51
|
Zhang S, Strayer N, Vessels T, Choi K, Wang GW, Li Y, Bejan CA, Hsi RS, Bick AG, Velez Edwards DR, Savona MR, Phillips EJ, Pulley JM, 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. J Am Med Inform Assoc 2024; 31:2440-2446. [PMID: 39127052 PMCID: PMC11491640 DOI: 10.1093/jamia/ocae182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 06/03/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
OBJECTIVES To address the need for interactive visualization tools and databases in characterizing multimorbidity patterns across different populations, we developed the Phenome-wide Multi-Institutional Multimorbidity Explorer (PheMIME). This tool leverages three large-scale EHR systems to facilitate efficient analysis and visualization of disease multimorbidity, aiming to reveal both robust and novel disease associations that are consistent across different systems and to provide insight for enhancing personalized healthcare strategies. MATERIALS AND METHODS PheMIME integrates summary statistics from phenome-wide analyses of disease multimorbidities, utilizing data from Vanderbilt University Medical Center, Mass General Brigham, and the UK Biobank. It offers interactive and multifaceted visualizations for exploring multimorbidity. Incorporating an enhanced version of associationSubgraphs, PheMIME also enables dynamic analysis and inference of disease clusters, promoting the discovery of complex multimorbidity patterns. A case study on schizophrenia demonstrates its capability for generating interactive visualizations of multimorbidity networks within and across multiple systems. Additionally, PheMIME supports diverse multimorbidity-based discoveries, detailed further in online case studies. RESULTS The PheMIME is accessible at https://prod.tbilab.org/PheMIME/. A comprehensive tutorial and multiple case studies for demonstration are available at https://prod.tbilab.org/PheMIME_supplementary_materials/. The source code can be downloaded from https://github.com/tbilab/PheMIME. DISCUSSION PheMIME represents a significant advancement in medical informatics, offering an efficient solution for accessing, analyzing, and interpreting the complex and noisy real-world patient data in electronic health records. CONCLUSION PheMIME provides an extensive multimorbidity knowledge base that consolidates data from three EHR systems, and it is a novel interactive tool designed to analyze and visualize multimorbidities across multiple EHR datasets. It stands out as the first of its kind to offer extensive multimorbidity knowledge integration with substantial support for efficient online analysis and interactive visualization.
Collapse
Affiliation(s)
- Siwei Zhang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37203, United States
| | | | - Tess Vessels
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Karmel Choi
- Psychiatric & Neuro Developmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, United States
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, United States
| | - Geoffrey W Wang
- Department of Statistics, North Carolina State University, Raleigh, NC 27695, United States
| | - Yajing Li
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37203, United States
| | - Cosmin A Bejan
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37203, United States
| | - Ryan S Hsi
- Department of Urology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Alexander G Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Digna R Velez Edwards
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Michael R Savona
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Elizabeth J Phillips
- Center for Drug Safety and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, WA 6150, Australia
| | - Jill M Pulley
- Vanderbilt Institute for Clinical and Translational Science, Vanderbilt University Medical Center, Nashville, TN 37203, United States
| | - Wesley H Self
- Vanderbilt Institute for Clinical and Translational Science, Vanderbilt University Medical Center, Nashville, TN 37203, United States
| | - Wilkins Consuelo Hopkins
- Vanderbilt Institute for Clinical and Translational Science, Vanderbilt University Medical Center, Nashville, TN 37203, United States
| | - Dan M Roden
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Jordan W Smoller
- Psychiatric & Neuro Developmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, United States
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, United States
- Stanley Center for Psychiatric Research, Broad Institute, Cambridge, MA 02142, United States
| | - Douglas M Ruderfer
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, United States
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37203, United States
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN 37212, United States
| | - Yaomin Xu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37203, United States
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37203, United States
| |
Collapse
|
52
|
Kelman G, Zucker R, Brandes N, Linial M. PWAS Hub for exploring gene-based associations of common complex diseases. Genome Res 2024; 34:1674-1686. [PMID: 39406500 PMCID: PMC11529988 DOI: 10.1101/gr.278916.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 08/30/2024] [Indexed: 11/01/2024]
Abstract
PWAS (proteome-wide association study) is an innovative genetic association approach that complements widely used methods like GWAS (genome-wide association study). The PWAS approach involves consecutive phases. Initially, machine learning modeling and probabilistic considerations quantify the impact of genetic variants on protein-coding genes' biochemical functions. Secondly, for each individual, aggregating the variants per gene determines a gene-damaging score. Finally, standard statistical tests are activated in the case-control setting to yield statistically significant genes per phenotype. The PWAS Hub offers a user-friendly interface for an in-depth exploration of gene-disease associations from the UK Biobank (UKB). Results from PWAS cover 99 common diseases and conditions, each with over 10,000 diagnosed individuals per phenotype. Users can explore genes associated with these diseases, with separate analyses conducted for males and females. For each phenotype, the analyses account for sex-based genetic effects, inheritance modes (dominant and recessive), and the pleiotropic nature of associated genes. The PWAS Hub showcases its usefulness for asthma by navigating through proteomic-genetic analyses. Inspecting PWAS asthma-listed genes (a total of 27) provide insights into the underlying cellular and molecular mechanisms. Comparison of PWAS-statistically significant genes for common diseases to the Open Targets benchmark shows partial but significant overlap in gene associations for most phenotypes. Graphical tools facilitate comparing genetic effects between PWAS and coding GWAS results, aiding in understanding the sex-specific genetic impact on common diseases. This adaptable platform is attractive to clinicians, researchers, and individuals interested in delving into gene-disease associations and sex-specific genetic effects.
Collapse
Affiliation(s)
- Guy Kelman
- The Jerusalem Center for Personalized Computational Medicine, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Roei Zucker
- The Rachel and Selim Benin School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Nadav Brandes
- Division of Rheumatology, Department of Medicine, University of California San Francisco, San Francisco, California 94143, USA
| | - Michal Linial
- Department of Biological Chemistry, Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| |
Collapse
|
53
|
Lovegrove CE, Howles SA, Furniss D, Holmes MV. Causal inference in health and disease: a review of the principles and applications of Mendelian randomization. J Bone Miner Res 2024; 39:1539-1552. [PMID: 39167758 PMCID: PMC11523132 DOI: 10.1093/jbmr/zjae136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/04/2024] [Accepted: 08/19/2024] [Indexed: 08/23/2024]
Abstract
Mendelian randomization (MR) is a genetic epidemiological technique that uses genetic variation to infer causal relationships between modifiable exposures and outcome variables. Conventional observational epidemiological studies are subject to bias from a range of sources; MR analyses can offer an advantage in that they are less prone to bias as they use genetic variants inherited at conception as "instrumental variables", which are proxies of an exposure. However, as with all research tools, MR studies must be carefully designed to yield valuable insights into causal relationships between exposures and outcomes, and to avoid biased or misleading results that undermine the validity of the causal inferences drawn from the study. In this review, we outline Mendel's laws of inheritance, the assumptions and principles that underlie MR, MR study designs and methods, and how MR analyses can be applied and reported. Using the example of serum phosphate concentrations on liability to kidney stone disease we illustrate how MR estimates may be visualized and, finally, we contextualize MR in bone and mineral research including exemplifying how this technique could be employed to inform clinical studies and future guidelines concerning BMD and fracture risk. This review provides a framework to enhance understanding of how MR may be used to triangulate evidence and progress research in bone and mineral metabolism as we strive to infer causal effects in health and disease.
Collapse
Affiliation(s)
- Catherine E Lovegrove
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, United Kingdom
| | - Sarah A Howles
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, United Kingdom
| | - Dominic Furniss
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, United Kingdom
| | - Michael V Holmes
- Medical Research Council, Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, United Kingdom
| |
Collapse
|
54
|
Mustafa R, Mens MMJ, van Hilten A, Huang J, Roshchupkin G, Huan T, Broer L, van Meurs JBJ, Elliott P, Levy D, Ikram MA, Evangelou M, Dehghan A, Ghanbari M. A comprehensive study of genetic regulation and disease associations of plasma circulatory microRNAs using population-level data. Genome Biol 2024; 25:276. [PMID: 39434104 PMCID: PMC11492503 DOI: 10.1186/s13059-024-03420-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/09/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are small non-coding RNAs that post-transcriptionally regulate gene expression. Perturbations in plasma miRNA levels are known to impact disease risk and have potential as disease biomarkers. Exploring the genetic regulation of miRNAs may yield new insights into their important role in governing gene expression and disease mechanisms. RESULTS We present genome-wide association studies of 2083 plasma circulating miRNAs in 2178 participants of the Rotterdam Study to identify miRNA-expression quantitative trait loci (miR-eQTLs). We identify 3292 associations between 1289 SNPs and 63 miRNAs, of which 65% are replicated in two independent cohorts. We demonstrate that plasma miR-eQTLs co-localise with gene expression, protein, and metabolite-QTLs, which help in identifying miRNA-regulated pathways. We investigate consequences of alteration in circulating miRNA levels on a wide range of clinical conditions in phenome-wide association studies and Mendelian randomisation using the UK Biobank data (N = 423,419), revealing the pleiotropic and causal effects of several miRNAs on various clinical conditions. In the Mendelian randomisation analysis, we find a protective causal effect of miR-1908-5p on the risk of benign colon neoplasm and show that this effect is independent of its host gene (FADS1). CONCLUSIONS This study enriches our understanding of the genetic architecture of plasma miRNAs and explores the signatures of miRNAs across a wide range of clinical conditions. The integration of population-based genomics, other omics layers, and clinical data presents opportunities to unravel potential clinical significance of miRNAs and provides tools for novel miRNA-based therapeutic target discovery.
Collapse
Affiliation(s)
- Rima Mustafa
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London, UK
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Michelle M J Mens
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Social and Behavorial Sciences, Harvard T.H Chan School of Public Health, Boston, MA, USA
| | - Arno van Hilten
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jian Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore
| | - Gennady Roshchupkin
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Tianxiao Huan
- Framingham Heart Study, Framingham, MA, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Linda Broer
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Joyce B J van Meurs
- Department of Internal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Orthopaedics and Sports Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - 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
- MRC Centre for Environment and Health, Imperial College London, London, UK
- Health Data Research (HDR) UK, Imperial College London, London, UK
- BHF Centre for Research Excellence, Imperial College London, London, UK
| | - Daniel Levy
- Framingham Heart Study, Framingham, MA, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - 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
- MRC Centre for Environment and Health, Imperial College London, London, UK
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
55
|
Shi X, Wang Y, Yang Z, Yuan W, Li MD. Identification and validation of a novel gene ARVCF associated with alcohol dependence among Chinese population. iScience 2024; 27:110976. [PMID: 39429782 PMCID: PMC11490727 DOI: 10.1016/j.isci.2024.110976] [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: 04/27/2024] [Revised: 08/24/2024] [Accepted: 09/13/2024] [Indexed: 10/22/2024] Open
Abstract
Alcohol dependence is a heritable disorder, yet its genetic basis and underlying mechanisms remain poorly understood, especially in Chinese population. In this study, we conducted gene-based and transcript-based association tests and found a significant association between ARVCF expression in the cortex and hippocampus of the brain and alcohol use in a cohort of 1,329 individuals with Chinese ancestry. Further analysis using the effective-median-based Mendelian randomization framework for inferring the causal genes (EMIC) revealed a causal relationship between ARVCF expression in the frontal cortex and alcohol use. Moreover, leveraging extensive European alcohol dependence data, our gene association tests and EMIC analysis showed that ARVCF expression in the nucleus accumbens was significantly associated with alcohol dependence. Finally, animal studies indicated that Arvcf knockout mice lacked conditioned place preference for alcohol. Together, our combined human genetic and animal studies indicate that ARVCF plays a crucial role in alcohol dependence.
Collapse
Affiliation(s)
- Xiaoqiang Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yan Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Zhongli Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Wenji Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Ming D. Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
- Research Center for Air Pollution and Health, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
56
|
Liu L, Zhu L, Monteiro-Martins S, Griffin A, Vlahos LJ, Fujita M, Berrouet C, Zanoni F, Marasa M, Zhang JY, Zhou XJ, Caliskan Y, Akchurin O, Al-Akash S, Jankauskiene A, Bodria M, Chishti A, Esposito C, Esposito V, Claes D, Tesar V, Davis TK, Samsonov D, Kaminska D, Hryszko T, Zaza G, Flynn JT, Iorember F, Lugani F, Rizk D, Julian BA, Hidalgo G, Kallash M, Biancone L, Amoroso A, Bono L, Mani LY, Vogt B, Lin F, Sreedharan R, Weng P, Ranch D, Xiao N, Quiroga A, Matar RB, Rheault MN, Wenderfer S, Selewski D, Lundberg S, Silva C, Mason S, Mahan JD, Vasylyeva TL, Mucha K, Foroncewicz B, Pączek L, Florczak M, Olszewska M, Gradzińska A, Szczepańska M, Machura E, Badeński A, Krakowczyk H, Sikora P, Kwella N, Miklaszewska M, Drożdż D, Zaniew M, Pawlaczyk K, SiniewiczLuzeńczyk K, Bomback AS, Appel GB, Izzi C, Scolari F, Materna-Kiryluk A, Mizerska-Wasiak M, Berthelot L, Pillebout E, Monteiro RC, Novak J, Green TJ, Smoyer WE, Hastings MC, Wyatt RJ, Nelson R, Martin J, González-Gay MA, De Jager PL, Köttgen A, Califano A, Gharavi AG, Zhang H, Kiryluk K. Genome-wide studies define new genetic mechanisms of IgA vasculitis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.10.24315041. [PMID: 39417133 PMCID: PMC11482997 DOI: 10.1101/2024.10.10.24315041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
IgA vasculitis (IgAV) is a pediatric disease with skin and systemic manifestations. Here, we conducted genome, transcriptome, and proteome-wide association studies in 2,170 IgAV cases and 5,928 controls, generated IgAV-specific maps of gene expression and splicing from blood of 255 pediatric cases, and reconstructed myeloid-specific regulatory networks to define disease master regulators modulated by the newly identified disease driver genes. We observed significant association at the HLA-DRB1 (OR=1.55, P=1.1×10-25) and fine-mapped specific amino-acid risk substitutions in DRβ1. We discovered two novel non-HLA loci: FCAR (OR=1.51, P=1.0×10-20) encoding a myeloid IgA receptor FcαR, and INPP5D (OR=1.34, P=2.2×10-9) encoding a known inhibitor of FcαR signaling. The FCAR risk locus co-localized with a cis-eQTL increasing FCAR expression; the risk alleles disrupted a PRDM1 binding motif within a myeloid enhancer of FCAR. Another risk locus was associated with a higher genetically predicted levels of plasma IL6R. The IL6R risk haplotype carried a missense variant contributing to accelerated cleavage of IL6R into a soluble form. Using systems biology approaches, we prioritized IgAV master regulators co-modulated by FCAR, INPP5D and IL6R in myeloid cells. We additionally identified 21 shared loci in a cross-phenotype analysis of IgAV with IgA nephropathy, including novel loci PAID4, WLS, and ANKRD55.
Collapse
Affiliation(s)
- Lili Liu
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Li Zhu
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - Sara Monteiro-Martins
- Institute of Genetic Epidemiology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Aaron Griffin
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Lukas J. Vlahos
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Masashi Fujita
- Division of Neuroimmunology, Department of Neurology, Columbia University, New York, NY, USA
| | - Cecilia Berrouet
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Francesca Zanoni
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Maddalena Marasa
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Jun Y. Zhang
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Xu-jie Zhou
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - Yasar Caliskan
- Division of Nephrology, Saint Louis University, Saint Louis, MO, USA
| | | | | | | | - Monica Bodria
- MONICA BODRIA, MD, PHD, Primary Care Unit, Ausl Parma, south east district, Parma, Italy
| | - Aftab Chishti
- Division of Pediatric Nephrology, University of Kentucky, Kentucky Children’s Hospital, Lexington, KY, USA
| | - Ciro Esposito
- Istituti Clinico Scientifici Maugeri IRCCS, University of Pavia, Pavia, Italy
| | - Vittoria Esposito
- Istituti Clinico Scientifici Maugeri IRCCS, University of Pavia, Pavia, Italy
| | - Donna Claes
- Cinncinnati Children’s Hospital, Cincinnati, OH, USA
| | - Vladimir Tesar
- Department of Nephrology, 1st School of Medicine, Charles University Prague, Czech Republic
| | | | - Dmitry Samsonov
- Maria Fareri Children’s Hospital (MCF), New York Medical College, New York, NY, USA
| | - Dorota Kaminska
- Department of Non-Procedural Clinical Sciences, Faculty of Medicine, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Tomasz Hryszko
- 2nd Department of Nephrology, Hypertension and Internal Medicine, Medical University of Bialystok, Poland
| | - Gianluigi Zaza
- Renal, Dialysis and Transplant Unit, Department of Pharmacy, Health and Nutritional Sciences (DFSSN), University of Calabria
| | - Joseph T. Flynn
- Department of Pediatrics, University of Washington; and Division of Nephrology, Seattle Children’s Hospital
| | | | | | - Dana Rizk
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | | | | - Luisa Bono
- Nephrology and Dialysis, A.R.N.A.S. Civico and Benfratellio, Palermo, Italy
| | - Laila-Yasmin Mani
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Bruno Vogt
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Fangming Lin
- Division of Pediatric Nephrology, Department of Medicine, Columbia University, New York, NY, USA
| | | | | | | | | | | | | | | | - Scott Wenderfer
- Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Dave Selewski
- Mott Children’s Hospital, University of Michigan, Ann Arbor, MI, USA
| | - Sigrid Lundberg
- Danderyd University Hospital, Karolinska Institute, Stockholm, Sweden
| | - Cynthia Silva
- Connecticut Children’s Medical Center, Hartford, CT, USA
| | - Sherene Mason
- Connecticut Children’s Medical Center, Hartford, CT, USA
| | | | | | - Krzysztof Mucha
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Bartosz Foroncewicz
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Leszek Pączek
- Department of Clinical Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Michał Florczak
- Department of Transplantology, Immunology, Nephrology and Internal Diseases, Medical University of Warsaw, Warsaw, Poland
| | | | - Agnieszka Gradzińska
- Department of Dermatology and Pediatric Dermatology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Maria Szczepańska
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Edyta Machura
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Andrzej Badeński
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Helena Krakowczyk
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | - Przemysław Sikora
- Department of Pediatric Nephrology, Medical University of Lublin, Lublin, Poland
| | - Norbert Kwella
- Department of Nephrology, Transplantology and Internal Diseases, University of Warmia and Mazury, Olsztyn, Poland
| | - Monika Miklaszewska
- Department of Pediatric Nephrology and Hypertension, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Dorota Drożdż
- Department of Pediatric Nephrology and Hypertension, Jagiellonian University Medical College, Krakow, Poland
| | - Marcin Zaniew
- Department of Pediatrics, University of Zielona Góra, Zielona Góra, Poland
| | - Krzysztof Pawlaczyk
- Department of Nephrology, Transplantology and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna SiniewiczLuzeńczyk
- Department of Paediatrics, Immunology and Nephrology, Polish Mother’s Memorial Hospital Research Institute, Lodz, Poland
| | | | | | - Claudia Izzi
- Department of Medical and Surgical Specialties and Nephrology Unit, University of Brescia-ASST Spedali Civili, Brescia, Italy
| | - Francesco Scolari
- Department of Medical and Surgical Specialties and Nephrology Unit, University of Brescia-ASST Spedali Civili, Brescia, Italy
| | | | | | - Laureline Berthelot
- Nantes University, Inserm, CR2TI Center of Research on Translational Transplantation and Immunology, Nantes, France
| | - Evangeline Pillebout
- Center for Research on Inflammation, Paris Cité University, INSERM and CNRS, Paris, France
| | - Renato C. Monteiro
- Center for Research on Inflammation, Paris Cité University, INSERM and CNRS, Paris, France
| | - Jan Novak
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | - Robert J. Wyatt
- Department of Pediatrics, University of Tennessee Health Sciences Center, Memphis, Tennessee
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, Tennessee
| | | | - Javier Martin
- Institute of Parasitology and Biomedicine Lopez-Neyra, Spanish National Research Council (CSIC), Granada, Spain
| | - Miguel A. González-Gay
- Division of Rheumatology, IIS-Fundación Jiménez Díaz, Madrid, Spain
- Medicine and Psychiatry Department, University of Cantabria, Santander, Spain
| | - Philip L. De Jager
- Division of Neuroimmunology, Department of Neurology, Columbia University, New York, NY, USA
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Medical Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS – Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Andrea Califano
- Department of Systems Biology, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
- Department of Biochemistry and Molecular Biophysics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Biomedical Informatics, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Chan Zuckerberg Biohub New York, New York, NY, USA
| | - Ali G. Gharavi
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| | - Hong Zhang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
| | - Krzysztof Kiryluk
- Department of Medicine, Division of Nephrology, Columbia University, College of Physicians & Surgeons, New York, NY, USA
| |
Collapse
|
57
|
Lu K, Chiu KY, Chen IC, Lin GC. Identification of GTF2I Polymorphisms as Potential Biomarkers for CKD in the Han Chinese Population : Multicentric Collaborative Cross-Sectional Cohort Study. KIDNEY360 2024; 5:1466-1476. [PMID: 39024039 PMCID: PMC11556913 DOI: 10.34067/kid.0000000000000517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 07/12/2024] [Indexed: 07/20/2024]
Abstract
Key Points Genetic factors are key players in CKD, with two linked single-nucleotide polymorphisms in the GTF2I gene, associated with CKD susceptibility in the Taiwanese population. Individuals with specific GTF2I genotypes (CT/TT for rs117026326 and CT/CC for rs73366469) show higher CKD prevalence and earlier onset. Men with the specific genotypes of rs117026326 and rs73366469 face a heightened CKD risk compared with women, particularly at lower eGFR. Background CKD poses a global health challenge, but its molecular mechanisms are poorly understood. Genetic factors play a critical role, and phenome-wide association studies and genome-wide association studies shed light on CKD's genetic architecture, shared variants, and biological pathways. Methods Using data from the multicenter collaborative precision medicine cohort, we conducted a retrospective prospectively maintained cross-sectional study. Participants with comprehensive information and genotyping data were selected, and genome-wide association study and phenome-wide association study analyses were performed using the curated Taiwan Biobank version 2 array to identify CKD-associated genetic variants and explore their phenotypic associations. Results Among 58,091 volunteers, 8420 participants were enrolled. Individuals with CKD exhibited higher prevalence of metabolic, cardiovascular, autoimmune, and nephritic disorders. Genetic analysis unveiled two closely linked single-nucleotide polymorphisms, rs117026326 and rs73366469, both associated with GTF2I and CKD (r 2 = 0.64). Further examination revealed significant associations between these single-nucleotide polymorphisms and various kidney-related diseases. The CKD group showed a higher proportion of individuals with specific genotypes (CT/TT for rs117026326 and CT/CC for rs73366469), suggesting potential associations with CKD susceptibility (P < 0.001). Furthermore, individuals with these genotypes developed CKD at an earlier age. Multiple logistic regression confirmed the independent association of these genetic variants with CKD. Subgroup analysis based on eGFR demonstrated an increased risk of CKD among carriers of the rs117026326 CT/TT genotypes (odds ratio [OR], 1.15; 95% confidence interval [CI], 1.07 to 1.24; P < 0.001; OR, 1.32, 95% CI, 1.04 to 1.66; P = 0.02, respectively) and carriers of the rs73366469 CT/CC genotypes (OR, 1.13; 95% CI, 1.05 to 1.21; P < 0.001; OR, 1.31; 95% CI, 1.08 to 1.58; P = 0.0049, respectively). In addition, men had a higher CKD risk than women at lower eGFR levels (OR, 1.35; 95% CI, 1.13 to 1.61; P < 0.001). Conclusions Our study reveals important links between genetic variants GTF2I and susceptibility to CKD, advancing our understanding of CKD development in the Taiwanese population and suggesting potential for personalized prevention and management strategies. More research is needed to validate and explore these variants in diverse populations.
