones may causally affect risk of several health outcomes.
Collapse
Affiliation(s)
- Shuai Yuan
- Department of Big Data in Health Science, Center of Clinical Big Data and Analytics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lijuan Wang
- Department of Big Data in Health Science, Center of Clinical Big Data and Analytics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Sun
- Department of Big Data in Health Science, Center of Clinical Big Data and Analytics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lili Yu
- Department of Big Data in Health Science, Center of Clinical Big Data and Analytics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuan Zhou
- Department of Big Data in Health Science, Center of Clinical Big Data and Analytics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Yang
- Department of Big Data in Health Science, Center of Clinical Big Data and Analytics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yimin Zhu
- Department of Big Data in Health Science, Center of Clinical Big Data and Analytics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Stephen Burgess
- MRC Biostatistics Unit, Cambridge Institute of Public Health, Cambridge, UK
| | - Joshua C Denny
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Susanna C Larsson
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Xue Li
- Corresponding author. School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. E-mail:
| |
Collapse
|
142
|
Larsson SC, Wang L, Li X, Jiang F, Chen X, Mantzoros CS. Circulating lipoprotein(a) levels and health outcomes: Phenome-wide Mendelian randomization and disease-trajectory analyses. Metabolism 2022; 137:155347. [PMID: 36396079 DOI: 10.1016/j.metabol.2022.155347] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Lipoprotein(a) [Lp(a)] is a risk factor for atherosclerotic and valvular diseases, but its possible role in other diseases has not yet been established. We conducted phenome-wide Mendelian randomization and disease-trajectory analyses to assess any associations of circulating Lp(a) levels with a broad range of diseases. METHODS A weighted polygenic risk score was constructed using independent genetic variants in the LPA gene and with an established effect on Lp(a) levels. The PheWAS analysis included 1081 phenotype outcomes ascertained among 385,917 White participants of the UK Biobank. Novel findings were investigated in MR analysis using data from the FinnGen consortium. Disease-trajectory and comorbidity analyses were further conducted to explore the sequential patterns of multiple morbidities related to high circulating Lp(a) levels. RESULTS PheWAS revealed statistically significant associations of higher circulating Lp(a) levels with increased risk of a large number of circulatory system diseases (including various cardiac diseases, peripheral vascular disease, hypertension, and valvular and cerebrovascular diseases) as well as some endocrine/metabolic diseases (including hyperlipidemia, hypercholesterolemia, disorders of lipoid metabolism, and type 2 diabetes), genitourinary system diseases (renal failure), and hematologic diseases (including different types of anemia). Two-sample MR analysis supported the association between Lp(a) and risk of anemia, showed a suggestive association with type 2 diabetes, but found no association with renal failure. Disease-trajectory and comorbidity analyses identified 3 major sequential patterns of multiple morbidities, mainly in the cardiovascular, metabolic, and mental disorders, related to high circulating Lp(a) levels. CONCLUSIONS Genetically predicted higher circulating Lp(a) levels were associated with increased risk of many circulatory system diseases and anemia. Additionally, this study identified three major sequential patterns of multiple morbidities related to high Lp(a).
