501
|
Zhao X, Geng X, Srinivasasainagendra V, Chaudhary N, Judd S, Wadley V, Gutiérrez OM, Wang H, Lange EM, Lange LA, Woo D, Unverzagt FW, Safford M, Cushman M, Limdi N, Quarells R, Arnett DK, Irvin MR, Zhi D. A PheWAS study of a large observational epidemiological cohort of African Americans from the REGARDS study. BMC Med Genomics 2019; 12:26. [PMID: 30704471 PMCID: PMC6357353 DOI: 10.1186/s12920-018-0462-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Cardiovascular disease, diabetes, and kidney disease are among the leading causes of death and disability worldwide. However, knowledge of genetic determinants of those diseases in African Americans remains limited. RESULTS In our study, associations between 4956 GWAS catalog reported SNPs and 67 traits were examined among 7726 African Americans from the REasons for Geographic and Racial Differences in Stroke (REGARDS) study, which is focused on identifying factors that increase stroke risk. The prevalent and incident phenotypes studied included inflammation, kidney traits, cardiovascular traits and cognition. Our results validated 29 known associations, of which eight associations were reported for the first time in African Americans. CONCLUSION Our cross-racial validation of GWAS findings provide additional evidence for the important roles of these loci in the disease process and may help identify genes especially important for future functional validation.
Collapse
Affiliation(s)
- Xueyan Zhao
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL 35233 USA
| | - Xin Geng
- BGI-Shenzhen, Shenzhen, 518083 China
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030 USA
| | | | - Ninad Chaudhary
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL 35233 USA
| | - Suzanne Judd
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL 35233 USA
| | - Virginia Wadley
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233 USA
| | - Orlando M. Gutiérrez
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL 35233 USA
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233 USA
| | - Henry Wang
- Department of Emergency Medicine, University of Alabama at Birmingham, Birmingham, AL 35233 USA
| | - Ethan M. Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 USA
| | - Leslie A. Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 USA
| | - Daniel Woo
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267 USA
| | - Frederick W. Unverzagt
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Monika Safford
- Division of General Internal Medicine, Weill Cornell Medical College, Cornell University, New York, NY 10065 USA
| | - Mary Cushman
- Department of Medicine and Pathology, Larner College of Medicine at the University of Vermont, Burlington, VT 05405 USA
| | - Nita Limdi
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Rakale Quarells
- Cardiovascular Research Institute, Department of Community Health and Preventive Medicine, Morehouse School of Medicine, Atlanta, GA 30310 USA
| | - Donna K. Arnett
- College of Public Health, University of Kentucky, Lexington, KY 40506 USA
| | - Marguerite R. Irvin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL 35233 USA
| | - Degui Zhi
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030 USA
- School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030 USA
| |
Collapse
|
502
|
Using phenome-wide association to investigate the function of a schizophrenia risk locus at SLC39A8. Transl Psychiatry 2019; 9:45. [PMID: 30696806 PMCID: PMC6351652 DOI: 10.1038/s41398-019-0386-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 11/08/2018] [Accepted: 11/13/2018] [Indexed: 12/18/2022] Open
Abstract
While nearly all common genomic variants associated with schizophrenia have no known function, one corresponds to a missense variant associated with change in efficiency of a metal ion transporter, ZIP8, coded by SLC39A8. This variant has been linked to a range of phenotypes and is believed to be under recent selection pressure, but its impact on health is poorly understood. We sought to understand phenotypic implications of this variant in a large genomic biobank using an unbiased phenome-wide approach. Specifically, we generated 50 topics based on diagnostic codes using latent Dirichlet allocation, and examined them for association with the risk variant. Then, any significant topics were further characterized by examining association with individual diagnostic codes contributing to the topic. Among 50 topics, 1 was associated at an experiment-wide significance threshold (beta = 0.003, uncorrected p = 0.00049), comprising predominantly brain-related codes, including intracranial hemorrhage, cerebrovascular disease, and delirium/dementia. These results suggest that a functional variant previously associated with schizophrenia risk also increases liability to cerebrovascular disease. They further illustrate the utility of a topic-based approach to phenome-wide association.