Collapse
Affiliation(s)
- Kevin Lu
- College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Urology, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Kun-Yuan Chiu
- Department of Urology, Taichung Veterans General Hospital, Taichung, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - I-Chieh Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Guan-Cheng Lin
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| |
Collapse
|
58
|
Guo J, Kiryluk K, Wang S. PheW 2P2V: a phenome-wide prediction framework with weighted patient representations using electronic health records. JAMIA Open 2024; 7:ooae084. [PMID: 39282083 PMCID: PMC11401611 DOI: 10.1093/jamiaopen/ooae084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 09/05/2024] [Indexed: 09/18/2024] Open
Abstract
Objective Electronic health records (EHRs) provide opportunities for the development of computable predictive tools. Conventional machine learning methods and deep learning methods have been widely used for this task, with the approach of usually designing one tool for one clinical outcome. Here we developed PheW2P2V, a Phenome-Wide prediction framework using Weighted Patient Vectors. PheW2P2V conducts tailored predictions for phenome-wide phenotypes using numeric representations of patients' past medical records weighted based on their similarities with individual phenotypes. Materials and Methods PheW2P2V defines clinical disease phenotypes using Phecode mapping based on International Classification of Disease codes, which reduces redundancy and case-control misclassification in real-life EHR datasets. Through upweighting medical records of patients that are more relevant to a phenotype of interest in calculating patient vectors, PheW2P2V achieves tailored incidence risk prediction of a phenotype. The calculation of weighted patient vectors is computationally efficient, and the weighting mechanism ensures tailored predictions across the phenome. We evaluated prediction performance of PheW2P2V and baseline methods with simulation studies and clinical applications using the MIMIC-III database. Results Across 942 phenome-wide predictions using the MIMIC-III database, PheW2P2V has median area under the receiver operating characteristic curve (AUC-ROC) 0.74 (baseline methods have values ≤0.72), median max F1-score 0.20 (baseline methods have values ≤0.19), and median area under the precision-recall curve (AUC-PR) 0.10 (baseline methods have values ≤0.10). Discussion PheW2P2V can predict phenotypes efficiently by using medical concept embeddings and upweighting relevant past medical histories. By leveraging both labeled and unlabeled data, PheW2P2V reduces overfitting and improves predictions for rare phenotypes, making it a useful screening tool for early diagnosis of high-risk conditions, though further research is needed to assess the transferability of embeddings across different databases. Conclusions PheW2P2V is fast, flexible, and has superior prediction performance for many clinical disease phenotypes across the phenome of the MIMIC-III database compared to that of several popular baseline methods.
Collapse
Affiliation(s)
- Jia Guo
- Department of Biostatistics, Columbia University, New York, NY 10032, United States
| | - Krzysztof Kiryluk
- Department of Medicine, Columbia University, New York, NY 10032, United States
| | - Shuang Wang
- Department of Biostatistics, Columbia University, New York, NY 10032, United States
| |
Collapse
|
59
|
Jin W, Boss J, Bakulski KM, Goutman SA, Feldman EL, Fritsche LG, Mukherjee B. Improving prediction models of amyotrophic lateral sclerosis (ALS) using polygenic, pre-existing conditions, and survey-based risk scores in the UK Biobank. J Neurol 2024; 271:6923-6934. [PMID: 39249108 DOI: 10.1007/s00415-024-12644-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/12/2024] [Accepted: 08/16/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND AND OBJECTIVES Amyotrophic lateral sclerosis (ALS) causes profound impairments in neurological function, and a cure for this devastating disease remains elusive. This study aimed to identify pre-disposing genetic, phenotypic, and exposure-related factors for amyotrophic lateral sclerosis using multi-modal data and assess their joint predictive potential. METHODS Utilizing data from the UK (United Kingdom) Biobank, we analyzed an unrelated set of 292 ALS cases and 408,831 controls of European descent. Two polygenic risk scores (PRS) are constructed: "GWAS Hits PRS" and "PRS-CS," reflecting oligogenic and polygenic ALS risk profiles, respectively. Time-restricted phenome-wide association studies (PheWAS) were performed to identify pre-existing conditions increasing ALS risk, integrated into phenotypic risk scores (PheRS). A poly-exposure score ("PXS") captures the influence of environmental exposures measured through survey questionnaires. We evaluate the performance of these scores for predicting ALS incidence and stratifying risk, adjusting for baseline demographic covariates. RESULTS Both PRSs modestly predicted ALS diagnosis but with increased predictive power when combined (covariate-adjusted receiver operating characteristic [AAUC] = 0.584 [0.525, 0.639]). PheRS incorporated diagnoses 1 year before ALS onset (PheRS1) modestly discriminated cases from controls (AAUC = 0.515 [0.472, 0.564]). The "PXS" did not significantly predict ALS. However, a model incorporating PRSs and PheRS1 improved the prediction of ALS (AAUC = 0.604 [0.547, 0.667]), outperforming a model combining all risk scores. This combined risk score identified the top 10% of risk score distribution with a fourfold higher ALS risk (95% CI [2.04, 7.73]) versus those in the 40%-60% range. DISCUSSION By leveraging UK Biobank data, our study uncovers pre-disposing ALS factors, highlighting the improved effectiveness of multi-factorial prediction models to identify individuals at highest risk for ALS.
Collapse
Affiliation(s)
- Weijia Jin
- Department of Biostatistics, University of Florida, Gainesville, FL, 32603, USA
| | - Jonathan Boss
- Department of Biostatistics, University of Michigan, University of Michigan, Ann Arbor, MI, 48109, USA
- Center for Precision Health Data Science, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kelly M Bakulski
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Stephen A Goutman
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Lars G Fritsche
- Department of Biostatistics, University of Michigan, University of Michigan, Ann Arbor, MI, 48109, USA.
- Center for Precision Health Data Science, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Bhramar Mukherjee
- Department of Biostatistics, University of Michigan, University of Michigan, Ann Arbor, MI, 48109, USA.
- Center for Precision Health Data Science, University of Michigan, Ann Arbor, MI, 48109, USA.
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, 48109, USA.
- Michigan Institute for Data Science, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
60
|
Yin R, Wack M, Hassen-Khodja C, McDuffie MT, Bliss G, Horn EJ, Kothari C, McLarney B, Davis R, Hanson K, O'Boyle M, Betancur C, Avillach P. Phenome-wide profiling identifies genotype-phenotype associations in Phelan-McDermid syndrome using family-sourced data from an international registry. Mol Autism 2024; 15:40. [PMID: 39350236 PMCID: PMC11443936 DOI: 10.1186/s13229-024-00619-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Phelan-McDermid syndrome (PMS) is a rare neurodevelopmental disorder caused by 22q13 deletions that include the SHANK3 gene or pathogenic sequence variants in SHANK3. It is characterized by global developmental delay, intellectual disability, speech impairment, autism spectrum disorder, and hypotonia; other variable features include epilepsy, brain and renal malformations, and mild dysmorphic features. Here, we conducted genotype-phenotype correlation analyses using the PMS International Registry, a family-driven registry that compiles clinical data in the form of family-reported outcomes and family-sourced genetic test results. METHODS Data from the registry were harmonized and integrated into the i2b2/tranSMART clinical and genomics data warehouse. We gathered information from 401 individuals with 22q13 deletions including SHANK3 (n = 350, ranging in size from 10 kb to 9.1 Mb) or pathogenic or likely pathogenic SHANK3 sequence variants (n = 51), and used regression models with deletion size as a potential predictor of clinical outcomes for 328 phenotypes. RESULTS Our results showed that increased deletion size was significantly associated with delay in gross and fine motor acquisitions, a spectrum of conditions related to poor muscle tone, renal malformations, mild dysmorphic features (e.g., large fleshy hands, sacral dimple, dysplastic toenails, supernumerary teeth), lymphedema, congenital heart defects, and more frequent neuroimaging abnormalities and infections. These findings indicate that genes upstream of SHANK3 also contribute to some of the manifestations of PMS in individuals with larger deletions. We also showed that self-help skills, verbal ability and a range of psychiatric diagnoses (e.g., autism, ADHD, anxiety disorder) were more common among individuals with smaller deletions and SHANK3 variants. LIMITATIONS Some participants were tested with targeted 22q microarrays rather than genome-wide arrays, and karyotypes were unavailable in many cases, thus precluding the analysis of the effect of other copy number variants or chromosomal rearrangements on the phenotype. CONCLUSIONS This is the largest reported case series of individuals with PMS. Overall, we demonstrate the feasibility of using data from a family-sourced registry to conduct genotype-phenotype analyses in rare genetic disorders. We replicate and strengthen previous findings, and reveal novel associations between larger 22q13 deletions and congenital heart defects, neuroimaging abnormalities and recurrent infections.
Collapse
Affiliation(s)
- Rui Yin
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, USA
| | - Maxime Wack
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
| | - Claire Hassen-Khodja
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
| | - Michael T McDuffie
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
| | | | | | - Cartik Kothari
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
| | | | - Rebecca Davis
- Phelan-McDermid Syndrome Foundation, Osprey, FL, 34229, USA
| | - Kristen Hanson
- Phelan-McDermid Syndrome Foundation, Osprey, FL, 34229, USA
| | - Megan O'Boyle
- Phelan-McDermid Syndrome Foundation, Osprey, FL, 34229, USA
| | - Catalina Betancur
- INSERM, CNRS, Neuroscience Paris Seine, Institut de Biologie Paris Seine, Sorbonne Université, 75005, Paris, France.
| | - Paul Avillach
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
61
|
Wilson M, Lee H, Dall'Aglio L, Li X, Kumar A, Colvin MK, Smoller JW, Beardslee WR, Choi KW. Time Trends in Adolescent Diagnoses of Major Depressive Disorder and Co-occurring Psychiatric Conditions in Electronic Health Records. RESEARCH SQUARE 2024:rs.3.rs-4925993. [PMID: 39372932 PMCID: PMC11451741 DOI: 10.21203/rs.3.rs-4925993/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Major depressive disorder (MDD) is highly prevalent in youth and generally characterized by psychiatric comorbidities. Secular trends in co-occurring diagnoses remain unclear, especially in healthcare settings. Using large-scale electronic health records data from a major U.S. healthcare system, we examined the prevalence of MDD diagnoses and co-occurring psychiatric conditions during adolescence (12-18 years; N = 133,753) across four generations (birth years spanning 1985 to 2002) and by sex. Then using a phenome-wide association analysis, we explored which of 67 psychiatric conditions were associated with adolescent MDD diagnosis in earlier versus recent generations. Adolescent MDD diagnosis prevalence increased (8.9 to 11.4%) over time. Over 60% with an MDD diagnosis had co-occurring psychiatric diagnoses, especially neurodevelopmental and anxiety disorders. Co-occurring diagnoses generally increased over time, especially for anxiety disorders (14 to 50%) and suicidal behaviors (6 to 23%), across both sexes. Eight comorbidities interacted with generation, showing stronger associations with MDD diagnosis in earlier (e.g., conduct disorder) versus more recent (e.g., suicidal ideation and behaviors) generations. The findings underscore the importance of assessing psychiatric complexity in adolescents diagnosed with MDD, applying transdiagnostic approaches to address co-occurring presentations, and further investigating potential causes for generational increases.
Collapse
Affiliation(s)
- Marina Wilson
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital
| | - Hyunjoon Lee
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital
| | - Lorenza Dall'Aglio
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital
| | - Xinyun Li
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital
| | - Anushka Kumar
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital
| | - Mary K Colvin
- Department of Psychiatry, Massachusetts General Hospital
| | - Jordan W Smoller
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital
| | | | - Karmel W Choi
- Center for Precision Psychiatry, Department of Psychiatry, Massachusetts General Hospital
| |
Collapse
|
62
|
Mukherjee S, McCaw ZR, Pei J, Merkoulovitch A, Soare T, Tandon R, Amar D, Somineni H, Klein C, Satapati S, Lloyd D, Probert C, Koller D, O’Dushlaine C, Karaletsos T. EmbedGEM: a framework to evaluate the utility of embeddings for genetic discovery. BIOINFORMATICS ADVANCES 2024; 4:vbae135. [PMID: 39664859 PMCID: PMC11632179 DOI: 10.1093/bioadv/vbae135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/13/2024] [Indexed: 12/13/2024]
Abstract
Summary Machine learning-derived embeddings are a compressed representation of high content data modalities. Embeddings can capture detailed information about disease states and have been qualitatively shown to be useful in genetic discovery. Despite their promise, embeddings have a major limitation: it is unclear if genetic variants associated with embeddings are relevant to the disease or trait of interest. In this work, we describe EmbedGEM (Embedding Genetic Evaluation Methods), a framework to systematically evaluate the utility of embeddings in genetic discovery. EmbedGEM focuses on comparing embeddings along two axes: heritability and disease relevance. As measures of heritability, we consider the number of genome-wide significant associations and the meanχ 2 statistic at significant loci. For disease relevance, we compute polygenic risk scores for each embedding principal component, then evaluate their association with high-confidence disease or trait labels in a held-out evaluation patient set. While our development of EmbedGEM is motivated by embeddings, the approach is generally applicable to multivariate traits and can readily be extended to accommodate additional metrics along the evaluation axes. We demonstrate EmbedGEM's utility by evaluating embeddings and multivariate traits in two separate datasets: (i) a synthetic dataset simulated to demonstrate the ability of the framework to correctly rank traits based on their heritability and disease relevance and (ii) a real data from the UK Biobank, including metabolic and liver-related traits. Importantly, we show that greater disease relevance does not automatically follow from greater heritability. Availability and implementation https://github.com/insitro/EmbedGEM.
Collapse
Affiliation(s)
- Sumit Mukherjee
- Insitro Inc, South San Francisco, California 94080, United States
| | - Zachary R McCaw
- Insitro Inc, South San Francisco, California 94080, United States
| | - Jingwen Pei
- Insitro Inc, South San Francisco, California 94080, United States
| | | | - Tom Soare
- Insitro Inc, South San Francisco, California 94080, United States
| | - Raghav Tandon
- Center for Machine Learning, Georgia Institute of Technology, Georgia 30332, United States
| | - David Amar
- Insitro Inc, South San Francisco, California 94080, United States
| | - Hari Somineni
- Insitro Inc, South San Francisco, California 94080, United States
| | - Christoph Klein
- Insitro Inc, South San Francisco, California 94080, United States
| | | | - David Lloyd
- Insitro Inc, South San Francisco, California 94080, United States
| | | | | | - Daphne Koller
- Insitro Inc, South San Francisco, California 94080, United States
| | - Colm O’Dushlaine
- Insitro Inc, South San Francisco, California 94080, United States
| | | |
Collapse
|
63
|
Wen A, Wang L, He H, Fu S, Liu S, Hanauer DA, Harris DR, Kavuluru R, Zhang R, Natarajan K, Pavinkurve NP, Hajagos J, Rajupet S, Lingam V, Saltz M, Elowsky C, Moffitt RA, Koraishy FM, Palchuk MB, Donovan J, Lingrey L, Stone-DerHagopian G, Miller RT, Williams AE, Leese PJ, Kovach PI, Pfaff ER, Zemmel M, Pates RD, Guthe N, Haendel MA, Chute CG, Liu H. A Case Demonstration of the Open Health Natural Language Processing Toolkit From the National COVID-19 Cohort Collaborative and the Researching COVID to Enhance Recovery Programs for a Natural Language Processing System for COVID-19 or Postacute Sequelae of SARS CoV-2 Infection: Algorithm Development and Validation. JMIR Med Inform 2024; 12:e49997. [PMID: 39250782 PMCID: PMC11420592 DOI: 10.2196/49997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 12/11/2023] [Accepted: 03/01/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND A wealth of clinically relevant information is only obtainable within unstructured clinical narratives, leading to great interest in clinical natural language processing (NLP). While a multitude of approaches to NLP exist, current algorithm development approaches have limitations that can slow the development process. These limitations are exacerbated when the task is emergent, as is the case currently for NLP extraction of signs and symptoms of COVID-19 and postacute sequelae of SARS-CoV-2 infection (PASC). OBJECTIVE This study aims to highlight the current limitations of existing NLP algorithm development approaches that are exacerbated by NLP tasks surrounding emergent clinical concepts and to illustrate our approach to addressing these issues through the use case of developing an NLP system for the signs and symptoms of COVID-19 and PASC. METHODS We used 2 preexisting studies on PASC as a baseline to determine a set of concepts that should be extracted by NLP. This concept list was then used in conjunction with the Unified Medical Language System to autonomously generate an expanded lexicon to weakly annotate a training set, which was then reviewed by a human expert to generate a fine-tuned NLP algorithm. The annotations from a fully human-annotated test set were then compared with NLP results from the fine-tuned algorithm. The NLP algorithm was then deployed to 10 additional sites that were also running our NLP infrastructure. Of these 10 sites, 5 were used to conduct a federated evaluation of the NLP algorithm. RESULTS An NLP algorithm consisting of 12,234 unique normalized text strings corresponding to 2366 unique concepts was developed to extract COVID-19 or PASC signs and symptoms. An unweighted mean dictionary coverage of 77.8% was found for the 5 sites. CONCLUSIONS The evolutionary and time-critical nature of the PASC NLP task significantly complicates existing approaches to NLP algorithm development. In this work, we present a hybrid approach using the Open Health Natural Language Processing Toolkit aimed at addressing these needs with a dictionary-based weak labeling step that minimizes the need for additional expert annotation while still preserving the fine-tuning capabilities of expert involvement.