Collapse
Affiliation(s)
- Susanna C Larsson
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden; Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lijuan Wang
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xue Li
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Fangyuan Jiang
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiangjun Chen
- Eye Center of the Second Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Section of Endocrinology, Boston VA Healthcare System, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
143
|
Haupert SR, Shi X, Chen C, Fritsche LG, Mukherjee B. A Case-Crossover Phenome-wide association study (PheWAS) for understanding Post-COVID-19 diagnosis patterns. J Biomed Inform 2022; 136:104237. [PMID: 36283580 PMCID: PMC9595430 DOI: 10.1016/j.jbi.2022.104237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/30/2022] [Accepted: 10/19/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Post COVID-19 condition (PCC) is known to affect a large proportion of COVID-19 survivors. Robust study design and methods are needed to understand post-COVID-19 diagnosis patterns in all survivors, not just those clinically diagnosed with PCC. METHODS We applied a case-crossover Phenome-Wide Association Study (PheWAS) in a retrospective cohort of COVID-19 survivors, comparing the occurrences of 1,671 diagnosis-based phenotype codes (PheCodes) pre- and post-COVID-19 infection periods in the same individual using a conditional logistic regression. We studied how this pattern varied by COVID-19 severity and vaccination status, and we compared to test negative and test negative but flu positive controls. RESULTS In 44,198 SARS-CoV-2-positive patients, we foundenrichment in respiratory,circulatory, and mental health disorders post-COVID-19-infection. Top hits included anxiety disorder (p = 2.8e-109, OR = 1.7 [95 % CI: 1.6-1.8]), cardiac dysrhythmias (p = 4.9e-87, OR = 1.7 [95 % CI: 1.6-1.8]), and respiratory failure, insufficiency, arrest (p = 5.2e-75, OR = 2.9 [95 % CI: 2.6-3.3]). In severe patients, we found stronger associations with respiratory and circulatory disorders compared to mild/moderate patients. Fully vaccinated patients had mental health and chronic circulatory diseases rise to the top of the association list, similar to the mild/moderate cohort. Both control groups (test negative, test negative and flu positive) showed a different pattern of hits to SARS-CoV-2 positives. CONCLUSIONS Patients experience myriad symptoms more than 28 days after SARS-CoV-2 infection, but especially respiratory, circulatory, and mental health disorders. Our case-crossover PheWAS approach controls for within-person confounders that are time-invariant. Comparison to test negatives and test negative but flu positive patients with a similar design helped identify enrichment specific to COVID-19. This design may be applied other emerging diseases with long-lasting effects other than a SARS-CoV-2 infection. Given the potential for bias from observational data, these results should be considered exploratory. As we look into the future, we must be aware of COVID-19 survivors' healthcare needs.
Collapse
Affiliation(s)
- Spencer R Haupert
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Xu Shi
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Chen Chen
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Lars G Fritsche
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; Center for Precision Health Data Science, University of Michigan, Ann Arbor, MI 48109, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Bhramar Mukherjee
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; Center for Precision Health Data Science, University of Michigan, Ann Arbor, MI 48109, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA; Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA.
| |
Collapse
|
144
|
Robinson JR, Carroll RJ, Bastarache L, Chen Q, Pirruccello J, Mou Z, Wei WQ, Connolly J, Mentch F, Crane PK, Hebbring SJ, Crosslin DR, Gordon AS, Rosenthal EA, Stanaway IB, Hayes MG, Wei W, Petukhova L, Namjou-Khales B, Zhang G, Safarova MS, Walton NA, Still C, Bottinger EP, Loos RJF, Murphy SN, Jackson GP, Abumrad N, Kullo IJ, Jarvik GP, Larson EB, Weng C, Roden D, Khera AV, Denny JC. Quantifying the phenome-wide disease burden of obesity using electronic health records and genomics. Obesity (Silver Spring) 2022; 30:2477-2488. [PMID: 36372681 PMCID: PMC9691570 DOI: 10.1002/oby.23561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVE High BMI is associated with many comorbidities and mortality. This study aimed to elucidate the overall clinical risk of obesity using a genome- and phenome-wide approach. METHODS This study performed a phenome-wide association study of BMI using a clinical cohort of 736,726 adults. This was followed by genetic association studies using two separate cohorts: one consisting of 65,174 adults in the Electronic Medical Records and Genomics (eMERGE) Network and another with 405,432 participants in the UK Biobank. RESULTS Class 3 obesity was associated with 433 phenotypes, representing 59.3% of all billing codes in individuals with severe obesity. A genome-wide polygenic risk score for BMI, accounting for 7.5% of variance in BMI, was associated with 296 clinical diseases, including strong associations with type 2 diabetes, sleep apnea, hypertension, and chronic liver disease. In all three cohorts, 199 phenotypes were associated with class 3 obesity and polygenic risk for obesity, including novel associations such as increased risk of renal failure, venous insufficiency, and gastroesophageal reflux. CONCLUSIONS This combined genomic and phenomic systematic approach demonstrated that obesity has a strong genetic predisposition and is associated with a considerable burden of disease across all disease classes.