Collapse
|
503
|
Zhang X, Basile AO, Pendergrass SA, Ritchie MD. Real world scenarios in rare variant association analysis: the impact of imbalance and sample size on the power in silico. BMC Bioinformatics 2019; 20:46. [PMID: 30669967 PMCID: PMC6343276 DOI: 10.1186/s12859-018-2591-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 12/26/2018] [Indexed: 11/11/2022] Open
Abstract
Background The development of sequencing techniques and statistical methods provides great opportunities for identifying the impact of rare genetic variation on complex traits. However, there is a lack of knowledge on the impact of sample size, case numbers, the balance of cases vs controls for both burden and dispersion based rare variant association methods. For example, Phenome-Wide Association Studies may have a wide range of case and control sample sizes across hundreds of diagnoses and traits, and with the application of statistical methods to rare variants, it is important to understand the strengths and limitations of the analyses. Results We conducted a large-scale simulation of randomly selected low-frequency protein-coding regions using twelve different balanced samples with an equal number of cases and controls as well as twenty-one unbalanced sample scenarios. We further explored statistical performance of different minor allele frequency thresholds and a range of genetic effect sizes. Our simulation results demonstrate that using an unbalanced study design has an overall higher type I error rate for both burden and dispersion tests compared with a balanced study design. Regression has an overall higher type I error with balanced cases and controls, while SKAT has higher type I error for unbalanced case-control scenarios. We also found that both type I error and power were driven by the number of cases in addition to the case to control ratio under large control group scenarios. Based on our power simulations, we observed that a SKAT analysis with case numbers larger than 200 for unbalanced case-control models yielded over 90% power with relatively well controlled type I error. To achieve similar power in regression, over 500 cases are needed. Moreover, SKAT showed higher power to detect associations in unbalanced case-control scenarios than regression. Conclusions Our results provide important insights into rare variant association study designs by providing a landscape of type I error and statistical power for a wide range of sample sizes. These results can serve as a benchmark for making decisions about study design for rare variant analyses. Electronic supplementary material The online version of this article (10.1186/s12859-018-2591-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xinyuan Zhang
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anna O Basile
- Department of Biomedical Informatics, Columbia University, New York, NY, USA
| | - Sarah A Pendergrass
- Biomedical and Translational Informatics Institute, Geisinger, Danville, PA, USA
| | - Marylyn D Ritchie
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA. .,Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
504
|
Wang Y, Sohn S, Liu S, Shen F, Wang L, Atkinson EJ, Amin S, Liu H. A clinical text classification paradigm using weak supervision and deep representation. BMC Med Inform Decis Mak 2019; 19:1. [PMID: 30616584 PMCID: PMC6322223 DOI: 10.1186/s12911-018-0723-6] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 12/10/2018] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Automatic clinical text classification is a natural language processing (NLP) technology that unlocks information embedded in clinical narratives. Machine learning approaches have been shown to be effective for clinical text classification tasks. However, a successful machine learning model usually requires extensive human efforts to create labeled training data and conduct feature engineering. In this study, we propose a clinical text classification paradigm using weak supervision and deep representation to reduce these human efforts. METHODS We develop a rule-based NLP algorithm to automatically generate labels for the training data, and then use the pre-trained word embeddings as deep representation features for training machine learning models. Since machine learning is trained on labels generated by the automatic NLP algorithm, this training process is called weak supervision. We evaluat the paradigm effectiveness on two institutional case studies at Mayo Clinic: smoking status classification and proximal femur (hip) fracture classification, and one case study using a public dataset: the i2b2 2006 smoking status classification shared task. We test four widely used machine learning models, namely, Support Vector Machine (SVM), Random Forest (RF), Multilayer Perceptron Neural Networks (MLPNN), and Convolutional Neural Networks (CNN), using this paradigm. Precision, recall, and F1 score are used as metrics to evaluate performance. RESULTS CNN achieves the best performance in both institutional tasks (F1 score: 0.92 for Mayo Clinic smoking status classification and 0.97 for fracture classification). We show that word embeddings significantly outperform tf-idf and topic modeling features in the paradigm, and that CNN captures additional patterns from the weak supervision compared to the rule-based NLP algorithms. We also observe two drawbacks of the proposed paradigm that CNN is more sensitive to the size of training data, and that the proposed paradigm might not be effective for complex multiclass classification tasks. CONCLUSION The proposed clinical text classification paradigm could reduce human efforts of labeled training data creation and feature engineering for applying machine learning to clinical text classification by leveraging weak supervision and deep representation. The experimental experiments have validated the effectiveness of paradigm by two institutional and one shared clinical text classification tasks.