Collapse
Affiliation(s)
- Andrew Wen
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN, United States
- McWilliams School of Biomedical Informatics, University of Texas Health Sciences Center at Houston, Houston, TX, United States
| | - Liwei Wang
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN, United States
- McWilliams School of Biomedical Informatics, University of Texas Health Sciences Center at Houston, Houston, TX, United States
| | - Huan He
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN, United States
| | - Sunyang Fu
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN, United States
- McWilliams School of Biomedical Informatics, University of Texas Health Sciences Center at Houston, Houston, TX, United States
| | - Sijia Liu
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN, United States
| | - David A Hanauer
- Department of Learning Health Sciences, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Daniel R Harris
- Institute for Pharmaceutical Outcomes and Policy, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Ramakanth Kavuluru
- Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky, Lexington, KY, United States
| | - Rui Zhang
- Division of Health Data Science, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Karthik Natarajan
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, United States
| | - Nishanth P Pavinkurve
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, United States
| | - Janos Hajagos
- Department of Biomedical Informatics, Stony Brook Medicine, Stony Brook, NY, United States
| | - Sritha Rajupet
- Department of Biomedical Informatics, Stony Brook Medicine, Stony Brook, NY, United States
| | - Veena Lingam
- Department of Biomedical Informatics, Stony Brook Medicine, Stony Brook, NY, United States
| | - Mary Saltz
- Department of Biomedical Informatics, Stony Brook Medicine, Stony Brook, NY, United States
| | - Corey Elowsky
- Department of Biomedical Informatics, Stony Brook Medicine, Stony Brook, NY, United States
| | - Richard A Moffitt
- Department of Biomedical Informatics, Stony Brook Medicine, Stony Brook, NY, United States
| | - Farrukh M Koraishy
- Division of Nephrology, Stony Brook Medicine, Stony Brook, NY, United States
| | | | | | | | | | - Robert T Miller
- Clinical and Translational Science Institute, Tufts Medical Center, Boston, MA, United States
| | - Andrew E Williams
- Clinical and Translational Science Institute, Tufts Medical Center, Boston, MA, United States
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, United States
| | - Peter J Leese
- North Carolina Translational and Clinical Sciences Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Paul I Kovach
- North Carolina Translational and Clinical Sciences Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Emily R Pfaff
- North Carolina Translational and Clinical Sciences Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Mikhail Zemmel
- University of Virginia, Charlottesville, VA, United States
| | - Robert D Pates
- University of Virginia, Charlottesville, VA, United States
| | - Nick Guthe
- Department of Population Health, New York University Grossman School of Medicine, New York, NY, United States
| | - Melissa A Haendel
- University of Colorado Anschutz Medical Campus, Denver, CO, United States
| | - Christopher G Chute
- Schools of Medicine, Public Health, and Nursing, Johns Hopkins University, Baltimore, MD, United States
| | - Hongfang Liu
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN, United States
- McWilliams School of Biomedical Informatics, University of Texas Health Sciences Center at Houston, Houston, TX, United States
| |
Collapse
|
64
|
Sanchez-Ruiz JA, Coombes BJ, Pazdernik VM, Melhuish Beaupre LM, Jenkins GD, Pendegraft RS, Batzler A, Ozerdem A, McElroy SL, Gardea-Resendez MA, Cuellar-Barboza AB, Prieto ML, Frye MA, Biernacka JM. Clinical and genetic contributions to medical comorbidity in bipolar disorder: a study using electronic health records-linked biobank data. Mol Psychiatry 2024; 29:2701-2713. [PMID: 38548982 PMCID: PMC11544602 DOI: 10.1038/s41380-024-02530-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 02/21/2024] [Accepted: 03/13/2024] [Indexed: 06/14/2024]
Abstract
Bipolar disorder is a chronic and complex polygenic disease with high rates of comorbidity. However, the independent contribution of either diagnosis or genetic risk of bipolar disorder to the medical comorbidity profile of individuals with the disease remains unresolved. Here, we conducted a multi-step phenome-wide association study (PheWAS) of bipolar disorder using phenomes derived from the electronic health records of participants enrolled in the Mayo Clinic Biobank and the Mayo Clinic Bipolar Disorder Biobank. First, we explored the conditions associated with a diagnosis of bipolar disorder by conducting a phenotype-based PheWAS followed by LASSO-penalized regression to account for correlations within the phenome. Then, we explored the conditions associated with bipolar disorder polygenic risk score (BD-PRS) using a PRS-based PheWAS with a sequential exclusion approach to account for the possibility that diagnosis, instead of genetic risk, may drive such associations. 53,386 participants (58.7% women) with a mean age at analysis of 67.8 years (SD = 15.6) were included. A bipolar disorder diagnosis (n = 1479) was associated with higher rates of psychiatric conditions, injuries and poisonings, endocrine/metabolic and neurological conditions, viral hepatitis C, and asthma. BD-PRS was associated with psychiatric comorbidities but, in contrast, had no positive associations with general medical conditions. While our findings warrant confirmation with longitudinal-prospective studies, the limited associations between bipolar disorder genetics and medical conditions suggest that shared environmental effects or environmental consequences of diagnosis may have a greater impact on the general medical comorbidity profile of individuals with bipolar disorder than its genetic risk.
Collapse
Affiliation(s)
| | - Brandon J Coombes
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | - Greg D Jenkins
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | - Anthony Batzler
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Aysegul Ozerdem
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA
| | - Susan L McElroy
- Lindner Center of HOPE/University of Cincinnati, Cincinnati, OH, USA
| | - Manuel A Gardea-Resendez
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA
- Department of Psychiatry, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Alfredo B Cuellar-Barboza
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA
- Department of Psychiatry, Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Miguel L Prieto
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA
- Department of Psychiatry, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
- Mental Health Service, Clínica Universidad de los Andes, Santiago, Chile
| | - Mark A Frye
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA
| | - Joanna M Biernacka
- Department of Psychiatry & Psychology, Mayo Clinic, Rochester, MN, USA.
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
65
|
Breeyear JH, Hellwege JN, Schroeder PH, House JS, Poisner HM, Mitchell SL, Charest B, Khakharia A, Basnet TB, Halladay CW, Reaven PD, Meigs JB, Rhee MK, Sun Y, Lynch MG, Bick AG, Wilson OD, Hung AM, Nealon CL, Iyengar SK, Rotroff DM, Buse JB, Leong A, Mercader JM, Sobrin L, Brantley MA, Peachey NS, Motsinger-Reif AA, Wilson PW, Sun YV, Giri A, Phillips LS, Edwards TL. Adaptive selection at G6PD and disparities in diabetes complications. Nat Med 2024; 30:2480-2488. [PMID: 38918629 PMCID: PMC11555759 DOI: 10.1038/s41591-024-03089-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024]
Abstract
Diabetes complications occur at higher rates in individuals of African ancestry. Glucose-6-phosphate dehydrogenase deficiency (G6PDdef), common in some African populations, confers malaria resistance, and reduces hemoglobin A1c (HbA1c) levels by shortening erythrocyte lifespan. In a combined-ancestry genome-wide association study of diabetic retinopathy, we identified nine loci including a G6PDdef causal variant, rs1050828 -T (Val98Met), which was also associated with increased risk of other diabetes complications. The effect of rs1050828 -T on retinopathy was fully mediated by glucose levels. In the years preceding diabetes diagnosis and insulin prescription, glucose levels were significantly higher and HbA1c significantly lower in those with versus without G6PDdef. In the Action to Control Cardiovascular Risk in Diabetes (ACCORD) trial, participants with G6PDdef had significantly higher hazards of incident retinopathy and neuropathy. At the same HbA1c levels, G6PDdef participants in both ACCORD and the Million Veteran Program had significantly increased risk of retinopathy. We estimate that 12% and 9% of diabetic retinopathy and neuropathy cases, respectively, in participants of African ancestry are due to this exposure. Across continentally defined ancestral populations, the differences in frequency of rs1050828 -T and other G6PDdef alleles contribute to disparities in diabetes complications. Diabetes management guided by glucose or potentially genotype-adjusted HbA1c levels could lead to more timely diagnoses and appropriate intensification of therapy, decreasing the risk of diabetes complications in patients with G6PDdef alleles.
Collapse
Affiliation(s)
- Joseph H Breeyear
- Biostatistics and Computational Biology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, Durham, NC, USA
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- VA Tennessee Valley Healthcare System (626), Nashville, TN, USA
| | - Jacklyn N Hellwege
- VA Tennessee Valley Healthcare System (626), Nashville, TN, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
| | - Philip H Schroeder
- Program in Metabolism, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - John S House
- Biostatistics and Computational Biology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, Durham, NC, USA
| | - Hannah M Poisner
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
| | - Sabrina L Mitchell
- VA Tennessee Valley Healthcare System (626), Nashville, TN, USA
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Brian Charest
- Massachusetts Veterans Epidemiology Research and Information Center, Boston, MA, USA
| | - Anjali Khakharia
- Atlanta VA Medical Center, Decatur, GA, USA
- Department of Medicine and Geriatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Til B Basnet
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Peter D Reaven
- Phoenix VA Health Care System, Phoenix, AZ, USA
- College of Medicine, University of Arizona, Phoenix, AZ, USA
| | - James B Meigs
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Mary K Rhee
- Atlanta VA Medical Center, Decatur, GA, USA
- Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
- Veterans Administration Palo Alto Health Care System, Palo Alto, California, USA
| | | | - Alexander G Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
| | - Otis D Wilson
- VA Tennessee Valley Healthcare System (626), Nashville, TN, USA
| | - Adriana M Hung
- VA Tennessee Valley Healthcare System (626), Nashville, TN, USA
| | - Cari L Nealon
- Eye Clinic, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
- Department of Ophthalmology & Visual Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Sudha K Iyengar
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Daniel M Rotroff
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Endocrinology and Metabolism Institute, Cleveland Clinic, Cleveland, OH, USA
- Center for Quantitative Metabolic Research, Cleveland Clinic, Cleveland, OH, USA
| | - John B Buse
- Division of Endocrinology & Metabolism, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Aaron Leong
- Program in Metabolism, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Josep M Mercader
- Program in Metabolism, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Lucia Sobrin
- Department of Ophthalmology, Mass Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Milam A Brantley
- VA Tennessee Valley Healthcare System (626), Nashville, TN, USA
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Neal S Peachey
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Alison A Motsinger-Reif
- Biostatistics and Computational Biology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, Durham, NC, USA
| | - Peter W Wilson
- Atlanta VA Medical Center, Decatur, GA, USA
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Yan V Sun
- Atlanta VA Medical Center, Decatur, GA, USA
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
| | - Ayush Giri
- VA Tennessee Valley Healthcare System (626), Nashville, TN, USA.
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA.
- Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Lawrence S Phillips
- Atlanta VA Medical Center, Decatur, GA, USA
- Division of Endocrinology, Metabolism, and Lipids, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- VA Tennessee Valley Healthcare System (626), Nashville, TN, USA.
| |
Collapse
|
66
|
Zheng NS, Annis J, Master H, Han L, Gleichauf K, Ching JH, Nasser M, Coleman P, Desine S, Ruderfer DM, Hernandez J, Schneider LD, Brittain EL. Sleep patterns and risk of chronic disease as measured by long-term monitoring with commercial wearable devices in the All of Us Research Program. Nat Med 2024; 30:2648-2656. [PMID: 39030265 PMCID: PMC11405268 DOI: 10.1038/s41591-024-03155-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 06/25/2024] [Indexed: 07/21/2024]
Abstract
Poor sleep health is associated with increased all-cause mortality and incidence of many chronic conditions. Previous studies have relied on cross-sectional and self-reported survey data or polysomnograms, which have limitations with respect to data granularity, sample size and longitudinal information. Here, using objectively measured, longitudinal sleep data from commercial wearable devices linked to electronic health record data from the All of Us Research Program, we show that sleep patterns, including sleep stages, duration and regularity, are associated with chronic disease incidence. Of the 6,785 participants included in this study, 71% were female, 84% self-identified as white and 71% had a college degree; the median age was 50.2 years (interquartile range = 35.7, 61.5) and the median sleep monitoring period was 4.5 years (2.5, 6.5). We found that rapid eye movement sleep and deep sleep were inversely associated with the odds of incident atrial fibrillation and that increased sleep irregularity was associated with increased odds of incident obesity, hyperlipidemia, hypertension, major depressive disorder and generalized anxiety disorder. Moreover, J-shaped associations were observed between average daily sleep duration and hypertension, major depressive disorder and generalized anxiety disorder. These findings show that sleep stages, duration and regularity are all important factors associated with chronic disease development and may inform evidence-based recommendations on healthy sleeping habits.
Collapse
Affiliation(s)
- Neil S Zheng
- Yale School of Medicine, Yale University, New Haven, CT, USA
- Brigham and Women's Hospital, Boston, MA, USA
| | - Jeffrey Annis
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Digital Genomic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hiral Master
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Digital Genomic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lide Han
- Center for Digital Genomic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | | | - Peyton Coleman
- Center for Digital Genomic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Stacy Desine
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Douglas M Ruderfer
- Center for Digital Genomic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, 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
| | | | - Logan D Schneider
- Google, Mountain View, CA, USA
- Sleep Medicine Center, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Redwood City, CA, USA
| | - Evan L Brittain
- Center for Digital Genomic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
67
|
Mackay TFC, Anholt RRH. Pleiotropy, epistasis and the genetic architecture of quantitative traits. Nat Rev Genet 2024; 25:639-657. [PMID: 38565962 PMCID: PMC11330371 DOI: 10.1038/s41576-024-00711-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2024] [Indexed: 04/04/2024]
Abstract
Pleiotropy (whereby one genetic polymorphism affects multiple traits) and epistasis (whereby non-linear interactions between genetic polymorphisms affect the same trait) are fundamental aspects of the genetic architecture of quantitative traits. Recent advances in the ability to characterize the effects of polymorphic variants on molecular and organismal phenotypes in human and model organism populations have revealed the prevalence of pleiotropy and unexpected shared molecular genetic bases among quantitative traits, including diseases. By contrast, epistasis is common between polymorphic loci associated with quantitative traits in model organisms, such that alleles at one locus have different effects in different genetic backgrounds, but is rarely observed for human quantitative traits and common diseases. Here, we review the concepts and recent inferences about pleiotropy and epistasis, and discuss factors that contribute to similarities and differences between the genetic architecture of quantitative traits in model organisms and humans.
Collapse
Affiliation(s)
- Trudy F C Mackay
- Center for Human Genetics, Clemson University, Greenwood, SC, USA.
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC, USA.
| | - Robert R H Anholt
- Center for Human Genetics, Clemson University, Greenwood, SC, USA.
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC, USA.
| |
Collapse
|
68
|
Bigdeli TB, Chatzinakos C, Bendl J, Barr PB, Venkatesh S, Gorman BR, Clarence T, Genovese G, Iyegbe CO, Peterson RE, Kolokotronis SO, Burstein D, Meyers JL, Li Y, Rajeevan N, Sayward F, Cheung KH, DeLisi LE, Kosten TR, Zhao H, Achtyes E, Buckley P, Malaspina D, Lehrer D, Rapaport MH, Braff DL, Pato MT, Fanous AH, Pato CN, Huang GD, Muralidhar S, Michael Gaziano J, Pyarajan S, Girdhar K, Lee D, Hoffman GE, Aslan M, Fullard JF, Voloudakis G, Harvey PD, Roussos P. Biological Insights from Schizophrenia-associated Loci in Ancestral Populations. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.27.24312631. [PMID: 39252912 PMCID: PMC11383513 DOI: 10.1101/2024.08.27.24312631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Large-scale genome-wide association studies of schizophrenia have uncovered hundreds of associated loci but with extremely limited representation of African diaspora populations. We surveyed electronic health records of 200,000 individuals of African ancestry in the Million Veteran and All of Us Research Programs, and, coupled with genotype-level data from four case-control studies, realized a combined sample size of 13,012 affected and 54,266 unaffected persons. Three genome-wide significant signals - near PLXNA4, PMAIP1, and TRPA1 - are the first to be independently identified in populations of predominantly African ancestry. Joint analyses of African, European, and East Asian ancestries across 86,981 cases and 303,771 controls, yielded 376 distinct autosomal loci, which were refined to 708 putatively causal variants via multi-ancestry fine-mapping. Utilizing single-cell functional genomic data from human brain tissue and two complementary approaches, transcriptome-wide association studies and enhancer-promoter contact mapping, we identified a consensus set of 94 genes across ancestries and pinpointed the specific cell types in which they act. We identified reproducible associations of schizophrenia polygenic risk scores with schizophrenia diagnoses and a range of other mental and physical health problems. Our study addresses a longstanding gap in the generalizability of research findings for schizophrenia across ancestral populations, underlining shared biological underpinnings of schizophrenia across global populations in the presence of broadly divergent risk allele frequencies.