Collapse
Affiliation(s)
- Jamie R. Robinson
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Robert J. Carroll
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lisa Bastarache
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Qingxia Chen
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James Pirruccello
- Center for Genomics Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Zongyang Mou
- Department of Surgery, University of California, San Diego, CA, USA
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John Connolly
- The Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Frank Mentch
- The Center for Applied Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Paul K. Crane
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Scott J. Hebbring
- Center for Human Genetics, Marshfield Clinic Research Institute, Marshfield, WI, USA
| | - David R. Crosslin
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA, USA
| | - Adam S. Gordon
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Elisabeth A. Rosenthal
- Departments of Medicine (Medical Genetics) and Genome Sciences, University of Washington Medical Center, Seattle, WA, USA
| | - Ian B. Stanaway
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA, USA
| | - M. Geoffrey Hayes
- Division of Endocrinology, Metabolism, and Molecular Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Wei Wei
- University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Lynn Petukhova
- Department of Epidemiology, Columbia University, New York, NY, USA
| | - Bahram Namjou-Khales
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Ge Zhang
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Mayya S. Safarova
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Nephi A. Walton
- Department of Biomedical and Translational Informatics, Geisinger Health System, Danville, PA, USA
| | - Christopher Still
- Department of Biomedical and Translational Informatics, Geisinger Health System, Danville, PA, USA
| | - Erwin P. Bottinger
- The Charles Bronfman Institute for Personalized Medicine at Mount Sinai, The Mindich Child Health and Development Institute, New York, NY, USA
| | - Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized Medicine at Mount Sinai, The Mindich Child Health and Development Institute, New York, NY, USA
| | - Shawn N. Murphy
- Department of Neurology, Partners Healthcare, Boston, MA, USA
| | - Gretchen P. Jackson
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Naji Abumrad
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Iftikhar J. Kullo
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Gail P. Jarvik
- Departments of Medicine (Medical Genetics) and Genome Sciences, University of Washington Medical Center, Seattle, WA, USA
| | - Eric B. Larson
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Chunhua Weng
- Department of Biomedical Informatics, Columbia University, New York, NY, USA
| | - Dan Roden
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Amit V. Khera
- Center for Genomics Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joshua C. Denny
- All of Us Research Program, National Institutes of Health, Bethesda, MD
| |
Collapse
|
145
|
Zagkos L, Dib MJ, Pinto R, Gill D, Koskeridis F, Drenos F, Markozannes G, Elliott P, Zuber V, Tsilidis K, Dehghan A, Tzoulaki I. Associations of genetically predicted fatty acid levels across the phenome: A mendelian randomisation study. PLoS Med 2022; 19:e1004141. [PMID: 36580444 PMCID: PMC9799317 DOI: 10.1371/journal.pmed.1004141] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 11/18/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Fatty acids are important dietary factors that have been extensively studied for their implication in health and disease. Evidence from epidemiological studies and randomised controlled trials on their role in cardiovascular, inflammatory, and other diseases remains inconsistent. The objective of this study was to assess whether genetically predicted fatty acid concentrations affect the risk of disease across a wide variety of clinical health outcomes. METHODS AND FINDINGS The UK Biobank (UKB) is a large study involving over 500,000 participants aged 40 to 69 years at recruitment from 2006 to 2010. We used summary-level data for 117,143 UKB samples (base dataset), to extract genetic associations of fatty acids, and individual-level data for 322,232 UKB participants (target dataset) to conduct our discovery analysis. We studied potentially causal relationships of circulating fatty acids with 845 clinical diagnoses, using mendelian randomisation (MR) approach, within a phenome-wide association study (PheWAS) framework. Regression models in PheWAS were adjusted for sex, age, and the first 10 genetic principal components. External summary statistics were used for replication. When several fatty acids were associated with a health outcome, multivariable MR and MR-Bayesian method averaging (MR-BMA) was applied to disentangle their causal role. Genetic predisposition to higher docosahexaenoic acid (DHA) was associated with cholelithiasis and cholecystitis (odds ratio per mmol/L: 0.76, 95% confidence interval: 0.66 to 0.87). This was supported in replication analysis (FinnGen study) and by the genetically predicted omega-3 fatty acids analyses. Genetically predicted linoleic acid (LA), omega-6, polyunsaturated fatty acids (PUFAs), and total fatty acids (total FAs) showed positive associations with cardiovascular outcomes with support from replication analysis. Finally, higher genetically predicted levels of DHA (0.83, 0.73 to 0.95) and omega-3 (0.83, 0.75 to 0.92) were found to have a protective effect on obesity, which was supported using body mass index (BMI) in the GIANT consortium as replication analysis. Multivariable MR analysis suggested a direct detrimental effect of LA (1.64, 1.07 to 2.50) and omega-6 fatty acids (1.81, 1.06 to 3.09) on coronary heart disease (CHD). MR-BMA prioritised LA and omega-6 fatty acids as the top risk factors for CHD. Although we present a range of sensitivity analyses to the address MR assumptions, horizontal pleiotropy may still bias the reported associations and further evaluation in clinical trials is needed. CONCLUSIONS Our study suggests potentially protective effects of circulating DHA and omega-3 concentrations on cholelithiasis and cholecystitis and on obesity, highlighting the need to further assess them as prevention treatments in clinical trials. Moreover, our findings do not support the supplementation of unsaturated fatty acids for cardiovascular disease prevention.