Collapse
Affiliation(s)
- Yanshan Wang
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, 200 1st ST SW, Rochester, MN 55905 USA
| | - Sunghwan Sohn
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, 200 1st ST SW, Rochester, MN 55905 USA
| | - Sijia Liu
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, 200 1st ST SW, Rochester, MN 55905 USA
| | - Feichen Shen
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, 200 1st ST SW, Rochester, MN 55905 USA
| | - Liwei Wang
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, 200 1st ST SW, Rochester, MN 55905 USA
| | - Elizabeth J. Atkinson
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, 200 1st ST SW, Rochester, MN 55905 USA
| | - Shreyasee Amin
- Division of Rheumatology, Department of Medicine, Mayo Clinic, 200 1st ST SW, Rochester, MN 55905 USA
- Division of Epidemiology, Department of Health Sciences Research, Mayo Clinic, 200 1st ST SW, Rochester, MN 55905 USA
| | - Hongfang Liu
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, 200 1st ST SW, Rochester, MN 55905 USA
| |
Collapse
|
505
|
Verma A, Bang L, Miller JE, Zhang Y, Lee MTM, Zhang Y, Byrska-Bishop M, Carey DJ, Ritchie MD, Pendergrass SA, Kim D. Human-Disease Phenotype Map Derived from PheWAS across 38,682 Individuals. Am J Hum Genet 2019; 104:55-64. [PMID: 30598166 PMCID: PMC6323551 DOI: 10.1016/j.ajhg.2018.11.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 11/12/2018] [Indexed: 12/17/2022] Open
Abstract
Phenome-wide association studies (PheWASs) have been a useful tool for testing associations between genetic variations and multiple complex traits or diagnoses. Linking PheWAS-based associations between phenotypes and a variant or a genomic region into a network provides a new way to investigate cross-phenotype associations, and it might broaden the understanding of genetic architecture that exists between diagnoses, genes, and pleiotropy. We created a network of associations from one of the largest PheWASs on electronic health record (EHR)-derived phenotypes across 38,682 unrelated samples from the Geisinger's biobank; the samples were genotyped through the DiscovEHR project. We computed associations between 632,574 common variants and 541 diagnosis codes. Using these associations, we constructed a "disease-disease" network (DDN) wherein pairs of diseases were connected on the basis of shared associations with a given genetic variant. The DDN provides a landscape of intra-connections within the same disease classes, as well as inter-connections across disease classes. We identified clusters of diseases with known biological connections, such as autoimmune disorders (type 1 diabetes, rheumatoid arthritis, and multiple sclerosis) and cardiovascular disorders. Previously unreported relationships between multiple diseases were identified on the basis of genetic associations as well. The network approach applied in this study can be used to uncover interactions between diseases as a result of their shared, potentially pleiotropic SNPs. Additionally, this approach might advance clinical research and even clinical practice by accelerating our understanding of disease mechanisms on the basis of similar underlying genetic associations.
Collapse
Affiliation(s)
- Anurag Verma
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA; The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Lisa Bang
- Biomedical and Translational Informatics Institute, Geisinger, Danville, PA 17821, USA
| | - Jason E Miller
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yanfei Zhang
- Genomic Medicine Institute, Geisinger, Danville, PA 17821, USA
| | | | - Yu Zhang
- Department of Statistics, The Pennsylvania State University, University Park, PA 16802, USA
| | - Marta Byrska-Bishop
- Biomedical and Translational Informatics Institute, Geisinger, Danville, PA 17821, USA
| | - David J Carey
- Weis Center for Research, Geisinger, Danville, PA 17821, USA
| | - Marylyn D Ritchie
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA; The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Sarah A Pendergrass
- Biomedical and Translational Informatics Institute, Geisinger, Danville, PA 17821, USA
| | - Dokyoon Kim
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA; Biomedical and Translational Informatics Institute, Geisinger, Danville, PA 17821, USA.