Collapse
Affiliation(s)
- Tim B. Bigdeli
- VA New York Harbor Healthcare System, Brooklyn, NY
- Department of Psychiatry and Behavioral Sciences and SUNY Downstate Health Sciences University, Brooklyn, NY
- Institute for Genomics in Health (IGH), SUNY Downstate Health Sciences University, Brooklyn, NY
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Chris Chatzinakos
- Department of Psychiatry and Behavioral Sciences and SUNY Downstate Health Sciences University, Brooklyn, NY
- Institute for Genomics in Health (IGH), SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Jaroslav Bendl
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
| | - Peter B. Barr
- VA New York Harbor Healthcare System, Brooklyn, NY
- Department of Psychiatry and Behavioral Sciences and SUNY Downstate Health Sciences University, Brooklyn, NY
- Institute for Genomics in Health (IGH), SUNY Downstate Health Sciences University, Brooklyn, NY
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Sanan Venkatesh
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Bryan R. Gorman
- Massachusetts Area Veterans Epidemiology, Research, and Information Center (MAVERIC), Jamaica Plain, MA
| | - Tereza Clarence
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
| | - Giulio Genovese
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA
- Harvard Medical School, Boston, MA
| | - Conrad O. Iyegbe
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
| | - Roseann E. Peterson
- VA New York Harbor Healthcare System, Brooklyn, NY
- Department of Psychiatry and Behavioral Sciences and SUNY Downstate Health Sciences University, Brooklyn, NY
- Institute for Genomics in Health (IGH), SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Sergios-Orestis Kolokotronis
- Institute for Genomics in Health (IGH), SUNY Downstate Health Sciences University, Brooklyn, NY
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY
- Division of Infectious Diseases, Department of Medicine, College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY
- Department of Cell Biology, College of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY
| | - David Burstein
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA
- Mental Illness Research, Education and Clinical Center VISN2, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Jacquelyn L. Meyers
- Department of Psychiatry and Behavioral Sciences and SUNY Downstate Health Sciences University, Brooklyn, NY
- Institute for Genomics in Health (IGH), SUNY Downstate Health Sciences University, Brooklyn, NY
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY
| | - Yuli Li
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Yale University School of Medicine, New Haven, CT
| | - Nallakkandi Rajeevan
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Yale University School of Medicine, New Haven, CT
| | - Frederick Sayward
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Yale University School of Medicine, New Haven, CT
| | - Kei-Hoi Cheung
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Yale University School of Medicine, New Haven, CT
| | | | | | | | - Lynn E. DeLisi
- Department of Psychiatry, Cambridge Health Alliance, Cambridge, MA
| | - Thomas R. Kosten
- Michael E. DeBakey VA Medical Center, Houston, TX
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX
| | - Hongyu Zhao
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Yale University School of Medicine, New Haven, CT
| | - Eric Achtyes
- Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI
| | - Peter Buckley
- University of Tennessee Health Science Center in Memphis, TN
| | - Dolores Malaspina
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
| | - Douglas Lehrer
- Department of Psychiatry, Wright State University, Dayton, OH
| | - Mark H. Rapaport
- Huntsman Mental Health Institute, Department of Psychiatry, University of Utah, Salt Lake City, UT
| | - David L. Braff
- Department of Psychiatry, University of California, San Diego, CA
- VA San Diego Healthcare System, San Diego, CA
| | - Michele T. Pato
- Department of Psychiatry, Robert Wood Johnson Medical School, New Brunswick, NJ
| | - Ayman H. Fanous
- Department of Psychiatry, University of Arizona College of Medicine-Phoenix, Phoenix, AZ
- Department of Psychiatry, VA Phoenix Healthcare System, Phoenix, AZ
| | - Carlos N. Pato
- Department of Psychiatry, Robert Wood Johnson Medical School, New Brunswick, NJ
| | | | | | | | - Grant D. Huang
- Office of Research and Development, Veterans Health Administration, Washington, DC
| | - Sumitra Muralidhar
- Office of Research and Development, Veterans Health Administration, Washington, DC
| | - J. Michael Gaziano
- Massachusetts Area Veterans Epidemiology, Research, and Information Center (MAVERIC), Jamaica Plain, MA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Saiju Pyarajan
- Massachusetts Area Veterans Epidemiology, Research, and Information Center (MAVERIC), Jamaica Plain, MA
| | - Kiran Girdhar
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
| | - Donghoon Lee
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
| | - Gabriel E. Hoffman
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA
- Mental Illness Research, Education and Clinical Center VISN2, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Mihaela Aslan
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT
- Yale University School of Medicine, New Haven, CT
| | - John F. Fullard
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
| | - Georgios Voloudakis
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA
- Mental Illness Research, Education and Clinical Center VISN2, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Philip D. Harvey
- Bruce W. Carter Miami Veterans Affairs (VA) Medical Center, Miami, FL
- University of Miami School of Medicine, Miami, FL
| | - Panos Roussos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, NY
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA
- Mental Illness Research, Education and Clinical Center VISN2, James J. Peters VA Medical Center, Bronx, NY, USA
| |
Collapse
|
69
|
Jensen M, Smolen C, Tyryshkina A, Pizzo L, Banerjee D, Oetjens M, Shimelis H, Taylor CM, Pounraja VK, Song H, Rohan L, Huber E, El Khattabi L, van de Laar I, Tadros R, Bezzina C, van Slegtenhorst M, Kammeraad J, Prontera P, Caberg JH, Fraser H, Banka S, Van Dijck A, Schwartz C, Voorhoeve E, Callier P, Mosca-Boidron AL, Marle N, Lefebvre M, Pope K, Snell P, Boys A, Lockhart PJ, Ashfaq M, McCready E, Nowacyzk M, Castiglia L, Galesi O, Avola E, Mattina T, Fichera M, Bruccheri MG, Mandarà GML, Mari F, Privitera F, Longo I, Curró A, Renieri A, Keren B, Charles P, Cuinat S, Nizon M, Pichon O, Bénéteau C, Stoeva R, Martin-Coignard D, Blesson S, Le Caignec C, Mercier S, Vincent M, Martin C, Mannik K, Reymond A, Faivre L, Sistermans E, Kooy RF, Amor DJ, Romano C, Andrieux J, Girirajan S. Genetic modifiers and ascertainment drive variable expressivity of complex disorders. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.27.24312158. [PMID: 39252907 PMCID: PMC11383473 DOI: 10.1101/2024.08.27.24312158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Variable expressivity of disease-associated variants implies a role for secondary variants that modify clinical features. We assessed the effects of modifier variants towards clinical outcomes of 2,252 individuals with primary variants. Among 132 families with the 16p12.1 deletion, distinct rare and common variant classes conferred risk for specific developmental features, including short tandem repeats for neurological defects and SNVs for microcephaly, while additional disease-associated variants conferred multiple genetic diagnoses. Within disease and population cohorts of 773 individuals with the 16p12.1 deletion, we found opposing effects of secondary variants towards clinical features across ascertainments. Additional analysis of 1,479 probands with other primary variants, such as 16p11.2 deletion and CHD8 variants, and 1,084 without primary variants, showed that phenotypic associations differed by primary variant context and were influenced by synergistic interactions between primary and secondary variants. Our study provides a paradigm to dissect the genomic architecture of complex disorders towards personalized treatment.
Collapse
Affiliation(s)
- Matthew Jensen
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
- Bioinformatics and Genomics Graduate program, Pennsylvania State University, University Park, PA 16802, USA
| | - Corrine Smolen
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
- Bioinformatics and Genomics Graduate program, Pennsylvania State University, University Park, PA 16802, USA
| | - Anastasia Tyryshkina
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Lucilla Pizzo
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Deepro Banerjee
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Matthew Oetjens
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA 17837, USA
| | - Hermela Shimelis
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA 17837, USA
| | - Cora M. Taylor
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA 17837, USA
| | - Vijay Kumar Pounraja
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
- Bioinformatics and Genomics Graduate program, Pennsylvania State University, University Park, PA 16802, USA
| | - Hyebin Song
- Department of Statistics, Pennsylvania State University, University Park, PA 16802, USA
| | - Laura Rohan
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Emily Huber
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Laila El Khattabi
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris Cité, CARPEM, Paris, France
| | - Ingrid van de Laar
- Department of Clinical Genetics, Erasmus MC, Univ. Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rafik Tadros
- Department of Clinical Genetics, Erasmus MC, Univ. Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Connie Bezzina
- Department of Clinical Genetics, Erasmus MC, Univ. Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marjon van Slegtenhorst
- Department of Clinical Genetics, Erasmus MC, Univ. Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Janneke Kammeraad
- Department of Clinical Genetics, Erasmus MC, Univ. Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Paolo Prontera
- Medical Genetics Unit, Hospital Santa Maria della Misericordia, Perugia, Italy
| | - Jean-Hubert Caberg
- Centre Hospitalier Universitaire de Liège. Domaine Universitaire du Sart Tilman, Liège, Belgium
| | - Harry Fraser
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Siddhartha Banka
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, St. Mary’s Hospital, Central Manchester University Hospitals, NHS Foundation Trust Manchester Academic Health Sciences Centre, Manchester, UK
| | - Anke Van Dijck
- Department of Medical Genetics, University and University Hospital Antwerp, Antwerp, Belgium
| | | | - Els Voorhoeve
- Department of Clinical Genetics, Amsterdam UMC, Amsterdam, The Netherlands
| | - Patrick Callier
- Center for Rare Diseases and Reference Developmental Anomalies and Malformation Syndromes, CHU Dijon, Dijon, France
| | - Anne-Laure Mosca-Boidron
- Center for Rare Diseases and Reference Developmental Anomalies and Malformation Syndromes, CHU Dijon, Dijon, France
| | - Nathalie Marle
- Center for Rare Diseases and Reference Developmental Anomalies and Malformation Syndromes, CHU Dijon, Dijon, France
| | - Mathilde Lefebvre
- Laboratoire de Genetique Chromosomique et Moleculaire, CHU Dijon, France
| | - Kate Pope
- Bruce Lefroy Centre, Murdoch Children’s Research Institute, Melbourne, Australia
| | - Penny Snell
- Bruce Lefroy Centre, Murdoch Children’s Research Institute, Melbourne, Australia
| | - Amber Boys
- Bruce Lefroy Centre, Murdoch Children’s Research Institute, Melbourne, Australia
| | - Paul J. Lockhart
- Bruce Lefroy Centre, Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Myla Ashfaq
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Elizabeth McCready
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Margaret Nowacyzk
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Lucia Castiglia
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute-IRCCS, Troina, Italy
| | - Ornella Galesi
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute-IRCCS, Troina, Italy
| | - Emanuela Avola
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute-IRCCS, Troina, Italy
| | - Teresa Mattina
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute-IRCCS, Troina, Italy
| | - Marco Fichera
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute-IRCCS, Troina, Italy
- Section of Clinical Biochemistry and Medical Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania School of Medicine, Catania, Italy
| | - Maria Grazia Bruccheri
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute-IRCCS, Troina, Italy
| | | | - Francesca Mari
- Laboratory of Clinical Molecular Genetics and Cytogenetics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Flavia Privitera
- Laboratory of Clinical Molecular Genetics and Cytogenetics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ilaria Longo
- Laboratory of Clinical Molecular Genetics and Cytogenetics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Aurora Curró
- Laboratory of Clinical Molecular Genetics and Cytogenetics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessandra Renieri
- Laboratory of Clinical Molecular Genetics and Cytogenetics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Boris Keren
- Département de Génétique, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, 75019 Paris, France
| | - Perrine Charles
- Département de Génétique, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, 75019 Paris, France
| | | | | | | | | | - Radka Stoeva
- CHU Nantes, Medical Genetics Department, Nantes, France
| | | | - Sophia Blesson
- Department of Genetics, Bretonneau University Hospital, Tours, France
| | - Cedric Le Caignec
- CHU Toulouse, Department of Medical Genetics, Toulouse, France
- Toulouse Neuro Imaging, Center, Inserm, UPS, Université de Toulouse, Toulouse, France
| | - Sandra Mercier
- Department of Genetics, Bretonneau University Hospital, Tours, France
| | - Marie Vincent
- Department of Genetics, Bretonneau University Hospital, Tours, France
| | - Christa Martin
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA 17837, USA
| | - Katrin Mannik
- Institute of Genomics, University of Tartu, Estonia
- Health2030 Genome Center, Fondation Campus Biotech, Geneva, Switzerland
| | - Alexandre Reymond
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Switzerland
| | - Laurence Faivre
- Center for Rare Diseases and Reference Developmental Anomalies and Malformation Syndromes, CHU Dijon, Dijon, France
- Laboratoire de Genetique Chromosomique et Moleculaire, CHU Dijon, France
| | - Erik Sistermans
- Department of Clinical Genetics, Amsterdam UMC, Amsterdam, The Netherlands
| | - R. Frank Kooy
- Department of Medical Genetics, University and University Hospital Antwerp, Antwerp, Belgium
| | - David J. Amor
- Department of Clinical Genetics, Amsterdam UMC, Amsterdam, The Netherlands
| | - Corrado Romano
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute-IRCCS, Troina, Italy
- Section of Clinical Biochemistry and Medical Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania School of Medicine, Catania, Italy
| | - Joris Andrieux
- Institut de Genetique Medicale, Hopital Jeanne de Flandre, CHRU de Lille, Lille, France
| | - Santhosh Girirajan
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
- Bioinformatics and Genomics Graduate program, Pennsylvania State University, University Park, PA 16802, USA
- Department of Anthropology, Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
70
|
Weinstock JS, Chaudhry SA, Ioannou M, Viskadourou M, Reventun P, Jakubek YA, Liggett LA, Laurie C, Broome JG, Khan A, Taylor KD, Guo X, Peyser PA, Boerwinkle E, Chami N, Kenny EE, Loos RJ, Psaty BM, Russell TP, Brody JA, Yun JH, Cho MH, Vasan RS, Kardia SL, Smith JA, Raffield LM, Bidulescu A, O’Brien E, de Andrade M, Rotter JI, Rich SS, Tracy RP, Chen YDI, Gu CC, Hsiung CA, Kooperberg C, Haring B, Nassir R, Mathias R, Reiner A, Sankaran V, Lowenstein CJ, Blackwell TW, Abecasis GR, Smith AV, Kang HM, Natarajan P, Jaiswal S, Bick A, Post WS, Scheet P, Auer P, Karantanos T, Battle A, Arvanitis M. The Genetic Determinants and Genomic Consequences of Non-Leukemogenic Somatic Point Mutations. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.22.24312319. [PMID: 39228737 PMCID: PMC11370504 DOI: 10.1101/2024.08.22.24312319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Clonal hematopoiesis (CH) is defined by the expansion of a lineage of genetically identical cells in blood. Genetic lesions that confer a fitness advantage, such as point mutations or mosaic chromosomal alterations (mCAs) in genes associated with hematologic malignancy, are frequent mediators of CH. However, recent analyses of both single cell-derived colonies of hematopoietic cells and population sequencing cohorts have revealed CH frequently occurs in the absence of known driver genetic lesions. To characterize CH without known driver genetic lesions, we used 51,399 deeply sequenced whole genomes from the NHLBI TOPMed sequencing initiative to perform simultaneous germline and somatic mutation analyses among individuals without leukemogenic point mutations (LPM), which we term CH-LPMneg. We quantified CH by estimating the total mutation burden. Because estimating somatic mutation burden without a paired-tissue sample is challenging, we developed a novel statistical method, the Genomic and Epigenomic informed Mutation (GEM) rate, that uses external genomic and epigenomic data sources to distinguish artifactual signals from true somatic mutations. We performed a genome-wide association study of GEM to discover the germline determinants of CH-LPMneg. After fine-mapping and variant-to-gene analyses, we identified seven genes associated with CH-LPMneg (TCL1A, TERT, SMC4, NRIP1, PRDM16, MSRA, SCARB1), and one locus associated with a sex-associated mutation pathway (SRGAP2C). We performed a secondary analysis excluding individuals with mCAs, finding that the genetic architecture was largely unaffected by their inclusion. Functional analyses of SMC4 and NRIP1 implicated altered HSC self-renewal and proliferation as the primary mediator of mutation burden in blood. We then performed comprehensive multi-tissue transcriptomic analyses, finding that the expression levels of 404 genes are associated with GEM. Finally, we performed phenotypic association meta-analyses across four cohorts, finding that GEM is associated with increased white blood cell count and increased risk for incident peripheral artery disease, but is not significantly associated with incident stroke or coronary disease events. Overall, we develop GEM for quantifying mutation burden from WGS without a paired-tissue sample and use GEM to discover the genetic, genomic, and phenotypic correlates of CH-LPMneg.
Collapse
Affiliation(s)
- Joshua S. Weinstock
- Department of Human Genetics, School of Medicine, Emory University, Atlanta, GA, USA
| | - Sharjeel A. Chaudhry
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD
- Department of Surgery, Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Maria Ioannou
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine
| | - Maria Viskadourou
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | - Paula Reventun
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | | | - L. Alexander Liggett
- Division of Hematology/Oncology, Boston Childrens Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Cecelia Laurie
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Jai G. Broome
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Alyna Khan
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA USA
| | - Patricia A. Peyser
- Department of Epidemiology, School of Public Health, Boston University, Boxton, MA USA
| | - Eric Boerwinkle
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Nathalie Chami
- The Charles Bronfman Institute of Personalized Medicine
- The Mindich Child Health and Developlement Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Ruth J. Loos
- The Charles Bronfman Institute of Personalized Medicine
- The Mindich Child Health and Developlement Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 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
| | - Tracy P. Russell
- Department of Pathology & Laboratory Medicine and Biochemistry, Larner College of Medicine at the University of Vermont, Colchester, VT, USA
| | - Jennifer A. Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jeong H. Yun
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA USA
| | - Michael H. Cho
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA USA
| | - Ramachandran S. Vasan
- National Heart Lung and Blood Institute’s, Boston University’s Framingham Heart Study, Framingham, MA, USA
| | - Sharon L. Kardia
- Department of Epidemiology, University of Michigan, Ann Arbor, MI
| | - Jennifer A. Smith
- Department of Epidemiology, University of Michigan, Ann Arbor, MI
- Survey Research Center, Institute for Social Research, University of Michgian, Ann Arbor, MI
| | - Laura M. Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, NC, 27514
| | - Aurelian Bidulescu
- Department of Epidemiology and Biostatistics, Indiana University School of Public Health Bloomington, Bloomington, IN, USA
| | | | - Mariza de Andrade
- Mayo Clinic, Department of Health Sciences Research, Rochester, MN, USA
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA USA
| | - Stephen S. Rich
- Department of Public Health Sciences, Center for Public Health Genomics, University of Virginia, Charlottesville, VA USA
| | - Russell P. Tracy
- Department of Pathology & Laboratory Medicine and Biochemistry, Larner College of Medicine at the University of Vermont, Colchester, VT, USA
| | - Yii Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA USA
| | - C. Charles. Gu
- Center for Biostatistics and Data Sciences, Washington University, St. Louis, MO USA
| | - Chao A. Hsiung
- Department of Medicine, Taipei Veterans General Hospital, Taipei Taiwan - 201 Shi-Pai Rd. Sec. 2, Taipei Taiwan
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Bernhard Haring
- Department of Medicine III, Saarland University Hospital, Homburg, Saarland, Germany - Department of Medicine I, University of Wrzburg, Wrzburg, Bavaria, Germany
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, USA. Electronic address
| | - Rami Nassir
- University of California Davis, Davis, CA, USA
| | - Rasika Mathias
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alex Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Vijay Sankaran
- Division of Hematology/Oncology, Boston Childrens Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | | | - Thomas W. Blackwell
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Goncalo R. Abecasis
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Albert V. Smith
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Hyun M. Kang
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Pradeep Natarajan
- Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics, Broad Institute of Harvard & MIT, Cambridge, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | | | - Alexander Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Wendy S. Post
- Department of Medicine, Cardiology Division, Johns Hopkins University
| | - Paul Scheet
- Department of Epidemiology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Paul Auer
- Department of Biostatistics, Medical College of WisconsinDivision of Biostatistics, Institute for Health and Equity, and Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Theodoros Karantanos
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine
| | - Alexis Battle
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD
- Department of Computer Science, Johns Hopkins University, Baltimore, MD
| | - Marios Arvanitis
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
71
|
Saltoun K, Yeo BTT, Paul L, Diedrichsen J, Bzdok D. Longitudinal changes in brain asymmetry track lifestyle and disease. RESEARCH SQUARE 2024:rs.3.rs-4798448. [PMID: 39149493 PMCID: PMC11326383 DOI: 10.21203/rs.3.rs-4798448/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Human beings may have evolved the largest asymmetries of brain organization in the animal kingdom. Hemispheric left-vs-right specialization is especially pronounced in our species-unique capacities. Yet, brain asymmetry features appear to be strongly shaped by non-genetic influences. We hence charted the largest longitudinal brain-imaging adult resource, yielding evidence that brain asymmetry changes continuously in a manner suggestive of neural plasticity. In the UK Biobank population cohort, we demonstrate that asymmetry changes show robust associations across 959 distinct phenotypic variables spanning 11 categories. We also find that changes in brain asymmetry over years co-occur with changes among specific lifestyle markers. Finally, we reveal relevance of brain asymmetry changes to major disease categories across thousands of medical diagnoses. Our results challenge the tacit assumption that asymmetrical neural systems are highly conserved throughout adulthood.
Collapse
Affiliation(s)
- Karin Saltoun
- The Neuro - Montreal Neurological Institute (MNI), McConnell Brain Imaging Centre, Department of Biomedical Engineering, Faculty of Medicine, School of Computer Science, McGill University, Montreal, Canada
- Mila - Quebec Artificial Intelligence Institute, Montreal, QC, Canada
| | - B T Thomas Yeo
- Centre for Sleep and Cognition & Centre for Translational MR Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Medicine, Human Potential Translational Research Programme & Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore
- N.1 Institute for Health, National University of Singapore, Singapore
- Integrative Sciences and Engineering Programme (ISEP), National University of Singapore
- Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
| | - Lynn Paul
- Division of the Humanities and Social Sciences, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Jorn Diedrichsen
- Western Institute of Neuroscience, Western University, London, Ontario, Canada
| | - Danilo Bzdok
- The Neuro - Montreal Neurological Institute (MNI), McConnell Brain Imaging Centre, Department of Biomedical Engineering, Faculty of Medicine, School of Computer Science, McGill University, Montreal, Canada
- Mila - Quebec Artificial Intelligence Institute, Montreal, QC, Canada
| |
Collapse
|
72
|
Bigdeli TB, Barr PB, Rajeevan N, Graham DP, Li Y, Meyers JL, Gorman BR, Peterson RE, Sayward F, Radhakrishnan K, Natarajan S, Nielsen DA, Wilkinson AV, Malhotra AK, Zhao H, Brophy M, Shi Y, O'Leary TJ, Gleason T, Przygodzki R, Pyarajan S, Muralidhar S, Gaziano JM, Huang GD, Concato J, Siever LJ, DeLisi LE, Kimbrel NA, Beckham JC, Swann AC, Kosten TR, Fanous AH, Aslan M, Harvey PD. Correlates of suicidal behaviors and genetic risk among United States veterans with schizophrenia or bipolar I disorder. Mol Psychiatry 2024; 29:2399-2407. [PMID: 38491344 DOI: 10.1038/s41380-024-02472-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 01/22/2024] [Accepted: 01/31/2024] [Indexed: 03/18/2024]
Abstract
Persons diagnosed with schizophrenia (SCZ) or bipolar I disorder (BPI) are at high risk for self-injurious behavior, suicidal ideation, and suicidal behaviors (SB). Characterizing associations between diagnosed health problems, prior pharmacological treatments, and polygenic scores (PGS) has potential to inform risk stratification. We examined self-reported SB and ideation using the Columbia Suicide Severity Rating Scale (C-SSRS) among 3,942 SCZ and 5,414 BPI patients receiving care within the Veterans Health Administration (VHA). These cross-sectional data were integrated with electronic health records (EHRs), and compared across lifetime diagnoses, treatment histories, follow-up screenings, and mortality data. PGS were constructed using available genomic data for related traits. Genome-wide association studies were performed to identify and prioritize specific loci. Only 20% of the veterans who reported SB had a corroborating ICD-9/10 EHR code. Among those without prior SB, more than 20% reported new-onset SB at follow-up. SB were associated with a range of additional clinical diagnoses, and with treatment with specific classes of psychotropic medications (e.g., antidepressants, antipsychotics, etc.). PGS for externalizing behaviors, smoking initiation, suicide attempt, and major depressive disorder were associated with SB. The GWAS for SB yielded no significant loci. Among individuals with a diagnosed mental illness, self-reported SB were strongly associated with clinical variables across several EHR domains. Analyses point to sequelae of substance-related and psychiatric comorbidities as strong correlates of prior and subsequent SB. Nonetheless, past SB was frequently not documented in health records, underscoring the value of regular screening with direct, in-person assessments, especially among high-risk individuals.