Collapse
Affiliation(s)
- Loukas Zagkos
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Marie-Joe Dib
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Rui Pinto
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- UK Dementia Research Institute, Imperial College London, London, United Kingdom
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- Chief Scientific Advisor Office, Research and Early Development, Novo Nordisk, Copenhagen, Denmark
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom
| | - Fotios Koskeridis
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Fotios Drenos
- Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
- Institute of Cardiovascular Sciences, University College London, London, United Kingdom
| | - Georgios Markozannes
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- UK Dementia Research Institute, Imperial College London, London, United Kingdom
- BHF Centre of Excellence at Imperial College London, London, United Kingdom
| | - Verena Zuber
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- UK Dementia Research Institute, Imperial College London, London, United Kingdom
| | - Kostas Tsilidis
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- UK Dementia Research Institute, Imperial College London, London, United Kingdom
| | - Ioanna Tzoulaki
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- UK Dementia Research Institute, Imperial College London, London, United Kingdom
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
- BHF Centre of Excellence at Imperial College London, London, United Kingdom
| |
Collapse
|
146
|
Associations of Genetically Predicted Vitamin B 12 Status across the Phenome. Nutrients 2022; 14:nu14235031. [PMID: 36501061 PMCID: PMC9740080 DOI: 10.3390/nu14235031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/16/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Variation in vitamin B12 levels has been associated with a range of diseases across the life-course, the causal nature of which remains elusive. We aimed to interrogate genetically predicted vitamin B12 status in relation to a plethora of clinical outcomes available in the UK Biobank. Genome-wide association study (GWAS) summary data obtained from a Danish and Icelandic cohort of 45,576 individuals were used to identify 8 genetic variants associated with vitamin B12 levels, serving as genetic instruments for vitamin B12 status in subsequent analyses. We conducted a Mendelian randomisation (MR)-phenome-wide association study (PheWAS) of vitamin B12 status with 945 distinct phenotypes in 439,738 individuals from the UK Biobank using these 8 genetic instruments to proxy alterations in vitamin B12 status. We used external GWAS summary statistics for replication of significant findings. Correction for multiple testing was taken into consideration using a 5% false discovery rate (FDR) threshold. MR analysis identified an association between higher genetically predicted vitamin B12 status and lower risk of vitamin B deficiency (including all B vitamin deficiencies), serving as a positive control outcome. We further identified associations between higher genetically predicted vitamin B12 status and a reduced risk of megaloblastic anaemia (OR = 0.35, 95% CI: 0.20-0.50) and pernicious anaemia (0.29, 0.19-0.45), which was supported in replication analyses. Our study highlights that higher genetically predicted vitamin B12 status is potentially protective of risk of vitamin B12 deficiency associated with pernicious anaemia diagnosis, and reduces risk of megaloblastic anaemia. The potential use of genetically predicted vitamin B12 status in disease diagnosis, progression and management remains to be investigated.