| |
Collapse
|
506
|
Yu S, Ma Y, Gronsbell J, Cai T, Ananthakrishnan AN, Gainer VS, Churchill SE, Szolovits P, Murphy SN, Kohane IS, Liao KP, Cai T. Enabling phenotypic big data with PheNorm. J Am Med Inform Assoc 2019; 25:54-60. [PMID: 29126253 DOI: 10.1093/jamia/ocx111] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 09/14/2017] [Indexed: 01/20/2023] Open
Abstract
Objective Electronic health record (EHR)-based phenotyping infers whether a patient has a disease based on the information in his or her EHR. A human-annotated training set with gold-standard disease status labels is usually required to build an algorithm for phenotyping based on a set of predictive features. The time intensiveness of annotation and feature curation severely limits the ability to achieve high-throughput phenotyping. While previous studies have successfully automated feature curation, annotation remains a major bottleneck. In this paper, we present PheNorm, a phenotyping algorithm that does not require expert-labeled samples for training. Methods The most predictive features, such as the number of International Classification of Diseases, Ninth Revision, Clinical Modification (ICD-9-CM) codes or mentions of the target phenotype, are normalized to resemble a normal mixture distribution with high area under the receiver operating curve (AUC) for prediction. The transformed features are then denoised and combined into a score for accurate disease classification. Results We validated the accuracy of PheNorm with 4 phenotypes: coronary artery disease, rheumatoid arthritis, Crohn's disease, and ulcerative colitis. The AUCs of the PheNorm score reached 0.90, 0.94, 0.95, and 0.94 for the 4 phenotypes, respectively, which were comparable to the accuracy of supervised algorithms trained with sample sizes of 100-300, with no statistically significant difference. Conclusion The accuracy of the PheNorm algorithms is on par with algorithms trained with annotated samples. PheNorm fully automates the generation of accurate phenotyping algorithms and demonstrates the capacity for EHR-driven annotations to scale to the next level - phenotypic big data.
Collapse
Affiliation(s)
- Sheng Yu
- Center for Statistical Science, Tsinghua University, Beijing, China.,Department of Industrial Engineering, Tsinghua University, Beijing, China
| | - Yumeng Ma
- Department of Mathematical Sciences, Tsinghua University, Beijing, China
| | - Jessica Gronsbell
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Tianrun Cai
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Vivian S Gainer
- Research Information Science and Computing, Partners HealthCare, Charlestown, MA, USA
| | - Susanne E Churchill
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Peter Szolovits
- Computer Science and Artificial Intelligence Laboratory (CSAIL), Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shawn N Murphy
- Research Information Science and Computing, Partners HealthCare, Charlestown, MA, USA.,Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Isaac S Kohane
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Katherine P Liao
- Department of Medicine, Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, MA, USA
| | - Tianxi Cai
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
507
|
Pendergrass SA, Crawford DC. Using Electronic Health Records To Generate Phenotypes For Research. CURRENT PROTOCOLS IN HUMAN GENETICS 2019; 100:e80. [PMID: 30516347 PMCID: PMC6318047 DOI: 10.1002/cphg.80] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Electronic health records contain patient-level data collected during and for clinical care. Data within the electronic health record include diagnostic billing codes, procedure codes, vital signs, laboratory test results, clinical imaging, and physician notes. With repeated clinic visits, these data are longitudinal, providing important information on disease development, progression, and response to treatment or intervention strategies. The near universal adoption of electronic health records nationally has the potential to provide population-scale real-world clinical data accessible for biomedical research, including genetic association studies. For this research potential to be realized, high-quality research-grade variables must be extracted from these clinical data warehouses. We describe here common and emerging electronic phenotyping approaches applied to electronic health records, as well as current limitations of both the approaches and the biases associated with these clinically collected data that impact their use in research. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Sarah A. Pendergrass
- Biomedical and Translational Informatics Institute,
Geisinger Research, Rockville MD
| | - Dana C. Crawford
- Institute for Computational Biology, Department of
Population and Quantitative Health Sciences, Case Western Reserve University,
Cleveland, OH
| |
Collapse
|
508
|
Giri A, Hellwege JN, Keaton JM, Park J, Qiu C, Warren HR, Torstenson ES, Kovesdy CP, Sun YV, Wilson OD, Robinson-Cohen C, Roumie CL, Chung CP, Birdwell KA, Damrauer SM, DuVall SL, Klarin D, Cho K, Wang Y, Evangelou E, Cabrera CP, Wain LV, Shrestha R, Mautz BS, Akwo EA, Sargurupremraj M, Debette S, Boehnke M, Scott LJ, Luan J, Zhao JH, Willems SM, Thériault S, Shah N, Oldmeadow C, Almgren P, Li-Gao R, Verweij N, Boutin TS, Mangino M, Ntalla I, Feofanova E, Surendran P, Cook JP, Karthikeyan S, Lahrouchi N, Liu C, Sepúlveda N, Richardson TG, Kraja A, Amouyel P, Farrall M, Poulter NR, Laakso M, Zeggini E, Sever P, Scott RA, Langenberg C, Wareham NJ, Conen D, Palmer CNA, Attia J, Chasman DI, Ridker PM, Melander O, Mook-Kanamori DO, Harst PVD, Cucca F, Schlessinger D, Hayward C, Spector TD, Jarvelin MR, Hennig BJ, Timpson NJ, Wei WQ, Smith JC, Xu Y, Matheny ME, Siew EE, Lindgren C, Herzig KH, Dedoussis G, Denny JC, Psaty BM, Howson JMM, Munroe PB, Newton-Cheh C, Caulfield MJ, Elliott P, Gaziano JM, Concato J, Wilson PWF, Tsao PS, Velez Edwards DR, Susztak K, O'Donnell CJ, Hung AM, Edwards TL. Trans-ethnic association study of blood pressure determinants in over 750,000 individuals. Nat Genet 2019; 51:51-62. [PMID: 30578418 PMCID: PMC6365102 DOI: 10.1038/s41588-018-0303-9] [Citation(s) in RCA: 299] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 10/31/2018] [Indexed: 12/15/2022]