Collapse
Affiliation(s)
- Tim B Bigdeli
- VA New York Harbor Healthcare System, Brooklyn, NY, US.
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, US.
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, NY, US.
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY, US.
| | - Peter B Barr
- VA New York Harbor Healthcare System, Brooklyn, NY, US
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, US
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, NY, US
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY, US
| | - Nallakkandi Rajeevan
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT, USA
- Yale University School of Medicine, New Haven, CT, USA
| | - David P Graham
- Michael E. DeBakey VA Medical Center, Houston, TX, USA
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Yuli Li
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT, USA
- Yale University School of Medicine, New Haven, CT, USA
| | - Jacquelyn L Meyers
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, US
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, NY, US
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY, US
| | - Bryan R Gorman
- Massachusetts Area Veterans Epidemiology, Research and Information Center (MAVERIC), Jamaica Plain, MA, USA
| | - Roseann E Peterson
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Health Sciences University, Brooklyn, NY, US
- Institute for Genomics in Health, SUNY Downstate Health Sciences University, Brooklyn, NY, US
- Department of Epidemiology and Biostatistics, School of Public Health, SUNY Downstate Health Sciences University, Brooklyn, NY, US
| | - Frederick Sayward
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT, USA
- Yale University School of Medicine, New Haven, CT, USA
| | - Krishnan Radhakrishnan
- National Mental Health and Substance Use Policy Laboratory, Substance Abuse and Mental Health Services Administration, Rockville, MD, USA
| | | | - David A Nielsen
- Michael E. DeBakey VA Medical Center, Houston, TX, USA
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Anna V Wilkinson
- Michael E. DeBakey VA Medical Center, Houston, TX, USA
- Department of Epidemiology, Human Genetics and Environmental Science, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Anil K Malhotra
- Center for Psychiatric Neuroscience, Feinstein Institute for Medical Research, Manhasset, NY, USA
- Division of Psychiatry Research, The Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY, USA
- Department of Psychiatry, Hofstra Northwell School of Medicine, Hempstead, NY, USA
| | - Hongyu Zhao
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT, USA
- Yale University School of Medicine, New Haven, CT, USA
| | - Mary Brophy
- Massachusetts Area Veterans Epidemiology, Research and Information Center (MAVERIC), Jamaica Plain, MA, USA
- Boston University School of Medicine, Boston, MA, USA
| | - Yunling Shi
- Massachusetts Area Veterans Epidemiology, Research and Information Center (MAVERIC), Jamaica Plain, MA, USA
| | - Timothy J O'Leary
- Office of Research and Development, Veterans Health Administration, Washington, DC, USA
| | - Theresa Gleason
- Office of Research and Development, Veterans Health Administration, Washington, DC, USA
| | - Ronald Przygodzki
- Office of Research and Development, Veterans Health Administration, Washington, DC, USA
| | - Saiju Pyarajan
- Massachusetts Area Veterans Epidemiology, Research and Information Center (MAVERIC), Jamaica Plain, MA, USA
| | | | - J Michael Gaziano
- Massachusetts Area Veterans Epidemiology, Research and Information Center (MAVERIC), Jamaica Plain, MA, USA
- Harvard University, Boston, MA, USA
| | - Grant D Huang
- Office of Research and Development, Veterans Health Administration, Washington, DC, USA
| | - John Concato
- Yale University School of Medicine, New Haven, CT, USA
- Office of Research and Development, Veterans Health Administration, Washington, DC, USA
- Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Larry J Siever
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA
| | - Lynn E DeLisi
- Department of Psychiatry, Cambridge Health Alliance, Cambridge, MA, USA
| | - Nathan A Kimbrel
- Durham VA Health Care System, Durham, NC, USA
- VA Mid-Atlantic Mental Illness Research, Education and Clinical Center, Durham, NC, USA
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Jean C Beckham
- Durham VA Health Care System, Durham, NC, USA
- VA Mid-Atlantic Mental Illness Research, Education and Clinical Center, Durham, NC, USA
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Alan C Swann
- Michael E. DeBakey VA Medical Center, Houston, TX, USA
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Thomas R Kosten
- Michael E. DeBakey VA Medical Center, Houston, TX, USA
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Ayman H Fanous
- VA New York Harbor Healthcare System, Brooklyn, NY, US
- Department of Psychiatry, University of Arizona College of Medicine Phoenix, Phoenix, AZ, USA
| | - Mihaela Aslan
- Clinical Epidemiology Research Center (CERC), VA Connecticut Healthcare System, West Haven, CT, USA
- Yale University School of Medicine, New Haven, CT, USA
| | - Philip D Harvey
- Bruce W. Carter Miami Veterans Affairs (VA) Medical Center, Miami, FL, USA
- University of Miami School of Medicine, Miami, FL, USA
| |
Collapse
|
73
|
Upadhyay VR, Ramesh V, Kumar H, Somagond YM, Priyadarsini S, Kuniyal A, Prakash V, Sahoo A. Phenomics in Livestock Research: Bottlenecks and Promises of Digital Phenotyping and Other Quantification Techniques on a Global Scale. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:380-393. [PMID: 39012961 DOI: 10.1089/omi.2024.0109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Bottlenecks in moving genomics to real-life applications also include phenomics. This is true not only for genomics medicine and public health genomics but also in ecology and livestock phenomics. This expert narrative review explores the intricate relationship between genetic makeup and observable phenotypic traits across various biological levels in the context of livestock research. We unpack and emphasize the significance of precise phenotypic data in selective breeding outcomes and examine the multifaceted applications of phenomics, ranging from improvement to assessing welfare, reproductive traits, and environmental adaptation in livestock. As phenotypic traits exhibit strong correlations, their measurement alongside specific biological outcomes provides insights into performance, overall health, and clinical endpoints like morbidity and disease. In addition, automated assessment of livestock holds potential for monitoring the dynamic phenotypic traits across various species, facilitating a deeper comprehension of how they adapt to their environment and attendant stressors. A key challenge in genetic improvement in livestock is predicting individuals with optimal fitness without direct measurement. Temporal predictions from unmanned aerial systems can surpass genomic predictions, offering in-depth data on livestock. In the near future, digital phenotyping and digital biomarkers may further unravel the genetic intricacies of stress tolerance, adaptation and welfare aspects of animals enabling the selection of climate-resilient and productive livestock. This expert review thus delves into challenges associated with phenotyping and discusses technological advancements shaping the future of biological research concerning livestock.
Collapse
Affiliation(s)
| | - Vikram Ramesh
- ICAR-National Research Centre on Mithun, Medziphema, Nagaland, India
| | - Harshit Kumar
- ICAR-National Research Centre on Mithun, Medziphema, Nagaland, India
| | - Y M Somagond
- ICAR-National Research Centre on Mithun, Medziphema, Nagaland, India
| | | | - Aruna Kuniyal
- ICAR-National Research Centre on Camel, Bikaner, Rajasthan, India
| | - Ved Prakash
- ICAR-National Research Centre on Camel, Bikaner, Rajasthan, India
| | - Artabandhu Sahoo
- ICAR-National Research Centre on Camel, Bikaner, Rajasthan, India
| |
Collapse
|
74
|
Wang L, Mesa-Eguiagaray I, Campbell H, Wilson JF, Vitart V, Li X, Theodoratou E. A phenome-wide association and factorial Mendelian randomization study on the repurposing of uric acid-lowering drugs for cardiovascular outcomes. Eur J Epidemiol 2024; 39:869-880. [PMID: 38992218 PMCID: PMC11410910 DOI: 10.1007/s10654-024-01138-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 06/18/2024] [Indexed: 07/13/2024]
Abstract
Uric acid has been linked to various disease outcomes. However, it remains unclear whether uric acid-lowering therapy could be repurposed as a treatment for conditions other than gout. We first performed both observational phenome-wide association study (Obs-PheWAS) and polygenic risk score PheWAS (PRS-PheWAS) to identify associations of uric acid levels with a wide range of disease outcomes. Then, trajectory analysis was conducted to explore temporal progression patterns of the observed disease outcomes. Finally, we investigated whether uric acid-lowering drugs could be repurposed using a factorial Mendelian randomization (MR) study design. A total of 41 overlapping phenotypes associated with uric acid levels were identified by both Obs- and PRS- PheWASs, primarily cardiometabolic diseases. The trajectory analysis illustrated how elevated uric acid levels contribute to cardiometabolic diseases, and finally death. Meanwhile, we found that uric acid-lowering drugs exerted a protective role in reducing the risk of coronary atherosclerosis (OR = 0.96, 95%CI: 0.93, 1.00, P = 0.049), congestive heart failure (OR = 0.64, 95%CI: 0.42, 0.99, P = 0.043), occlusion of cerebral arteries (OR = 0.93, 95%CI: 0.87, 1.00, P = 0.044) and peripheral vascular disease (OR = 0.60, 95%CI: 0.38, 0.94, P = 0.025). Furthermore, the combination of uric acid-lowering therapy (e.g. xanthine oxidase inhibitors) with antihypertensive treatment (e.g. calcium channel blockers) exerted additive effects and was associated with a 6%, 8%, 8%, 10% reduction in risk of coronary atherosclerosis, heart failure, occlusion of cerebral arteries and peripheral vascular disease, respectively. Our findings support a role of elevated uric acid levels in advancing cardiovascular dysfunction and identify potential repurposing opportunities for uric acid-lowering drugs in cardiovascular treatment.
Collapse
Affiliation(s)
- Lijuan Wang
- Centre for Global Health, Usher Institute, The University of Edinburgh, Edinburgh, UK
| | - Ines Mesa-Eguiagaray
- Centre for Global Health, Usher Institute, The University of Edinburgh, Edinburgh, UK
| | - Harry Campbell
- Centre for Global Health, Usher Institute, The University of Edinburgh, Edinburgh, UK
| | - James F Wilson
- Centre for Global Health, Usher Institute, The University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Veronique Vitart
- MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Xue Li
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Evropi Theodoratou
- Centre for Global Health, Usher Institute, The University of Edinburgh, Edinburgh, UK.
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
75
|
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. Transl Psychiatry 2024; 14:311. [PMID: 39069519 PMCID: PMC11284231 DOI: 10.1038/s41398-024-03011-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/28/2024] [Accepted: 07/04/2024] [Indexed: 07/30/2024] Open
Abstract
Tics are a common feature of early-onset neurodevelopmental disorders, characterized by involuntary and repetitive movements or sounds. Despite affecting up to 2% of children and having a genetic contribution, the underlying causes remain poorly understood. In this study, we leverage dense phenotype information to identify features (i.e., symptoms and comorbid diagnoses) of tic disorders within the context of a clinical biobank. Using de-identified electronic health records (EHRs), we identified individuals with tic disorder diagnosis codes. We performed a phenome-wide association study (PheWAS) to identify the EHR features enriched in tic cases versus controls (n = 1406 and 7030; respectively) and found highly comorbid neuropsychiatric phenotypes, including: obsessive-compulsive disorder, attention-deficit/hyperactivity disorder, autism spectrum disorder, and anxiety (p < 7.396 × 10-5). These features (among others) were then used to generate a phenotype risk score (PheRS) for tic disorder, which was applied across an independent set of 90,051 individuals. A gold standard set of tic disorder cases identified by an EHR algorithm and confirmed by clinician chart review was then used to validate the tic disorder PheRS; the tic disorder PheRS was significantly higher among clinician-validated tic cases versus non-cases (p = 4.787 × 10-151; β = 1.68; SE = 0.06). Our findings provide support for the use of large-scale medical databases to better understand phenotypically complex and underdiagnosed conditions, such as tic disorders.
Collapse
Affiliation(s)
- Tyne W Miller-Fleming
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, TN, Nashville, 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, Nashville, 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, Nashville, USA.
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Biomedical Informatics, Vanderbilt University Medical Center, TN, Nashville, USA.
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, TN, Nashville, USA.
- Department of Molecular Physiology and Biophysics, Vanderbilt University, TN, Nashville, USA.
| |
Collapse
|
76
|
Manipur I, Reales G, Sul JH, Shin MK, Longerich S, Cortes A, Wallace C. CoPheScan: phenome-wide association studies accounting for linkage disequilibrium. Nat Commun 2024; 15:5862. [PMID: 38997278 PMCID: PMC11245513 DOI: 10.1038/s41467-024-49990-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 06/25/2024] [Indexed: 07/14/2024] Open
Abstract
Phenome-wide association studies (PheWAS) facilitate the discovery of associations between a single genetic variant with multiple phenotypes. For variants which impact a specific protein, this can help identify additional therapeutic indications or on-target side effects of intervening on that protein. However, PheWAS is restricted by an inability to distinguish confounding due to linkage disequilibrium (LD) from true pleiotropy. Here we describe CoPheScan (Coloc adapted Phenome-wide Scan), a Bayesian approach that enables an intuitive and systematic exploration of causal associations while simultaneously addressing LD confounding. We demonstrate its performance through simulation, showing considerably better control of false positive rates than a conventional approach not accounting for LD. We used CoPheScan to perform PheWAS of protein-truncating variants and fine-mapped variants from disease and pQTL studies, in 2275 disease phenotypes from the UK Biobank. Our results identify the complexity of known pleiotropic genes such as APOE, and suggest a new causal role for TGM3 in skin cancer.
Collapse
Affiliation(s)
- Ichcha Manipur
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK.
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 2QQ, UK.
| | - Guillermo Reales
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 2QQ, UK
| | | | | | | | - Adrian Cortes
- Human Genetics and Genomics, GSK, Heidelberg, 69117, Germany
| | - Chris Wallace
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 2QQ, UK
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
77
|
Meng W, Xu J, Huang Y, Wang C, Song Q, Ma A, Song L, Bian J, Ma Q, Yin R. Autoencoder to Identify Sex-Specific Sub-phenotypes in Alzheimer's Disease Progression Using Longitudinal Electronic Health Records. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.07.24310055. [PMID: 39040206 PMCID: PMC11261930 DOI: 10.1101/2024.07.07.24310055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Alzheimer's Disease (AD) is a complex neurodegenerative disorder significantly influenced by sex differences, with approximately two-thirds of AD patients being women. Characterizing the sex-specific AD progression and identifying its progression trajectory is a crucial step to developing effective risk stratification and prevention strategies. In this study, we developed an autoencoder to uncover sex-specific sub-phenotypes in AD progression leveraging longitudinal electronic health record (EHR) data from OneFlorida+ Clinical Research Consortium. Specifically, we first constructed temporal patient representation using longitudinal EHRs from a sex-stratified AD cohort. We used a long short-term memory (LSTM)-based autoencoder to extract and generate latent representation embeddings from sequential clinical records of patients. We then applied hierarchical agglomerative clustering to the learned representations, grouping patients based on their progression sub-phenotypes. The experimental results show we successfully identified five primary sex-based AD sub-phenotypes with corresponding progression pathways with high confidence. These sex-specific sub-phenotypes not only illustrated distinct AD progression patterns but also revealed differences in clinical characteristics and comorbidities between females and males in AD development. These findings could provide valuable insights for advancing personalized AD intervention and treatment strategies.
Collapse
Affiliation(s)
- Weimin Meng
- Department of Health Outcomes &Biomedical Informatics, University of Florida, Gainesville, FL, 32610, USA
| | - Jie Xu
- Department of Health Outcomes &Biomedical Informatics, University of Florida, Gainesville, FL, 32610, USA
| | - Yu Huang
- Department of Health Outcomes &Biomedical Informatics, University of Florida, Gainesville, FL, 32610, USA
| | - Cankun Wang
- Department of Biomedical Informatics, Ohio State University, Columbus, OH, 43210, USA
| | - Qianqian Song
- Department of Health Outcomes &Biomedical Informatics, University of Florida, Gainesville, FL, 32610, USA
| | - Anjun Ma
- Department of Biomedical Informatics, Ohio State University, Columbus, OH, 43210, USA
| | - Lixin Song
- School of Nursing, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Jiang Bian
- Department of Health Outcomes &Biomedical Informatics, University of Florida, Gainesville, FL, 32610, USA
| | - Qin Ma
- Department of Biomedical Informatics, Ohio State University, Columbus, OH, 43210, USA
| | - Rui Yin
- Department of Health Outcomes &Biomedical Informatics, University of Florida, Gainesville, FL, 32610, USA
| |
Collapse
|
78
|
Allaire P, Elsayed NS, Berg RL, Rose W, Shukla SK. Phenome-wide association study identifies new clinical phenotypes associated with Staphylococcus aureus infections. PLoS One 2024; 19:e0303395. [PMID: 38968223 PMCID: PMC11226111 DOI: 10.1371/journal.pone.0303395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 04/23/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Phenome-Wide Association study (PheWAS) is a powerful tool designed to systematically screen clinical observations derived from medical records (phenotypes) for association with a variable of interest. Despite their usefulness, no systematic screening of phenotypes associated with Staphylococcus aureus infections (SAIs) has been done leaving potential novel risk factors or complications undiscovered. METHOD AND COHORTS We tailored the PheWAS approach into a two-stage screening procedure to identify novel phenotypes correlating with SAIs. The first stage screened for co-occurrence of SAIs with other phenotypes within medical records. In the second stage, significant findings were examined for the correlations between their age of onset with that of SAIs. The PheWAS was implemented using the medical records of 754,401 patients from the Marshfield Clinic Health System. Any novel associations discovered were subsequently validated using datasets from TriNetX and All of Us, encompassing 109,884,571 and 118,538 patients respectively. RESULTS Forty-one phenotypes met the significance criteria of a p-value < 3.64e-5 and odds ratios of > 5. Out of these, we classified 23 associations either as risk factors or as complications of SAIs. Three novel associations were discovered and classified either as a risk (long-term use of aspirin) or complications (iron deficiency anemia and anemia of chronic disease). All novel associations were replicated in the TriNetX cohort. In the All of Us cohort, anemia of chronic disease was replicated according to our significance criteria. CONCLUSIONS The PheWAS of SAIs expands our understanding of SAIs interacting phenotypes. Additionally, the novel two-stage PheWAS approach developed in this study can be applied to examine other disease-disease interactions of interest. Due to the possibility of bias inherent in observational data, the findings of this study require further investigation.