Collapse
|
147
|
Yuan S, Sun J, Lu Y, Xu F, Li D, Jiang F, Wan Z, Li X, Qin LQ, Larsson SC. Health effects of milk consumption: phenome-wide Mendelian randomization study. BMC Med 2022; 20:455. [PMID: 36424608 PMCID: PMC9694907 DOI: 10.1186/s12916-022-02658-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 11/09/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND We performed phenome-wide Mendelian randomization analysis (MR-PheWAS), two-sample MR analysis, and systemic review to comprehensively explore the health effects of milk consumption in the European population. METHODS Rs4988235 located upstream of the LCT gene was used as the instrumental variable for milk consumption. MR-PheWAS analysis was conducted to map the association of genetically predicted milk consumption with 1081 phenotypes in the UK Biobank study (n=339,197). The associations identified in MR-PheWAS were examined by two-sample MR analysis using data from the FinnGen study (n=260,405) and international consortia. A systematic review of MR studies on milk consumption was further performed. RESULTS PheWAS and two-sample MR analyses found robust evidence in support of inverse associations of genetically predicted milk consumption with risk of cataract (odds ratio (OR) per 50 g/day increase in milk consumption, 0.89, 95% confidence interval (CI), 0.84-0.94; p=3.81×10-5), hypercholesterolemia (OR, 0.91, 95% CI 0.86-0.96; p=2.97×10-4), and anal and rectal polyps (OR, 0.85, 95% CI, 0.77-0.94; p=0.001). An inverse association for type 2 diabetes risk (OR, 0.92, 95% CI, 0.86-0.97; p=0.003) was observed in MR analysis based on genetic data with body mass index adjustment but not in the corresponding data without body mass index adjustment. The systematic review additionally found evidence that genetically predicted milk consumption was inversely associated with asthma, hay fever, multiple sclerosis, colorectal cancer, and Alzheimer's disease, and positively associated with Parkinson's disease, renal cell carcinoma, metabolic syndrome, overweight, and obesity. CONCLUSIONS This study suggests several health effects of milk consumption in the European population.
Collapse
Affiliation(s)
- Shuai Yuan
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jing Sun
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Lu
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fengzhe Xu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China.,Westlake Intelligent Biomarker Discovery Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Doudou Li
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fangyuan Jiang
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhongxiao Wan
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, China
| | - Xue Li
- Department of Big Data in Health Science School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Li-Qiang Qin
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, China.
| | - Susanna C Larsson
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden. .,Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
148
|
Fritsche LG, Jin W, Admon AJ, Mukherjee B. Characterizing and Predicting Post-Acute Sequelae of SARS CoV-2 infection (PASC) in a Large Academic Medical Center in the US. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.10.21.22281356. [PMID: 36415469 PMCID: PMC9681058 DOI: 10.1101/2022.10.21.22281356] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Objective A growing number of Coronavirus Disease-2019 (COVID-19) survivors are affected by Post-Acute Sequelae of SARS CoV-2 infection (PACS). Using electronic health records data, we aimed to characterize PASC-associated diagnoses and to develop risk prediction models. Methods In our cohort of 63,675 COVID-19 positive patients, 1,724 (2.7 %) had a recorded PASC diagnosis. We used a case control study design and phenome-wide scans to characterize PASC-associated phenotypes of the pre-, acute-, and post-COVID-19 periods. We also integrated PASC-associated phenotypes into Phenotype Risk Scores (PheRSs) and evaluated their predictive performance. Results In the post-COVID-19 period, known PASC symptoms (e.g., shortness of breath, malaise/fatigue) and musculoskeletal, infectious, and digestive disorders were enriched among PASC cases. We found seven phenotypes in the pre-COVID-19 period (e.g., irritable bowel syndrome, concussion, nausea/vomiting) and 69 phenotypes in the acute-COVID-19 period (predominantly respiratory, circulatory, neurological) associated with PASC. The derived pre- and acute-COVID-19 PheRSs stratified risk well, e.g., the combined PheRSs identified a quarter of the COVID-19 positive cohort with an at least 2.9-fold increased risk for PASC. Conclusions The uncovered PASC-associated diagnoses across categories highlighted a complex arrangement of presenting and likely predisposing features, some with a potential for risk stratification approaches.