Abstract
In this trans-ethnic multi-omic study, we reinterpret the genetic architecture of blood pressure to identify genes, tissues, phenomes and medication contexts of blood pressure homeostasis. We discovered 208 novel common blood pressure SNPs and 53 rare variants in genome-wide association studies of systolic, diastolic and pulse pressure in up to 776,078 participants from the Million Veteran Program (MVP) and collaborating studies, with analysis of the blood pressure clinical phenome in MVP. Our transcriptome-wide association study detected 4,043 blood pressure associations with genetically predicted gene expression of 840 genes in 45 tissues, and mouse renal single-cell RNA sequencing identified upregulated blood pressure genes in kidney tubule cells.
Collapse
Affiliation(s)
- Ayush Giri
- Division of Quantitative Sciences, Department of Obstetrics & Gynecology, Vanderbilt Genetics Institute, Vanderbilt Epidemiology Center, Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, TN, USA
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
| | - Jacklyn N Hellwege
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
- Division of Epidemiology, Department of Medicine, Institute for Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN, USA
| | - Jacob M Keaton
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
- Division of Epidemiology, Department of Medicine, Institute for Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN, USA
| | - Jihwan Park
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
| | - Chengxiang Qiu
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
| | - Helen R Warren
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK
| | - Eric S Torstenson
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
- Division of Epidemiology, Department of Medicine, Institute for Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN, USA
| | - Csaba P Kovesdy
- Nephrology Section, Memphis VA Medical Center, Memphis, TN, USA
| | - Yan V Sun
- 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
| | - Otis D Wilson
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cassianne Robinson-Cohen
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christianne L Roumie
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Geriatrics Research Education and Clinical Center, Tennessee Valley Health System, Veteran's Health Administration, Nashville, TN, USA
| | - Cecilia P Chung
- Division of Rheumatology and Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kelly A Birdwell
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Nephrology, Department of Medicine, Nashville Veteran Affairs Hospital, Nashville, TN, USA
| | - Scott M Damrauer
- Department of Surgery, Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Scott L DuVall
- VA Salt Lake City Health Care System, Salt Lake City, UT, USA
- University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Derek Klarin
- VA Boston Health Care System, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kelly Cho
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
- Division of Aging, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Yu Wang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Claudia P Cabrera
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK
| | - Louise V Wain
- Department of Health Sciences, University of Leicester, Leicester, UK
- National Institute for Health Research, Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Rojesh Shrestha
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
| | - Brian S Mautz
- Division of Epidemiology, Department of Medicine, Institute for Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN, USA
| | - Elvis A Akwo
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Stéphanie Debette
- University of Bordeaux, Bordeaux Population Health Research Center, INSERM UMR 1219, Bordeaux, France
- Department of Neurology, Bordeaux University Hospital, Bordeaux, France
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Laura J Scott
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Jing-Hua Zhao
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Sara M Willems
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Sébastien Thériault
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Quebec City, Quebec, Canada
| | - Nabi Shah
- Division of Molecular and Clinical Medicine, Pat Macpherson Centre for Pharmacogenetics and Pharmacogenomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad, Pakistan
| | | | - Peter Almgren
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Ruifang Li-Gao
- Leiden University Medical Center, Leiden, the Netherlands
| | - Niek Verweij
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Thibaud S Boutin
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, UK
- NIHR Biomedical Research Centre at Guy's and St Thomas' Foundation Trust, London, UK
| | - Ioanna Ntalla
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Elena Feofanova
- Human Genetics Center, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Praveen Surendran
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - James P Cook
- Department of Biostatistics, University of Liverpool, Liverpool, UK
| | - Savita Karthikeyan
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Najim Lahrouchi
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Research Center, Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Heart Center, Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Chunyu Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Nuno Sepúlveda