Collapse
Affiliation(s)
- Patrick Allaire
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, Marshfield, Wisconsin, United States of America
| | - Noha S. Elsayed
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, Marshfield, Wisconsin, United States of America
| | - Richard L. Berg
- Research Computing and Analytics, Marshfield Clinic Research Institute, Marshfield, Wisconsin, United States of America
| | - Warren Rose
- School of Pharmacy, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Sanjay K. Shukla
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, Marshfield, Wisconsin, United States of America
- Computational and Informatics in Biology and Medicine Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
79
|
Gilliland T, Dron JS, Selvaraj MS, Trinder M, Paruchuri K, Urbut SM, Haidermota S, Bernardo R, Uddin MM, Honigberg MC, Peloso GM, Natarajan P. Genetic Architecture and Clinical Outcomes of Combined Lipid Disturbances. Circ Res 2024; 135:265-276. [PMID: 38828614 PMCID: PMC11223949 DOI: 10.1161/circresaha.123.323973] [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: 11/10/2023] [Accepted: 05/20/2024] [Indexed: 06/05/2024]
Abstract
BACKGROUND Dyslipoproteinemia often involves simultaneous derangements of multiple lipid traits. We aimed to evaluate the phenotypic and genetic characteristics of combined lipid disturbances in a general population-based cohort. METHODS Among UK Biobank participants without prevalent coronary artery disease, we used blood lipid and apolipoprotein B concentrations to ascribe individuals into 1 of 6 reproducible and mutually exclusive dyslipoproteinemia subtypes. Incident coronary artery disease risk was estimated for each subtype using Cox proportional hazards models. Phenome-wide analyses and genome-wide association studies were performed for each subtype, followed by in silico causal gene prioritization and heritability analyses. Additionally, the prevalence of disruptive variants in causal genes for Mendelian lipid disorders was assessed using whole-exome sequence data. RESULTS Among 450 636 UK Biobank participants: 63 (0.01%) had chylomicronemia; 40 005 (8.9%) had hypercholesterolemia; 94 785 (21.0%) had combined hyperlipidemia; 13 998 (3.1%) had remnant hypercholesterolemia; 110 389 (24.5%) had hypertriglyceridemia; and 49 (0.01%) had mixed hypertriglyceridemia and hypercholesterolemia. Over a median (interquartile range) follow-up of 11.1 (10.4-11.8) years, incident coronary artery disease risk varied across subtypes, with combined hyperlipidemia exhibiting the largest hazard (hazard ratio, 1.92 [95% CI, 1.84-2.01]; P=2×10-16), even when accounting for non-HDL-C (hazard ratio, 1.45 [95% CI, 1.30-1.60]; P=2.6×10-12). Genome-wide association studies revealed 250 loci significantly associated with dyslipoproteinemia subtypes, of which 72 (28.8%) were not detected in prior single lipid trait genome-wide association studies. Mendelian lipid variant carriers were rare (2.0%) among individuals with dyslipoproteinemia, but polygenic heritability was high, ranging from 23% for remnant hypercholesterolemia to 54% for combined hyperlipidemia. CONCLUSIONS Simultaneous assessment of multiple lipid derangements revealed nuanced differences in coronary artery disease risk and genetic architectures across dyslipoproteinemia subtypes. These findings highlight the importance of looking beyond single lipid traits to better understand combined lipid and lipoprotein phenotypes and implications for disease risk.
Collapse
Affiliation(s)
- Thomas Gilliland
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Jacqueline S. Dron
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
| | - Margaret Sunitha Selvaraj
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Mark Trinder
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC
| | - Kaavya Paruchuri
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Sarah M. Urbut
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Sara Haidermota
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA
| | - Rachel Bernardo
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA
| | - Md Mesbah Uddin
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA
| | - Michael C. Honigberg
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Gina M. Peloso
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Pradeep Natarajan
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics and the Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA
- Department of Medicine, Harvard Medical School, Boston, MA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
80
|
Tang AS, Woldemariam SR, Miramontes S, Norgeot B, Oskotsky TT, Sirota M. Harnessing EHR data for health research. Nat Med 2024; 30:1847-1855. [PMID: 38965433 DOI: 10.1038/s41591-024-03074-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/17/2024] [Indexed: 07/06/2024]
Abstract
With the increasing availability of rich, longitudinal, real-world clinical data recorded in electronic health records (EHRs) for millions of patients, there is a growing interest in leveraging these records to improve the understanding of human health and disease and translate these insights into clinical applications. However, there is also a need to consider the limitations of these data due to various biases and to understand the impact of missing information. Recognizing and addressing these limitations can inform the design and interpretation of EHR-based informatics studies that avoid confusing or incorrect conclusions, particularly when applied to population or precision medicine. Here we discuss key considerations in the design, implementation and interpretation of EHR-based informatics studies, drawing from examples in the literature across hypothesis generation, hypothesis testing and machine learning applications. We outline the growing opportunities for EHR-based informatics studies, including association studies and predictive modeling, enabled by evolving AI capabilities-while addressing limitations and potential pitfalls to avoid.
Collapse
Affiliation(s)
- Alice S Tang
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Sarah R Woldemariam
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Silvia Miramontes
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | | | - Tomiko T Oskotsky
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA.
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
81
|
Martínez-Magaña JJ, Hurtado-Soriano J, Rivero-Segura NA, Montalvo-Ortiz JL, Garcia-delaTorre P, Becerril-Rojas K, Gomez-Verjan JC. Towards a Novel Frontier in the Use of Epigenetic Clocks in Epidemiology. Arch Med Res 2024; 55:103033. [PMID: 38955096 DOI: 10.1016/j.arcmed.2024.103033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/10/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024]
Abstract
Health problems associated with aging are a major public health concern for the future. Aging is a complex process with wide intervariability among individuals. Therefore, there is a need for innovative public health strategies that target factors associated with aging and the development of tools to assess the effectiveness of these strategies accurately. Novel approaches to measure biological age, such as epigenetic clocks, have become relevant. These clocks use non-sequential variable information from the genome and employ mathematical algorithms to estimate biological age based on DNA methylation levels. Therefore, in the present study, we comprehensively review the current status of the epigenetic clocks and their associations across the human phenome. We emphasize the potential utility of these tools in an epidemiological context, particularly in evaluating the impact of public health interventions focused on promoting healthy aging. Our review describes associations between epigenetic clocks and multiple traits across the life and health span. Additionally, we highlighted the evolution of studies beyond mere associations to establish causal mechanisms between epigenetic age and disease. We explored the application of epigenetic clocks to measure the efficacy of interventions focusing on rejuvenation.
Collapse
Affiliation(s)
- José Jaime Martínez-Magaña
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; U.S. Department of Veterans Affairs National Center for Post-Traumatic Stress Disorder, Clinical Neuroscience Division, West Haven, CT, USA; VA Connecticut Healthcare System, West Haven, CT, USA
| | | | | | - Janitza L Montalvo-Ortiz
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; U.S. Department of Veterans Affairs National Center for Post-Traumatic Stress Disorder, Clinical Neuroscience Division, West Haven, CT, USA; VA Connecticut Healthcare System, West Haven, CT, USA
| | - Paola Garcia-delaTorre
- Unidad de Investigación Epidemiológica y en Servicios de Salud, Área de Envejecimiento, Centro Médico Nacional, Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | | | | |
Collapse
|
82
|
Salvatore M, Kundu R, Shi X, Friese CR, Lee S, Fritsche LG, Mondul AM, Hanauer D, Pearce CL, Mukherjee B. To weight or not to weight? The effect of selection bias in 3 large electronic health record-linked biobanks and recommendations for practice. J Am Med Inform Assoc 2024; 31:1479-1492. [PMID: 38742457 PMCID: PMC11187425 DOI: 10.1093/jamia/ocae098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/14/2024] [Accepted: 04/18/2024] [Indexed: 05/16/2024] Open
Abstract
OBJECTIVES To develop recommendations regarding the use of weights to reduce selection bias for commonly performed analyses using electronic health record (EHR)-linked biobank data. MATERIALS AND METHODS We mapped diagnosis (ICD code) data to standardized phecodes from 3 EHR-linked biobanks with varying recruitment strategies: All of Us (AOU; n = 244 071), Michigan Genomics Initiative (MGI; n = 81 243), and UK Biobank (UKB; n = 401 167). Using 2019 National Health Interview Survey data, we constructed selection weights for AOU and MGI to represent the US adult population more. We used weights previously developed for UKB to represent the UKB-eligible population. We conducted 4 common analyses comparing unweighted and weighted results. RESULTS For AOU and MGI, estimated phecode prevalences decreased after weighting (weighted-unweighted median phecode prevalence ratio [MPR]: 0.82 and 0.61), while UKB estimates increased (MPR: 1.06). Weighting minimally impacted latent phenome dimensionality estimation. Comparing weighted versus unweighted phenome-wide association study for colorectal cancer, the strongest associations remained unaltered, with considerable overlap in significant hits. Weighting affected the estimated log-odds ratio for sex and colorectal cancer to align more closely with national registry-based estimates. DISCUSSION Weighting had a limited impact on dimensionality estimation and large-scale hypothesis testing but impacted prevalence and association estimation. When interested in estimating effect size, specific signals from untargeted association analyses should be followed up by weighted analysis. CONCLUSION EHR-linked biobanks should report recruitment and selection mechanisms and provide selection weights with defined target populations. Researchers should consider their intended estimands, specify source and target populations, and weight EHR-linked biobank analyses accordingly.
Collapse
Affiliation(s)
- Maxwell Salvatore
- Department of Epidemiology, University of Michigan, Ann Arbor, MI 48109-2029, United States
- Center for Precision Health Data Science, Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109-2029, United States
| | - Ritoban Kundu
- Center for Precision Health Data Science, Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109-2029, United States
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109-2029, United States
| | - Xu Shi
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109-2029, United States
| | - Christopher R Friese
- Rogel Cancer Center, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109-2029, United States
- Center for Improving Patient and Population Health, School of Nursing, University of Michigan, Ann Arbor, MI 48109-2029, United States
- Department of Health Management and Policy, University of Michigan, Ann Arbor, MI 48109-2029, United States
| | - Seunggeun Lee
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109-2029, United States
- Graduate School of Data Science, Seoul National University, Gwanak-gu, Seoul, Republic of Korea
| | - Lars G Fritsche
- Center for Precision Health Data Science, Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109-2029, United States
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109-2029, United States
- Rogel Cancer Center, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109-2029, United States
| | - Alison M Mondul
- Department of Epidemiology, University of Michigan, Ann Arbor, MI 48109-2029, United States
- Rogel Cancer Center, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109-2029, United States
| | - David Hanauer
- Department of Learning Health Sciences, University of Michigan Medical School, Ann Arbor, MI 48109-2054, United States
| | - Celeste Leigh Pearce
- Department of Epidemiology, University of Michigan, Ann Arbor, MI 48109-2029, United States
- Rogel Cancer Center, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109-2029, United States
| | - Bhramar Mukherjee
- Department of Epidemiology, University of Michigan, Ann Arbor, MI 48109-2029, United States
- Center for Precision Health Data Science, Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109-2029, United States
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109-2029, United States
| |
Collapse
|
83
|
Dalal T, Patel CJ. PYPE: A pipeline for phenome-wide association and Mendelian randomization in investigator-driven biobank scale analysis. PATTERNS (NEW YORK, N.Y.) 2024; 5:100982. [PMID: 39005490 PMCID: PMC11240175 DOI: 10.1016/j.patter.2024.100982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/30/2023] [Accepted: 04/08/2024] [Indexed: 07/16/2024]
Abstract
Phenome-wide association studies (PheWASs) serve as a way of documenting the relationship between genotypes and multiple phenotypes, helping to uncover unexplored genotype-phenotype associations (known as pleiotropy). Secondly, Mendelian randomization (MR) can be harnessed to make causal statements about a pair of phenotypes by comparing their genetic architecture. Thus, approaches that automate both PheWASs and MR can enhance biobank-scale analyses, circumventing the need for multiple tools by providing a comprehensive, end-to-end tool to drive scientific discovery. To this end, we present PYPE, a Python pipeline for running, visualizing, and interpreting PheWASs. PYPE utilizes input genotype or phenotype files to automatically estimate associations between the chosen independent variables and phenotypes. PYPE can also produce a variety of visualizations and can be used to identify nearby genes and functional consequences of significant associations. Finally, PYPE can identify possible causal relationships between phenotypes using MR under a variety of causal effect modeling scenarios.
Collapse
Affiliation(s)
- Taykhoom Dalal
- Harvard Medical School Department of Biomedical Informatics, Boston, MA 02115, USA
| | - Chirag J Patel
- Harvard Medical School Department of Biomedical Informatics, Boston, MA 02115, USA
| |
Collapse
|
84
|
Coon H, Shabalin A, DiBlasi E, Monson ET, Han S, Kaufman EA, Chen D, Kious B, Molina N, Yu Z, Staley M, Crockett DK, Colbert SM, Mullins N, Bakian AV, Docherty AR, Keeshin B. Absence of nonfatal suicidal behavior preceding suicide death reveals differences in clinical risks. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.05.24308493. [PMID: 38883733 PMCID: PMC11177925 DOI: 10.1101/2024.06.05.24308493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Nonfatal suicidality is the most robust predictor of suicide death. However, only ~10% of those who survive an attempt go on to die by suicide. Moreover, ~50% of suicide deaths occur in the absence of prior known attempts, suggesting risks other than nonfatal suicide attempt need to be identified. We studied data from 4,000 population-ascertained suicide deaths and 26,191 population controls to improve understanding of risks leading to suicide death. This study included 2,253 suicide deaths and 3,375 controls with evidence of nonfatal suicidality (SUI_SI/SB and CTL_SI/SB) from diagnostic codes and natural language processing of electronic health records notes. Characteristics of these groups were compared to 1,669 suicides with no prior nonfatal SI/SB (SUI_None) and 22,816 controls with no lifetime suicidality (CTL_None). The SUI_None and CTL_None groups had fewer diagnoses and were older than SUI_SI/SB and CTL_SI/SB. Mental health diagnoses were far less common in both the SUI_None and CTL_None groups; mental health problems were less associated with suicide death than with presence of SI/SB. Physical health diagnoses were conversely more often associated with risk of suicide death than with presence of SI/SB. Pending replication, results indicate highly significant clinical differences among suicide deaths with versus without prior nonfatal SI/SB.
Collapse
Affiliation(s)
- Hilary Coon
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Andrey Shabalin
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Emily DiBlasi
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Eric T. Monson
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Seonggyun Han
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Erin A. Kaufman
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Danli Chen
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Brent Kious
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | - Zhe Yu
- Pedigree & Population Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Michael Staley
- Utah State Office of the Medical Examiner, Utah Department of Health and Human Services, Salt Lake City, UT
| | | | - Sarah M. Colbert
- Department of Psychiatry, Mount Sinai School of Medicine, New York, NY
| | - Niamh Mullins
- Department of Psychiatry, Mount Sinai School of Medicine, New York, NY
| | - Amanda V. Bakian
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Anna R. Docherty
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Brooks Keeshin
- Department of Psychiatry & Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
- Department of Pediatrics, University of Utah, Salt Lake City, UT
- Primary Children’s Hospital Center for Safe and Healthy Families, Salt Lake City, UT
| |
Collapse
|
85
|
Lee H, Kennedy CJ, Tu A, Restivo J, Liu CH, Naslund JA, Patel V, Choi KW, Smoller JW. Patterns and correlates of mental healthcare utilization during the COVID-19 pandemic among individuals with pre-existing mental disorder. PLoS One 2024; 19:e0303079. [PMID: 38833458 PMCID: PMC11149861 DOI: 10.1371/journal.pone.0303079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 04/19/2024] [Indexed: 06/06/2024] Open
Abstract
How did mental healthcare utilization change during the COVID-19 pandemic period among individuals with pre-existing mental disorder? Understanding utilization patterns of these at-risk individuals and identifying those most likely to exhibit increased utilization could improve patient stratification and efficient delivery of mental health services. This study leveraged large-scale electronic health record (EHR) data to describe mental healthcare utilization patterns among individuals with pre-existing mental disorder before and during the COVID-19 pandemic and identify correlates of high mental healthcare utilization. Using EHR data from a large healthcare system in Massachusetts, we identified three "pre-existing mental disorder" groups (PMD) based on having a documented mental disorder diagnosis within the 6 months prior to the March 2020 lockdown, related to: (1) stress-related disorders (e.g., depression, anxiety) (N = 115,849), (2) serious mental illness (e.g., schizophrenia, bipolar disorders) (N = 11,530), or (3) compulsive behavior disorders (e.g., eating disorder, OCD) (N = 5,893). We also identified a "historical comparison" group (HC) for each PMD (N = 113,604, 11,758, and 5,387, respectively) from the previous year (2019). We assessed the monthly number of mental healthcare visits from March 13 to December 31 for PMDs in 2020 and HCs in 2019. Phenome-wide association analyses (PheWAS) were used to identify clinical correlates of high mental healthcare utilization. We found the overall number of mental healthcare visits per patient during the pandemic period in 2020 was 10-12% higher than in 2019. The majority of increased visits was driven by a subset of high mental healthcare utilizers (top decile). PheWAS results indicated that correlates of high utilization (prior mental disorders, chronic pain, insomnia, viral hepatitis C, etc.) were largely similar before and during the pandemic, though several conditions (e.g., back pain) were associated with high utilization only during the pandemic. Limitations included that we were not able to examine other risk factors previously shown to influence mental health during the pandemic (e.g., social support, discrimination) due to lack of social determinants of health information in EHR data. Mental healthcare utilization among patients with pre-existing mental disorder increased overall during the pandemic, likely due to expanded access to telemedicine. Given that clinical correlates of high mental healthcare utilization in a major hospital system were largely similar before and during the COVID-19 pandemic, resource stratification based on known risk factor profiles may aid hospitals in responding to heightened mental healthcare needs during a pandemic.