Collapse
Affiliation(s)
- Lars G. Fritsche
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan 48109, United States of America,Center for Precision Health Data Science, University of Michigan School of Public Health, Ann Arbor, Michigan 48109, United States of America,Correspondence: ,
| | - Weijia Jin
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan 48109, United States of America,Center for Precision Health Data Science, University of Michigan School of Public Health, Ann Arbor, Michigan 48109, United States of America
| | - Andrew J. Admon
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States of America,Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan 48109, United States of America,VA Center for Clinical Management Research, LTC Charles S. Kettles VA Medical Center, Ann Arbor, Michigan 48109, United States of America
| | - Bhramar Mukherjee
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan 48109, United States of America,Center for Precision Health Data Science, University of Michigan School of Public Health, Ann Arbor, Michigan 48109, United States of America,Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan 48109, United States of America,Michigan Institute for Data Science, University of Michigan, Ann Arbor, Michigan 48109, United States of America,Correspondence: ,
| |
Collapse
|
149
|
Mack T, Sanchez-Roige S, Davis LK. Genetic investigation of the contribution of body composition to anorexia nervosa in an electronic health record setting. Transl Psychiatry 2022; 12:486. [PMID: 36402754 PMCID: PMC9675730 DOI: 10.1038/s41398-022-02251-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 10/31/2022] [Accepted: 11/04/2022] [Indexed: 11/21/2022] Open
Abstract
Anorexia nervosa (AN) is a psychiatric disorder defined by anthropometric symptoms, such as low body weight, and cognitive-behavioral symptoms, such as restricted eating, fear of weight gain, and distorted body image. Recent studies have identified a genetic association between AN and metabolic/anthropometric factors, including body mass index (BMI). Although the reported associations may be under pleiotropic genetic influences, they may represent independent risk factors for AN. Here we examined the independent contributions of genetic predisposition to low body weight and polygenic risk (PRS) for AN in a clinical population (Vanderbilt University Medical Center biobank, BioVU). We fitted logistic and linear regression models in a retrospective case-control design (123 AN patients, 615 age-matched controls). We replicated the genetic correlations between PRSBMI and AN (p = 1.12 × 10-3, OR = 0.96), but this correlation disappeared when controlling for lowest BMI (p = 0.84, OR = 1.00). Additionally, we performed a phenome-wide association analysis of the PRSAN and found that the associations with metabolic phenotypes were attenuated when controlling for PRSBMI. These findings suggest that the genetic association between BMI and AN may be a consequence of the weight-related diagnostic criteria for AN and that genetically regulated anthropometric traits (like BMI) may be independent of AN psychopathology. If so, individuals with cognitive-behavioral symptomatology suggestive of AN, but with a higher PRSBMI, may be under-diagnosed given current diagnostic criteria. Furthermore, PRSBMI may serve as an independent risk factor for weight loss and weight gain during recovery.
Collapse
Affiliation(s)
- Taralynn Mack
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sandra Sanchez-Roige
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Psychiatry, University of California San Diego, La Jolla, CA, 92093, USA
| | - Lea K Davis
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
150
|
Koskeridis F, Evangelou E, Said S, Boyle JJ, Elliott P, Dehghan A, Tzoulaki I. Pleiotropic genetic architecture and novel loci for C-reactive protein levels. Nat Commun 2022; 13:6939. [PMID: 36376304 PMCID: PMC9663411 DOI: 10.1038/s41467-022-34688-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 11/02/2022] [Indexed: 11/16/2022] Open
Abstract
C-reactive protein is involved in a plethora of pathophysiological conditions. Many genetic loci associated with C-reactive protein are annotated to lipid and glucose metabolism genes supporting common biological pathways between inflammation and metabolic traits. To identify novel pleiotropic loci, we perform multi-trait analysis of genome-wide association studies on C-reactive protein levels along with cardiometabolic traits, followed by a series of in silico analyses including colocalization, phenome-wide association studies and Mendelian randomization. We find 41 novel loci and 19 gene sets associated with C-reactive protein with various pleiotropic effects. Additionally, 41 variants colocalize between C-reactive protein and cardiometabolic risk factors and 12 of them display unexpected discordant effects between the shared traits which are translated into discordant associations with clinical outcomes in subsequent phenome-wide association studies. Our findings provide insights into shared mechanisms underlying inflammation and lipid metabolism, representing potential preventive and therapeutic targets.
Collapse
Affiliation(s)
- Fotios Koskeridis
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece.
| | - Evangelos Evangelou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
- Institute of Biosciences, University Research Center of Ioannina, University of Ioannina, 45110, Ioannina, Greece
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Saredo Said
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Joseph J Boyle
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
- BHF Centre of Excellence, Imperial College London, London, UK
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
- BHF Centre of Excellence, Imperial College London, London, UK
| | - Ioanna Tzoulaki
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
- Institute of Biosciences, University Research Center of Ioannina, University of Ioannina, 45110, Ioannina, Greece
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
- BHF Centre of Excellence, Imperial College London, London, UK
| |
Collapse
|