- Immunology and Infection Department, London School of Hygiene & Tropical Medicine, London, UK
| | - Tom G Richardson
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School (Population Health Sciences), University of Bristol, Oakfield House, Oakfield Grove, Bristol, UK
| | - Aldi Kraja
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
- Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
- Division of Statistical Genomics, Washington University School of Medicine, St. Louis, MO, USA
| | - Philippe Amouyel
- Risk Factors and Molecular Determinants of Aging-Related Diseases (RID-AGE), University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167, Lille, France
| | - Martin Farrall
- Department of Cardiovascular Medicine, The Wellcome Trust Centre for Human Genetics, Oxford, UK
| | - Neil R Poulter
- International Centre for Circulatory Health, Imperial College London, London, UK
| | - Markku Laakso
- University of Eastern Finland, School of Medicine, Kuopio, Finland
| | | | - Peter Sever
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, London, UK
| | - Robert A Scott
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - David Conen
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Colin Neil Alexander Palmer
- Division of Molecular and Clinical Medicine, Pat Macpherson Centre for Pharmacogenetics and Pharmacogenomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - John Attia
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- Faculty of Health, University of Newcastle, Newcastle, New South Wales, Australia
| | - Daniel I Chasman
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Paul M Ridker
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | | | - Pim van der Harst
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato, Cagliari, Italy
- Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, Sassari, Italy
| | - David Schlessinger
- Laboratory of Genetics and Genomics, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, UK
| | - Marjo-Riitta Jarvelin
- MRC-PHE Centre for Environment & Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Unit of Primary Health Care, Oulu University Hospital, OYS, Oulu, Finland
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, Middlesex, UK
| | - Branwen J Hennig
- Wellcome Trust, London, UK
- MRC Unit The Gambia, Atlantic Boulevard, Fajara, Banjul, The Gambia
- London School of Hygiene & Tropical Medicine, London, UK
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School (Population Health Sciences), University of Bristol, Oakfield House, Oakfield Grove, Bristol, UK
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joshua C Smith
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yaomin Xu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael E Matheny
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Geriatrics Research Education and Clinical Center, Tennessee Valley Health System, Veteran's Health Administration, Nashville, TN, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Edward E Siew
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Geriatrics Research Education and Clinical Center, Tennessee Valley Health System, Veteran's Health Administration, Nashville, TN, USA
| | - Cecilia Lindgren
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Big Data Institute at the Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Karl-Heinz Herzig
- Institute of Biomedicine, Biocenter of Oulu, Medical Research Center, Oulu University and Oulu University Hospital, Oulu, Finland
- Department of Gastroenterology and Metabolism, Poznan University of Medical Sciences, Poznan, Poland
| | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Joshua C Denny
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bruce M Psaty
- Departments of Medicine, University of Washington, Seattle, WA, USA
- Departments of Epidemiology, University of Washington, Seattle, WA, USA
- Departments of Health Services, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Joanna M M Howson
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Patricia B Munroe
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK
| | - Christopher Newton-Cheh
- Cardiovascular Research Center, Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mark J Caulfield
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, UK
| | - Paul Elliott
- MRC-PHE Centre for Environment & Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- National Institute for Health Research Imperial Biomedical Research Centre, Imperial College Healthcare NHS Trust, Imperial College London, London, UK
- UK Dementia Research Institute at Imperial College London, London, UK
| | - J Michael Gaziano
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - John Concato
- Clinical Epidemiology Research Center (CERC), VA Cooperative Studies Program, VA Connecticut Healthcare System, West Haven, CT, USA
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Peter W F Wilson
- Atlanta VA Medical Center, Atlanta, GA, USA
- Emory Clinical Cardiovascular Research Institute, Atlanta, GA, USA
| | - Philip S Tsao
- VA Palo Alto Health Care System, Palo Alto, CA, USA
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Digna R Velez Edwards
- Division of Quantitative Sciences, Department of Obstetrics & Gynecology, Vanderbilt Genetics Institute, Vanderbilt Epidemiology Center, Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, TN, USA
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Katalin Susztak
- Department of Medicine, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Christopher J O'Donnell
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- VA Boston Healthcare, Section of Cardiology and Department of Medicine, Boston, MA, USA
| | - Adriana M Hung
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA.