Collapse
Affiliation(s)
- Hyunjoon Lee
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Psychiatry, Center for Precision Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Chris J. Kennedy
- Department of Psychiatry, Center for Precision Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Allison Tu
- Harvard College, Cambridge, Massachusetts, United States of America
| | - Juliana Restivo
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Cindy H. Liu
- Departments of Pediatrics and Psychiatry, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - John A. Naslund
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Vikram Patel
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Global Health and Population, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Karmel W. Choi
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Psychiatry, Center for Precision Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Jordan W. Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Psychiatry, Center for Precision Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| |
Collapse
|
86
|
Chiou JS, Lin YJ, Chang CYY, Liang WM, Liu TY, Yang JS, Chou CH, Lu HF, Chiu ML, Lin TH, Liao CC, Huang SM, Chou IC, Li TM, Huang PY, Chien TS, Chen HR, Tsai FJ. Menarche-a journey into womanhood: age at menarche and health-related outcomes in East Asians. Hum Reprod 2024; 39:1336-1350. [PMID: 38527428 DOI: 10.1093/humrep/deae060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 02/22/2024] [Indexed: 03/27/2024] Open
Abstract
STUDY QUESTION Are there associations of age at menarche (AAM) with health-related outcomes in East Asians? SUMMARY ANSWER AAM is associated with osteoporosis, Type 2 diabetes (T2D), glaucoma, and uterine fibroids, as demonstrated through observational studies, polygenic risk scores, genetic correlations, and Mendelian randomization (MR), with additional findings indicating a causal effect of BMI and T2D on earlier AAM. WHAT IS KNOWN ALREADY Puberty timing is linked to adult disease risk, but research predominantly focuses on European populations, with limited studies in other groups. STUDY DESIGN, SIZE, DURATION We performed an AAM genome-wide association study (GWAS) with 57 890 Han Taiwanese females and examined the association between AAM and 154 disease outcomes using the Taiwanese database. Additionally, we examined genetic correlations between AAM and 113 diseases and 67 phenotypes using Japanese GWAS summary statistics. PARTICIPANTS/MATERIALS, SETTING, METHODS We performed AAM GWAS and gene-based GWAS studies to obtain summary statistics and identify potential AAM-related genes. We applied phenotype, polygenic risk scores, and genetic correlation analyses of AAM to explore health-related outcomes, using multivariate regression and linkage disequilibrium score regression analyses. We also explored potential bidirectional causal relationships between AAM and related outcomes through univariable and multivariable MR analyses. MAIN RESULTS AND THE ROLE OF CHANCE Fifteen lead single-nucleotide polymorphisms and 24 distinct genes were associated with AAM in Taiwan. AAM was genetically associated with later menarche and menopause, greater height, increased osteoporosis risk, but lower BMI, and reduced risks of T2D, glaucoma, and uterine fibroids in East Asians. Bidirectional MR analyses indicated that higher BMI/T2D causally leads to earlier AAM. LIMITATIONS, REASONS FOR CAUTION Our findings were specific to Han Taiwanese individuals, with genetic correlation analyses conducted in East Asians. Further research in other ethnic groups is necessary. WIDER IMPLICATIONS OF THE FINDINGS Our study provides insights into the genetic architecture of AAM and its health-related outcomes in East Asians, highlighting causal links between BMI/T2D and earlier AAM, which may suggest potential prevention strategies for early puberty. STUDY FUNDING/COMPETING INTEREST(S) The work was supported by China Medical University, Taiwan (CMU110-S-17, CMU110-S-24, CMU110-MF-49, CMU111-SR-158, CMU111-MF-105, CMU111-MF-21, CMU111-S-35, CMU112-SR-30, and CMU112-MF-101), the China Medical University Hospital, Taiwan (DMR-111-062, DMR-111-153, DMR-112-042, DMR-113-038, and DMR-113-103), and the Ministry of Science and Technology, Taiwan (MOST 111-2314-B-039-063-MY3, MOST 111-2314-B-039-064-MY3, MOST 111-2410-H-039-002-MY3, and NSTC 112-2813-C-039-036-B). The funders had no influence on the data collection, analyses, or conclusions of the study. No conflict of interests to declare. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Jian-Shiun Chiou
- PhD Program for Health Science and Industry, College of Health Care, China Medical University, Taichung, Taiwan
- Department of Health Services Administration, China Medical University, Taichung, Taiwan
| | - Ying-Ju Lin
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Cherry Yin-Yi Chang
- Division of Minimal Invasive Endoscopy Surgery, Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Wen-Miin Liang
- Department of Health Services Administration, China Medical University, Taichung, Taiwan
| | - Ting-Yuan Liu
- Million-Person Precision Medicine Initiative, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Jai-Sing Yang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Chen-Hsing Chou
- PhD Program for Health Science and Industry, College of Health Care, China Medical University, Taichung, Taiwan
- Department of Health Services Administration, China Medical University, Taichung, Taiwan
| | - Hsing-Fang Lu
- Million-Person Precision Medicine Initiative, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Mu-Lin Chiu
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Ting-Hsu Lin
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Chiu-Chu Liao
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Shao-Mei Huang
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - I-Ching Chou
- Department of Pediatrics, China Medical University Children's Hospital, Taichung, Taiwan
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Te-Mao Li
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Peng-Yan Huang
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Tzu-Shun Chien
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Hou-Ren Chen
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Fuu-Jen Tsai
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Pediatrics, China Medical University Children's Hospital, Taichung, Taiwan
- Division of Medical Genetics, China Medical University Children's Hospital, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
87
|
Castaño A, Heitner SB, Masri A, Huda A, Calambur V, Bruno M, Schumacher J, Emir B, Isherwood C, Shah SJ. EstimATTR: A Simplified, Machine-Learning-Based Tool to Predict the Risk of Wild-Type Transthyretin Amyloid Cardiomyopathy. J Card Fail 2024; 30:778-787. [PMID: 38065306 DOI: 10.1016/j.cardfail.2023.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 11/14/2023] [Accepted: 11/14/2023] [Indexed: 02/02/2024]
Abstract
BACKGROUND Wild-type transthyretin amyloid cardiomyopathy (ATTRwt-CM), an increasingly recognized cause of heart failure (HF), often remains undiagnosed until later stages of the disease. METHODS AND RESULTS A previously developed machine learning algorithm was simplified to create a random forest model based on 11 selected phenotypes predictive of ATTRwt-CM to estimate ATTRwt-CM risk in hypothetical patient scenarios. Using U.S. medical claims datasets (IQVIA), International Classification of Diseases codes were extracted to identify a training cohort of patients with ATTRwt-CM (cases) or nonamyloid HF (controls). After assessment in a 20% test sample of the training cohort, model performance was validated in cohorts of patients with International Classification of Diseases codes for ATTRwt-CM or cardiac amyloidosis vs nonamyloid HF derived from medical claims (IQVIA) or electronic health records (Optum). The simplified model performed well in identifying patients with ATTRwt-CM vs nonamyloid HF in the test sample, with an accuracy of 74%, sensitivity of 77%, specificity of 72%, and area under the curve of 0.82; robust performance was also observed in the validation cohorts. CONCLUSIONS This simplified machine learning model accurately estimated the empirical probability of ATTRwt-CM in administrative datasets, suggesting it may serve as an easily implementable tool for clinical assessment of patient risk for ATTRwt-CM in the clinical setting. BRIEF LAY SUMMARY Wild-type transthyretin amyloid cardiomyopathy (ATTRwt-CM for short) is a frequently overlooked cause of heart failure. Finding ATTRwt-CM early is important because the disease can worsen rapidly without treatment. Researchers developed a computer program that predicts the risk of ATTRwt-CM in patients with heart failure. In this study, the program was used to check for 11 medical conditions linked to ATTRwt-CM in the medical claims records of patients with heart failure. The program was 74% accurate in identifying ATTRwt-CM in patients with heart failure and was then used to develop an educational online tool for doctors (the wtATTR-CM estimATTR).
Collapse
Affiliation(s)
| | - Stephen B Heitner
- The Amyloidosis Center, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Ahmad Masri
- The Amyloidosis Center, Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | | | | | | | | | | | | | - Sanjiv J Shah
- Northwestern University Feinberg School of Medicine, 633 N. St. Clair St., Chicago, Illinois.
| |
Collapse
|
88
|
Oikonomou EK, Holste G, Yuan N, Coppi A, McNamara RL, Haynes NA, Vora AN, Velazquez EJ, Li F, Menon V, Kapadia SR, Gill TM, Nadkarni GN, Krumholz HM, Wang Z, Ouyang D, Khera R. A Multimodal Video-Based AI Biomarker for Aortic Stenosis Development and Progression. JAMA Cardiol 2024; 9:534-544. [PMID: 38581644 PMCID: PMC10999005 DOI: 10.1001/jamacardio.2024.0595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 02/27/2024] [Indexed: 04/08/2024]
Abstract
Importance Aortic stenosis (AS) is a major public health challenge with a growing therapeutic landscape, but current biomarkers do not inform personalized screening and follow-up. A video-based artificial intelligence (AI) biomarker (Digital AS Severity index [DASSi]) can detect severe AS using single-view long-axis echocardiography without Doppler characterization. Objective To deploy DASSi to patients with no AS or with mild or moderate AS at baseline to identify AS development and progression. Design, Setting, and Participants This is a cohort study that examined 2 cohorts of patients without severe AS undergoing echocardiography in the Yale New Haven Health System (YNHHS; 2015-2021) and Cedars-Sinai Medical Center (CSMC; 2018-2019). A novel computational pipeline for the cross-modal translation of DASSi into cardiac magnetic resonance (CMR) imaging was further developed in the UK Biobank. Analyses were performed between August 2023 and February 2024. Exposure DASSi (range, 0-1) derived from AI applied to echocardiography and CMR videos. Main Outcomes and Measures Annualized change in peak aortic valve velocity (AV-Vmax) and late (>6 months) aortic valve replacement (AVR). Results A total of 12 599 participants were included in the echocardiographic study (YNHHS: n = 8798; median [IQR] age, 71 [60-80] years; 4250 [48.3%] women; median [IQR] follow-up, 4.1 [2.4-5.4] years; and CSMC: n = 3801; median [IQR] age, 67 [54-78] years; 1685 [44.3%] women; median [IQR] follow-up, 3.4 [2.8-3.9] years). Higher baseline DASSi was associated with faster progression in AV-Vmax (per 0.1 DASSi increment: YNHHS, 0.033 m/s per year [95% CI, 0.028-0.038] among 5483 participants; CSMC, 0.082 m/s per year [95% CI, 0.053-0.111] among 1292 participants), with values of 0.2 or greater associated with a 4- to 5-fold higher AVR risk than values less than 0.2 (YNHHS: 715 events; adjusted hazard ratio [HR], 4.97 [95% CI, 2.71-5.82]; CSMC: 56 events; adjusted HR, 4.04 [95% CI, 0.92-17.70]), independent of age, sex, race, ethnicity, ejection fraction, and AV-Vmax. This was reproduced across 45 474 participants (median [IQR] age, 65 [59-71] years; 23 559 [51.8%] women; median [IQR] follow-up, 2.5 [1.6-3.9] years) undergoing CMR imaging in the UK Biobank (for participants with DASSi ≥0.2 vs those with DASSi <.02, adjusted HR, 11.38 [95% CI, 2.56-50.57]). Saliency maps and phenome-wide association studies supported associations with cardiac structure and function and traditional cardiovascular risk factors. Conclusions and Relevance In this cohort study of patients without severe AS undergoing echocardiography or CMR imaging, a new AI-based video biomarker was independently associated with AS development and progression, enabling opportunistic risk stratification across cardiovascular imaging modalities as well as potential application on handheld devices.
Collapse
Affiliation(s)
- Evangelos K. Oikonomou
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Gregory Holste
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin
| | - Neal Yuan
- Department of Medicine, University of California, San Francisco
- Division of Cardiology, San Francisco Veterans Affairs Medical Center, San Francisco, California
| | - Andreas Coppi
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut
| | - Robert L. McNamara
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Norrisa A. Haynes
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Amit N. Vora
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Eric J. Velazquez
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Fan Li
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
- Center for Methods in Implementation and Prevention Science, Yale School of Public Health, New Haven, Connecticut
| | - Venu Menon
- Heart and Vascular Institute, Department of Cardiovascular Medicine, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Samir R. Kapadia
- Heart and Vascular Institute, Department of Cardiovascular Medicine, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Thomas M. Gill
- Section of Geriatrics, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Girish N. Nadkarni
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Harlan M. Krumholz
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut
| | - Zhangyang Wang
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin
| | - David Ouyang
- Smidt Heart Institute, Department of Cardiology, Cedars-Sinai Medical Center, Los Angeles, California
- Division of Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Rohan Khera
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut
- Section of Biomedical Informatics and Data Science, Yale School of Medicine, New Haven, Connecticut
- Section of Health Informatics, Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
- Associate Editor, JAMA
| |
Collapse
|
89
|
Bagheri M, Bombin A, Shi M, Murthy VL, Shah R, Mosley JD, Ferguson JF. Genotype-based "virtual" metabolomics in a clinical biobank identifies novel metabolite-disease associations. Front Genet 2024; 15:1392622. [PMID: 38812968 PMCID: PMC11133605 DOI: 10.3389/fgene.2024.1392622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/03/2024] [Indexed: 05/31/2024] Open
Abstract
Introduction: Circulating metabolites act as biomarkers of dysregulated metabolism and may inform disease pathophysiology. A portion of the inter-individual variability in circulating metabolites is influenced by common genetic variation. We evaluated whether a genetics-based "virtual" metabolomics approach can identify novel metabolite-disease associations. Methods: We examined the association between polygenic scores for 724 metabolites with 1,247 clinical phenotypes in the BioVU DNA biobank, comprising 57,735 European ancestry and 15,754 African ancestry participants. We applied Mendelian randomization (MR) to probe significant relationships and validated significant MR associations using independent GWAS of candidate phenotypes. Results and Discussion: We found significant associations between 336 metabolites and 168 phenotypes in European ancestry and 107 metabolites and 56 phenotypes in African ancestry. Of these metabolite-disease pairs, MR analyses confirmed associations between 73 metabolites and 53 phenotypes in European ancestry. Of 22 metabolitephenotype pairs evaluated for replication in independent GWAS, 16 were significant (false discovery rate p < 0.05). These included associations between bilirubin and X-21796 with cholelithiasis, phosphatidylcholine (16:0/22:5n3,18:1/20:4) and arachidonate with inflammatory bowel disease and Crohn's disease, and campesterol with coronary artery disease and myocardial infarction. These associations may represent biomarkers or potentially targetable mediators of disease risk.
Collapse
Affiliation(s)
- Minoo Bagheri
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Andrei Bombin
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Mingjian Shi
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Venkatesh L. Murthy
- Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Ravi Shah
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jonathan D. Mosley
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, United States
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jane F. Ferguson
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
90
|
Rämö JT, Gorman B, Weng LC, Jurgens SJ, Singhanetr P, Tieger MG, van Dijk EH, Halladay CW, Wang X, Brinks J, Choi SH, Luo Y, Pyarajan S, Nealon CL, Gorin MB, Wu WC, Sobrin L, Kaarniranta K, Yzer S, Palotie A, Peachey NS, Turunen JA, Boon CJ, Ellinor PT, Iyengar SK, Daly MJ, Rossin EJ. Rare genetic variation in VE-PTP is associated with central serous chorioretinopathy, venous dysfunction and glaucoma. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.08.24307013. [PMID: 38766240 PMCID: PMC11100937 DOI: 10.1101/2024.05.08.24307013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Central serous chorioretinopathy (CSC) is a fluid maculopathy whose etiology is not well understood. Abnormal choroidal veins in CSC patients have been shown to have similarities with varicose veins. To identify potential mechanisms, we analyzed genotype data from 1,477 CSC patients and 455,449 controls in FinnGen. We identified an association for a low-frequency (AF=0.5%) missense variant (rs113791087) in the gene encoding vascular endothelial protein tyrosine phosphatase (VE-PTP) (OR=2.85, P=4.5×10-9). This was confirmed in a meta-analysis of 2,452 CSC patients and 865,767 controls from 4 studies (OR=3.06, P=7.4×10-15). Rs113791087 was associated with a 56% higher prevalence of retinal abnormalities (35.3% vs 22.6%, P=8.0×10-4) in 708 UK Biobank participants and, surprisingly, with varicose veins (OR=1.31, P=2.3×10-11) and glaucoma (OR=0.82, P=6.9×10-9). Predicted loss-of-function variants in VEPTP, though rare in number, were associated with CSC in All of Us (OR=17.10, P=0.018). These findings highlight the significance of VE-PTP in diverse ocular and systemic vascular diseases.
Collapse
Affiliation(s)
- Joel T Rämö
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Massachusetts Eye and Ear, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Bryan Gorman
- Center for Data and Computational Sciences (C-DACS), VA Cooperative Studies Program, VA Boston Healthcare System, Boston, MA, USA
- Booz Allen Hamilton, McLean, VA, USA
| | - Lu-Chen Weng
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sean J Jurgens
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Panisa Singhanetr
- Massachusetts Eye and Ear, Boston, MA, USA
- Mettapracharak Eye Institute, Mettapracharak (Wat Rai Khing) Hospital, Nakhon Pathom, Thailand
| | - Marisa G Tieger
- New England Eye Center, Tufts Medical Center, Boston, MA, USA
| | - Elon Hc van Dijk
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Christopher W Halladay
- Center of Innovation in Long Term Services and Supports, Providence VA Medical Center, Providence, RI, USA
| | - Xin Wang
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joost Brinks
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Seung Hoan Choi
- Department of Biostatistics, Boston University, Boston, MA, USA
| | - Yuyang Luo
- Massachusetts Eye and Ear, Boston, MA, USA
| | - Saiju Pyarajan
- VA Cooperative Studies Program, VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard School of Medicine, Boston, MA, USA
| | - Cari L Nealon
- Eye Clinic, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
| | - Michael B Gorin
- Department of Ophthalmology, David Geffen School of Medicine, Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Wen-Chih Wu
- Section of Cardiology, Medical Service, VA Providence Healthcare System, Providence, RI, USA
| | - Lucia Sobrin
- Harvard Medical School Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
| | - Kai Kaarniranta
- Department of Ophthalmology, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Suzanne Yzer
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
- Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Neal S Peachey
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
- Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Joni A Turunen
- Folkhälsan Research Center, Biomedicum, Helsinki, Finland
- Department of Ophthalmology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Camiel Jf Boon
- Department of Ophthalmology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sudha K Iyengar
- Research Service, VA Northeast Ohio Healthcare System, Cleveland, OH, USA
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Mark J Daly
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Analytic and Translational Genetics Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Elizabeth J Rossin
- Harvard Medical School Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
91
|
Breeyear JH, Mautz BS, Keaton JM, Hellwege JN, Torstenson ES, Liang J, Bray MJ, Giri A, Warren HR, Munroe PB, Velez Edwards DR, Zhu X, Li C, Edwards TL. A new test for trait mean and variance detects unreported loci for blood-pressure variation. Am J Hum Genet 2024; 111:954-965. [PMID: 38614075 PMCID: PMC11080606 DOI: 10.1016/j.ajhg.2024.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/15/2024] Open
Abstract
Variability in quantitative traits has clinical, ecological, and evolutionary significance. Most genetic variants identified for complex quantitative traits have only a detectable effect on the mean of trait. We have developed the mean-variance test (MVtest) to simultaneously model the mean and log-variance of a quantitative trait as functions of genotypes and covariates by using estimating equations. The advantages of MVtest include the facts that it can detect effect modification, that multiple testing can follow conventional thresholds, that it is robust to non-normal outcomes, and that association statistics can be meta-analyzed. In simulations, we show control of type I error of MVtest over several alternatives. We identified 51 and 37 previously unreported associations for effects on blood-pressure variance and mean, respectively, in the UK Biobank. Transcriptome-wide association studies revealed 633 significant unique gene associations with blood-pressure mean variance. MVtest is broadly applicable to studies of complex quantitative traits and provides an important opportunity to detect novel loci.
Collapse
Affiliation(s)
- Joseph H Breeyear
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Biostatistics and Computational Biology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, Durham, NC, USA
| | - Brian S Mautz
- Population Analytics and Insights, Data Sciences, Janssen Research and Development, Spring House, PA, USA
| | - Jacob M Keaton
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jacklyn N Hellwege
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA; Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eric S Torstenson
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jingjing Liang
- Department of Pharmacy Practice and Science, University of Arizona, Tucson, AZ, USA
| | - Michael J Bray
- Department of Maternal and Fetal Medicine, Orlando Health, Orlando, FL, USA; Genetic Counseling Program, Bay Path University, Longmeadow, MA, USA
| | - Ayush Giri
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA; Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Helen R Warren
- Center of Clinical Pharmacology and Precision Medicine, Queen Mary University, London, England
| | - Patricia B Munroe
- Center of Clinical Pharmacology and Precision Medicine, Queen Mary University, London, England
| | - Digna R Velez Edwards
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA; Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xiaofeng Zhu
- Department of Epidemiology and Biostatistics, Institute for Computational Biology, Case Western Reserve University, Cleveland, OH, USA
| | - Chun Li
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
92
|
Johnson R, Stephens AV, Mester R, Knyazev S, Kohn LA, Freund MK, Bondhus L, Hill BL, Schwarz T, Zaitlen N, Arboleda VA, Bastarache LA, Pasaniuc B, Butte MJ. Electronic health record signatures identify undiagnosed patients with common variable immunodeficiency disease. Sci Transl Med 2024; 16:eade4510. [PMID: 38691621 PMCID: PMC11402387 DOI: 10.1126/scitranslmed.ade4510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 04/10/2024] [Indexed: 05/03/2024]
Abstract
Human inborn errors of immunity include rare disorders entailing functional and quantitative antibody deficiencies due to impaired B cells called the common variable immunodeficiency (CVID) phenotype. Patients with CVID face delayed diagnoses and treatments for 5 to 15 years after symptom onset because the disorders are rare (prevalence of ~1/25,000), and there is extensive heterogeneity in CVID phenotypes, ranging from infections to autoimmunity to inflammatory conditions, overlapping with other more common disorders. The prolonged diagnostic odyssey drives excessive system-wide costs before diagnosis. Because there is no single causal mechanism, there are no genetic tests to definitively diagnose CVID. Here, we present PheNet, a machine learning algorithm that identifies patients with CVID from their electronic health records (EHRs). PheNet learns phenotypic patterns from verified CVID cases and uses this knowledge to rank patients by likelihood of having CVID. PheNet could have diagnosed more than half of our patients with CVID 1 or more years earlier than they had been diagnosed. When applied to a large EHR dataset, followed by blinded chart review of the top 100 patients ranked by PheNet, we found that 74% were highly probable to have CVID. We externally validated PheNet using >6 million records from disparate medical systems in California and Tennessee. As artificial intelligence and machine learning make their way into health care, we show that algorithms such as PheNet can offer clinical benefits by expediting the diagnosis of rare diseases.
Collapse
Affiliation(s)
- Ruth Johnson
- Department of Computer Science, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Alexis V. Stephens
- Department of Pediatrics, Division of Immunology, Allergy and Rheumatology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Rachel Mester
- Department of Computer Science, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Sergey Knyazev
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Lisa A. Kohn
- Department of Pediatrics, Division of Immunology, Allergy and Rheumatology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Malika K. Freund
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Leroy Bondhus
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Brian L. Hill
- Department of Computer Science, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Tommer Schwarz
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Noah Zaitlen
- Department of Neurology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Valerie A. Arboleda
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Computational Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Lisa A. Bastarache
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, USA 37203
| | - Bogdan Pasaniuc
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Computational Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Manish J. Butte
- Department of Pediatrics, Division of Immunology, Allergy and Rheumatology, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
93
|
Nguyen TQ, Kerley CI, Key AP, Maxwell-Horn AC, Wells QS, Neul JL, Cutting LE, Landman BA. Phenotyping Down syndrome: discovery and predictive modelling with electronic medical records. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2024; 68:491-511. [PMID: 38303157 PMCID: PMC11023778 DOI: 10.1111/jir.13124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 11/20/2023] [Accepted: 12/27/2023] [Indexed: 02/03/2024]
Abstract
BACKGROUND Individuals with Down syndrome (DS) have a heightened risk for various co-occurring health conditions, including congenital heart disease (CHD). In this two-part study, electronic medical records (EMRs) were leveraged to examine co-occurring health conditions among individuals with DS (Study 1) and to investigate health conditions linked to surgical intervention among DS cases with CHD (Study 2). METHODS De-identified EMRs were acquired from Vanderbilt University Medical Center and facilitated creating a cohort of N = 2282 DS cases (55% females), along with comparison groups for each study. In Study 1, DS cases were one-by-two sex and age matched with samples of case-controls and of individuals with other intellectual and developmental difficulties (IDDs). The phenome-disease association study (PheDAS) strategy was employed to reveal co-occurring health conditions in DS versus comparison groups, which were then ranked for how often they are discussed in relation to DS using the PubMed database and Novelty Finding Index. In Study 2, a subset of DS individuals with CHD [N = 1098 (48%)] were identified to create longitudinal data for N = 204 cases with surgical intervention (19%) versus 204 case-controls. Data were included in predictive models and assessed which model-based health conditions, when more prevalent, would increase the likelihood of surgical intervention. RESULTS In Study 1, relative to case-controls and those with other IDDs, co-occurring health conditions among individuals with DS were confirmed to include heart failure, pulmonary heart disease, atrioventricular block, heart transplant/surgery and primary pulmonary hypertension (circulatory); hypothyroidism (endocrine/metabolic); and speech and language disorder and Alzheimer's disease (neurological/mental). Findings also revealed more versus less prevalent co-occurring health conditions in individuals with DS when comparing with those with other IDDs. Findings with high Novelty Finding Index were abnormal electrocardiogram, non-rheumatic aortic valve disorders and heart failure (circulatory); acid-base balance disorder (endocrine/metabolism); and abnormal blood chemistry (symptoms). In Study 2, the predictive models revealed that among individuals with DS and CHD, presence of health conditions such as congestive heart failure (circulatory), valvular heart disease and cardiac shunt (congenital), and pleural effusion and pulmonary collapse (respiratory) were associated with increased likelihood of surgical intervention. CONCLUSIONS Research efforts using EMRs and rigorous statistical methods could shed light on the complexity in health profile among individuals with DS and other IDDs and motivate precision-care development.
Collapse
Affiliation(s)
- T Q Nguyen
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
- Peabody College of Education and Human Development, Vanderbilt University, Nashville, TN, USA
| | - C I Kerley
- School of Engineering, Vanderbilt University, Nashville, TN, USA
| | - A P Key
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Speech and Hearing Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - A C Maxwell-Horn
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Q S Wells
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - J L Neul
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - L E Cutting
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
- Peabody College of Education and Human Development, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - B A Landman
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
- School of Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
94
|
Kolla BP, Mansukhani MP, Chakravorty S, Frank JA, Coombes BJ. Prevalence and associations of multiple hypnotic prescriptions in a clinical sample. J Clin Sleep Med 2024; 20:793-800. [PMID: 38189358 PMCID: PMC11063698 DOI: 10.5664/jcsm.10988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/09/2024]
Abstract
STUDY OBJECTIVES We examined the prevalence of multiple hypnotic prescriptions and its association with clinical and demographic characteristics from the electronic health record (EHR) in the Mayo Clinic Biobank. METHODS Adult participants enrolled in the Mayo Clinic Biobank with an EHR number of ≥ 1 year were included (n = 52,940). Clinical and demographic characteristics were compared between participants who were and were not prescribed any hypnotic approved for insomnia by the US Food and Drug Administration and/or trazodone and in those prescribed a single vs multiple (≥ 2) hypnotics. A phenotype-based, phenome-wide association study (PheWAS) examining associations between hypnotic prescriptions and diagnoses across the EHR was performed adjusting for demographic and other confounders. RESULTS A total of 17,662 (33%) participants were prescribed at least 1 hypnotic and 5,331 (10%) received ≥ 2 hypnotics. Participants who were prescribed a hypnotic were more likely to be older, female, White, with a longer EHR, and a greater number of diagnostic codes (all P < .001). Those with multiple hypnotic prescriptions were more likely to be younger, female, with a longer EHR, and a greater number of diagnostic codes (all P < .001) compared with those prescribed a single hypnotic. The PheWAS revealed that participants with multiple hypnotic prescriptions had higher rates of mood disorders, anxiety disorders, suicidal ideation, restless legs syndrome, and chronic pain (all P < 1 e-10). CONCLUSIONS Receiving multiple hypnotic prescriptions is common and associated with a greater prevalence of psychiatric, chronic pain, and sleep-related movement disorders. Future studies should examine potential genetic associations with multiple hypnotic prescriptions to personalize treatments for chronic insomnia. CITATION Kolla BP, Mansukhani MP, Chakravorty S, Frank JA, Coombes BJ. Prevalence and associations of multiple hypnotic prescriptions in a clinical sample. J Clin Sleep Med. 2024;20(5):793-800.
Collapse
Affiliation(s)
- Bhanu Prakash Kolla
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, Minnesota
- Center for Sleep Medicine, Mayo Clinic, Rochester, Minnesota
| | | | | | - Jacob A. Frank
- Department of Quantitative Health Sciences, Division of Computational Biology, Mayo Clinic, Rochester, Minnesota
| | - Brandon J. Coombes
- Department of Quantitative Health Sciences, Division of Computational Biology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
95
|
Yan C, Zhang Z, Nyemba S, Li Z. Generating Synthetic Electronic Health Record Data Using Generative Adversarial Networks: Tutorial. JMIR AI 2024; 3:e52615. [PMID: 38875595 PMCID: PMC11074891 DOI: 10.2196/52615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 01/24/2024] [Accepted: 03/07/2024] [Indexed: 06/16/2024]
Abstract
Synthetic electronic health record (EHR) data generation has been increasingly recognized as an important solution to expand the accessibility and maximize the value of private health data on a large scale. Recent advances in machine learning have facilitated more accurate modeling for complex and high-dimensional data, thereby greatly enhancing the data quality of synthetic EHR data. Among various approaches, generative adversarial networks (GANs) have become the main technical path in the literature due to their ability to capture the statistical characteristics of real data. However, there is a scarcity of detailed guidance within the domain regarding the development procedures of synthetic EHR data. The objective of this tutorial is to present a transparent and reproducible process for generating structured synthetic EHR data using a publicly accessible EHR data set as an example. We cover the topics of GAN architecture, EHR data types and representation, data preprocessing, GAN training, synthetic data generation and postprocessing, and data quality evaluation. We conclude this tutorial by discussing multiple important issues and future opportunities in this domain. The source code of the entire process has been made publicly available.
Collapse
Affiliation(s)
- Chao Yan
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Ziqi Zhang
- Department of Computer Science, Vanderbilt University, Nashville, TN, United States
| | - Steve Nyemba
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Zhuohang Li
- Department of Computer Science, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
96
|
Mosley JD, Shelley JP, Dickson AL, Zanussi J, Daniel LL, Zheng NS, Bastarache L, Wei WQ, Shi M, Jarvik GP, Rosenthal EA, Khan A, Sherafati A, Kullo IJ, Walunas TL, Glessner J, Hakonarson H, Cox NJ, Roden DM, Frangakis SG, Vanderwerff B, Stein CM, Van Driest SL, Borinstein SC, Shu XO, Zawistowski M, Chung CP, Kawai VK. Clinical associations with a polygenic predisposition to benign lower white blood cell counts. Nat Commun 2024; 15:3384. [PMID: 38649760 PMCID: PMC11035609 DOI: 10.1038/s41467-024-47804-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 04/10/2024] [Indexed: 04/25/2024] Open
Abstract
Polygenic variation unrelated to disease contributes to interindividual variation in baseline white blood cell (WBC) counts, but its clinical significance is uncharacterized. We investigated the clinical consequences of a genetic predisposition toward lower WBC counts among 89,559 biobank participants from tertiary care centers using a polygenic score for WBC count (PGSWBC) comprising single nucleotide polymorphisms not associated with disease. A predisposition to lower WBC counts was associated with a decreased risk of identifying pathology on a bone marrow biopsy performed for a low WBC count (odds-ratio = 0.55 per standard deviation increase in PGSWBC [95%CI, 0.30-0.94], p = 0.04), an increased risk of leukopenia (a low WBC count) when treated with a chemotherapeutic (n = 1724, hazard ratio [HR] = 0.78 [0.69-0.88], p = 4.0 × 10-5) or immunosuppressant (n = 354, HR = 0.61 [0.38-0.99], p = 0.04). A predisposition to benign lower WBC counts was associated with an increased risk of discontinuing azathioprine treatment (n = 1,466, HR = 0.62 [0.44-0.87], p = 0.006). Collectively, these findings suggest that there are genetically predisposed individuals who are susceptible to escalations or alterations in clinical care that may be harmful or of little benefit.
Collapse
Affiliation(s)
- Jonathan D Mosley
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - John P Shelley
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alyson L Dickson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jacy Zanussi
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Laura L Daniel
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Neil S Zheng
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Yale School of Medicine, New Haven, CT, USA
| | - Lisa Bastarache
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mingjian Shi
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gail P Jarvik
- Department of Genome Sciences, University of Washington Medical Center, Seattle, WA, USA
- Department of Medicine (Medical Genetics), University of Washington Medical Center, Seattle, WA, USA
| | - Elisabeth A Rosenthal
- Department of Medicine (Medical Genetics), University of Washington Medical Center, Seattle, WA, USA
| | - Atlas Khan
- Division of Nephrology, Dept of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Alborz Sherafati
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Iftikhar J Kullo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Theresa L Walunas
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Joseph Glessner
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Hakon Hakonarson
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nancy J Cox
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dan M Roden
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Stephan G Frangakis
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Brett Vanderwerff
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - C Michael Stein
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sara L Van Driest
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Scott C Borinstein
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xiao-Ou Shu
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Matthew Zawistowski
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | | | - Vivian K Kawai
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
97
|
Tao Y, Zhao R, Yang B, Han J, Li Y. Dissecting the shared genetic landscape of anxiety, depression, and schizophrenia. J Transl Med 2024; 22:373. [PMID: 38637810 PMCID: PMC11025255 DOI: 10.1186/s12967-024-05153-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/01/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Numerous studies highlight the genetic underpinnings of mental disorders comorbidity, particularly in anxiety, depression, and schizophrenia. However, their shared genetic loci are not well understood. Our study employs Mendelian randomization (MR) and colocalization analyses, alongside multi-omics data, to uncover potential genetic targets for these conditions, thereby informing therapeutic and drug development strategies. METHODS We utilized the Consortium for Linkage Disequilibrium Score Regression (LDSC) and Mendelian Randomization (MR) analysis to investigate genetic correlations among anxiety, depression, and schizophrenia. Utilizing GTEx V8 eQTL and deCODE Genetics pQTL data, we performed a three-step summary-data-based Mendelian randomization (SMR) and protein-protein interaction analysis. This helped assess causal and comorbid loci for these disorders and determine if identified loci share coincidental variations with psychiatric diseases. Additionally, phenome-wide association studies, drug prediction, and molecular docking validated potential drug targets. RESULTS We found genetic correlations between anxiety, depression, and schizophrenia, and under a meta-analysis of MR from multiple databases, the causal relationships among these disorders are supported. Based on this, three-step SMR and colocalization analyses identified ITIH3 and CCS as being related to the risk of developing depression, while CTSS and DNPH1 are related to the onset of schizophrenia. BTN3A1, PSMB4, and TIMP4 were identified as comorbidity loci for both disorders. Molecules that could not be determined through colocalization analysis were also presented. Drug prediction and molecular docking showed that some drugs and proteins have good binding affinity and available structural data. CONCLUSIONS Our study indicates genetic correlations and shared risk loci between anxiety, depression, and schizophrenia. These findings offer insights into the underlying mechanisms of their comorbidities and aid in drug development.
Collapse
Affiliation(s)
- Yiming Tao
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hankou, Wuhan, 430030, China
- Department of Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250101, Shandong, China
| | - Rui Zhao
- Department of Laboratory Medicine, The First Afliated Hospital of Chongqing Medical University, Chongqing, 400042, China
| | - Bin Yang
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hankou, Wuhan, 430030, China
| | - Jie Han
- Department of Emergency, School of Medicine, Qingdao Municipal Hospital, Qingdao University, Qingdao, 266071, China.
| | - Yongsheng Li
- Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hankou, Wuhan, 430030, China.
| |
Collapse
|
98
|
Baldridge D, Kaster L, Sancimino C, Srivastava S, Molholm S, Gupta A, Oh I, Lanzotti V, Grewal D, Riggs ER, Savatt JM, Hauck R, Sveden A, Constantino JN, Piven J, Gurnett CA, Chopra M, Hazlett H, Payne PRO. The Brain Gene Registry: a data snapshot. J Neurodev Disord 2024; 16:17. [PMID: 38632549 PMCID: PMC11022437 DOI: 10.1186/s11689-024-09530-3] [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: 10/17/2023] [Accepted: 03/27/2024] [Indexed: 04/19/2024] Open
Abstract
Monogenic disorders account for a large proportion of population-attributable risk for neurodevelopmental disabilities. However, the data necessary to infer a causal relationship between a given genetic variant and a particular neurodevelopmental disorder is often lacking. Recognizing this scientific roadblock, 13 Intellectual and Developmental Disabilities Research Centers (IDDRCs) formed a consortium to create the Brain Gene Registry (BGR), a repository pairing clinical genetic data with phenotypic data from participants with variants in putative brain genes. Phenotypic profiles are assembled from the electronic health record (EHR) and a battery of remotely administered standardized assessments collectively referred to as the Rapid Neurobehavioral Assessment Protocol (RNAP), which include cognitive, neurologic, and neuropsychiatric assessments, as well as assessments for attention deficit hyperactivity disorder (ADHD) and autism spectrum disorder (ASD). Co-enrollment of BGR participants in the Clinical Genome Resource's (ClinGen's) GenomeConnect enables display of variant information in ClinVar. The BGR currently contains data on 479 participants who are 55% male, 6% Asian, 6% Black or African American, 76% white, and 12% Hispanic/Latine. Over 200 genes are represented in the BGR, with 12 or more participants harboring variants in each of these genes: CACNA1A, DNMT3A, SLC6A1, SETD5, and MYT1L. More than 30% of variants are de novo and 43% are classified as variants of uncertain significance (VUSs). Mean standard scores on cognitive or developmental screens are below average for the BGR cohort. EHR data reveal developmental delay as the earliest and most common diagnosis in this sample, followed by speech and language disorders, ASD, and ADHD. BGR data has already been used to accelerate gene-disease validity curation of 36 genes evaluated by ClinGen's BGR Intellectual Disability (ID)-Autism (ASD) Gene Curation Expert Panel. In summary, the BGR is a resource for use by stakeholders interested in advancing translational research for brain genes and continues to recruit participants with clinically reported variants to establish a rich and well-characterized national resource to promote research on neurodevelopmental disorders.
Collapse
Affiliation(s)
- Dustin Baldridge
- Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA.
| | - Levi Kaster
- Institute for Informatics, Data Science and Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Catherine Sancimino
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Siddharth Srivastava
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA
| | - Sophie Molholm
- Departments of Pediatrics and Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Aditi Gupta
- Institute for Informatics, Data Science and Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Inez Oh
- Institute for Informatics, Data Science and Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Virginia Lanzotti
- Department of Psychiatry, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Daleep Grewal
- Institute for Informatics, Data Science and Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Erin Rooney Riggs
- Autism and Developmental Medicine Institute, Geisinger, Danville, PA, USA
| | | | - Rachel Hauck
- Institute for Informatics, Data Science and Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Abigail Sveden
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA
| | - John N Constantino
- Division of Behavioral and Mental Health, Departments of Psychiatry and Pediatrics, Children's Healthcare of Atlanta, Emory University, Atlanta, GA, USA
| | - Joseph Piven
- The Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, NC, USA
| | - Christina A Gurnett
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Maya Chopra
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, MA, USA
| | - Heather Hazlett
- The Carolina Institute for Developmental Disabilities, University of North Carolina, Chapel Hill, NC, USA
| | - Philip R O Payne
- Institute for Informatics, Data Science and Biostatistics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| |
Collapse
|
99
|
Yun JS, Jung SH, Lee SN, Jung SM, Won HH, Kim D, Choi JA. Polygenic risk score-based phenome-wide association for glaucoma and its impact on disease susceptibility in two large biobanks. J Transl Med 2024; 22:355. [PMID: 38622600 PMCID: PMC11020996 DOI: 10.1186/s12967-024-05152-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/01/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND Glaucoma is a leading cause of worldwide irreversible blindness. Considerable uncertainty remains regarding the association between a variety of phenotypes and the genetic risk of glaucoma, as well as the impact they exert on the glaucoma development. METHODS We investigated the associations of genetic liability for primary open angle glaucoma (POAG) with a wide range of potential risk factors and to assess its impact on the risk of incident glaucoma. The phenome-wide association study (PheWAS) approach was applied to determine the association of POAG polygenic risk score (PRS) with a wide range of phenotypes in 377, 852 participants from the UK Biobank study and 43,623 participants from the Penn Medicine Biobank study, all of European ancestry. Participants were stratified into four risk tiers: low, intermediate, high, and very high-risk. Cox proportional hazard models assessed the relationship of POAG PRS and ocular factors with new glaucoma events. RESULTS In both discovery and replication set in the PheWAS, a higher genetic predisposition to POAG was specifically correlated with ocular disease phenotypes. The POAG PRS exhibited correlations with low corneal hysteresis, refractive error, and ocular hypertension, demonstrating a strong association with the onset of glaucoma. Individuals carrying a high genetic burden exhibited a 9.20-fold, 11.88-fold, and 28.85-fold increase in glaucoma incidence when associated with low corneal hysteresis, high myopia, and elevated intraocular pressure, respectively. CONCLUSION Genetic susceptibility to POAG primarily influences ocular conditions, with limited systemic associations. Notably, the baseline polygenic risk for POAG robustly associates with new glaucoma events, revealing a large combined effect of genetic and ocular risk factors on glaucoma incidents.
Collapse
Affiliation(s)
- Jae-Seung Yun
- Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sang-Hyuk Jung
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Su-Nam Lee
- Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Min Jung
- Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hong-Hee Won
- Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea.
- Samsung Genome Institute, Samsung Medical Center, Seoul, Republic of Korea.
| | - Dokyoon Kim
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA, USA.
| | - Jin A Choi
- Department of Ophthalmology, College of Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
100
|
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. CELL GENOMICS 2024; 4:100523. [PMID: 38508198 PMCID: PMC11019356 DOI: 10.1016/j.xgen.2024.100523] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/15/2023] [Accepted: 02/20/2024] [Indexed: 03/22/2024]
Abstract
Polygenic risk scores (PRSs) are an emerging tool to predict the clinical phenotypes and outcomes of individuals. We propose PRSmix, a framework that 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 for 47 and 32 diseases/traits in European and South Asian ancestries, respectively. PRSmix demonstrated a mean prediction accuracy improvement of 1.20-fold (95% confidence interval [CI], [1.10; 1.3]; p = 9.17 × 10-5) and 1.19-fold (95% CI, [1.11; 1.27]; p = 1.92 × 10-6), and PRSmix+ improved the prediction accuracy by 1.72-fold (95% CI, [1.40; 2.04]; p = 7.58 × 10-6) and 1.42-fold (95% CI, [1.25; 1.59]; p = 8.01 × 10-7) in European and South Asian ancestries, respectively. Compared to the previously cross-trait-combination methods with scores from pre-defined correlated traits, we demonstrated that our method improved prediction accuracy for coronary artery disease up to 3.27-fold (95% CI, [2.1; 4.44]; p value after false discovery rate (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, USA; Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
| | - Leland E Hull
- Division of General Internal Medicine, Massachusetts General Hospital, 100 Cambridge Street, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - 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, USA; Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
| | - 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, USA; Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
| | - 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, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - 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, USA; Center for Genomic Medicine and Cardiovascular Research Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA.
| |
Collapse
|