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Todd L Edwards
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, TN, USA.
- Division of Epidemiology, Department of Medicine, Institute for Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
509
|
Beaulieu-Jones BK, Kohane IS, Beam AL. Learning Contextual Hierarchical Structure of Medical Concepts with Poincairé Embeddings to Clarify Phenotypes. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2019; 24:8-17. [PMID: 30864306 PMCID: PMC6417814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Biomedical association studies are increasingly done using clinical concepts, and in particular diagnostic codes from clinical data repositories as phenotypes. Clinical concepts can be represented in a meaningful, vector space using word embedding models. These embeddings allow for comparison between clinical concepts or for straightforward input to machine learning models. Using traditional approaches, good representations require high dimensionality, making downstream tasks such as visualization more difficult. We applied Poincaré embeddings in a 2-dimensional hyperbolic space to a large-scale administrative claims database and show performance comparable to 100-dimensional embeddings in a euclidean space. We then examine disease relationships under different disease contexts to better understand potential phenotypes.
Collapse
Affiliation(s)
| | - Isaac S. Kohane
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew L. Beam
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
510
|
Zeng Z, Deng Y, Li X, Naumann T, Luo Y. Natural Language Processing for EHR-Based Computational Phenotyping. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2019; 16:139-153. [PMID: 29994486 PMCID: PMC6388621 DOI: 10.1109/tcbb.2018.2849968] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
This article reviews recent advances in applying natural language processing (NLP) to Electronic Health Records (EHRs) for computational phenotyping. NLP-based computational phenotyping has numerous applications including diagnosis categorization, novel phenotype discovery, clinical trial screening, pharmacogenomics, drug-drug interaction (DDI), and adverse drug event (ADE) detection, as well as genome-wide and phenome-wide association studies. Significant progress has been made in algorithm development and resource construction for computational phenotyping. Among the surveyed methods, well-designed keyword search and rule-based systems often achieve good performance. However, the construction of keyword and rule lists requires significant manual effort, which is difficult to scale. Supervised machine learning models have been favored because they are capable of acquiring both classification patterns and structures from data. Recently, deep learning and unsupervised learning have received growing attention, with the former favored for its performance and the latter for its ability to find novel phenotypes. Integrating heterogeneous data sources have become increasingly important and have shown promise in improving model performance. Often, better performance is achieved by combining multiple modalities of information. Despite these many advances, challenges and opportunities remain for NLP-based computational phenotyping, including better model interpretability and generalizability, and proper characterization of feature relations in clinical narratives.
Collapse
Affiliation(s)
- Zexian Zeng
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611.
| | - Yu Deng
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611.
| | - Xiaoyu Li
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA 02115.
| | - Tristan Naumann
- Science and Artificial Intelligence Lab, Massachusetts Institue of Technology, Cambridge, MA 02139.
| | - Yuan Luo
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611.
| |
Collapse
|
511
|
|
512
|
Mehta D, Czamara D. GWAS of Behavioral Traits. Curr Top Behav Neurosci 2019; 42:1-34. [PMID: 31407241 DOI: 10.1007/7854_2019_105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Over the past decade, genome-wide association studies (GWAS) have evolved into a powerful tool to investigate genetic risk factors for human diseases via a hypothesis-free scan of the genome. The success of GWAS for psychiatric disorders and behavioral traits have been somewhat mixed, partly owing to the complexity and heterogeneity of these traits. Significant progress has been made in the last few years in the development and implementation of complex statistical methods and algorithms incorporating GWAS. Such advanced statistical methods applied to GWAS hits in combination with incorporation of different layers of genomics data have catapulted the search for novel genes for behavioral traits and improved our understanding of the complex polygenic architecture of these traits.This chapter will give a brief overview on GWAS and statistical methods currently used in GWAS. The chapter will focus on reviewing the current literature and highlight some of the most important GWAS on psychiatric and other behavioral traits and will conclude with a discussion on future directions.
Collapse
Affiliation(s)
- Divya Mehta
- School of Psychology and Counselling, Faculty of Health, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia.
| | - Darina Czamara
- Department of Translational Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
513
|
Chaganti S, Mawn LA, Kang H, Egan J, Resnick SM, Beason-Held LL, Landman BA, Lasko TA. Electronic Medical Record Context Signatures Improve Diagnostic Classification Using Medical Image Computing. IEEE J Biomed Health Inform 2018; 23:2052-2062. [PMID: 30602428 DOI: 10.1109/jbhi.2018.2890084] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Composite models that combine medical imaging with electronic medical records (EMR) improve predictive power when compared to traditional models that use imaging alone. The digitization of EMR provides potential access to a wealth of medical information, but presents new challenges in algorithm design and inference. Previous studies, such as Phenome Wide Association Study (PheWAS), have shown that EMR data can be used to investigate the relationship between genotypes and clinical conditions. Here, we introduce Phenome-Disease Association Study to extend the statistical capabilities of the PheWAS software through a custom Python package, which creates diagnostic EMR signatures to capture system-wide co-morbidities for a disease population within a given time interval. We investigate the effect of integrating these EMR signatures with radiological data to improve diagnostic classification in disease domains known to have confounding factors because of variable and complex clinical presentation. Specifically, we focus on two studies: First, a study of four major optic nerve related conditions; and second, a study of diabetes. Addition of EMR signature vectors to radiologically derived structural metrics improves the area under the curve (AUC) for diagnostic classification using elastic net regression, for diseases of the optic nerve. For glaucoma, the AUC improves from 0.71 to 0.83, for intrinsic optic nerve disease it increases from 0.72 to 0.91, for optic nerve edema it increases from 0.95 to 0.96, and for thyroid eye disease from 0.79 to 0.89. The EMR signatures recapitulate known comorbidities with diabetes, such as abnormal glucose, but do not significantly modulate image-derived features. In summary, EMR signatures present a scalable and readily applicable.
Collapse
|
514
|
Zeng Z, Espino S, Roy A, Li X, Khan SA, Clare SE, Jiang X, Neapolitan R, Luo Y. Using natural language processing and machine learning to identify breast cancer local recurrence. BMC Bioinformatics 2018; 19:498. [PMID: 30591037 PMCID: PMC6309052 DOI: 10.1186/s12859-018-2466-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Identifying local recurrences in breast cancer from patient data sets is important for clinical research and practice. Developing a model using natural language processing and machine learning to identify local recurrences in breast cancer patients can reduce the time-consuming work of a manual chart review. METHODS We design a novel concept-based filter and a prediction model to detect local recurrences using EHRs. In the training dataset, we manually review a development corpus of 50 progress notes and extract partial sentences that indicate breast cancer local recurrence. We process these partial sentences to obtain a set of Unified Medical Language System (UMLS) concepts using MetaMap, and we call it positive concept set. We apply MetaMap on patients' progress notes and retain only the concepts that fall within the positive concept set. These features combined with the number of pathology reports recorded for each patient are used to train a support vector machine to identify local recurrences. RESULTS We compared our model with three baseline classifiers using either full MetaMap concepts, filtered MetaMap concepts, or bag of words. Our model achieved the best AUC (0.93 in cross-validation, 0.87 in held-out testing). CONCLUSIONS Compared to a labor-intensive chart review, our model provides an automated way to identify breast cancer local recurrences. We expect that by minimally adapting the positive concept set, this study has the potential to be replicated at other institutions with a moderately sized training dataset.
Collapse
Affiliation(s)
- Zexian Zeng
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Sasa Espino
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ankita Roy
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Xiaoyu Li
- Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Seema A Khan
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Susan E Clare
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Xia Jiang
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Richard Neapolitan
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Yuan Luo
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
515
|
Liu Y, Wan Z, Xia W, Kantarcioglu M, Vorobeychik Y, Clayton EW, Kho A, Carrell D, Malin BA. Detecting the Presence of an Individual in Phenotypic Summary Data. AMIA ... ANNUAL SYMPOSIUM PROCEEDINGS. AMIA SYMPOSIUM 2018; 2018:760-769. [PMID: 30815118 PMCID: PMC6371366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
As the quantity and detail of association studies between clinical phenotypes and genotypes grows, there is a push to make summary statistics widely available. Genome wide summary statistics have been shown to be vulnerable to the inference of a targeted individual's presence. In this paper, we show that presence attacks are feasible with phenome wide summary statistics as well. We use data from three healthcare organizations and an online resource that publishes summary statistics. We introduce a novel attack that achieves over 80% recall and precision within a population of 16,346, where 8,173 individuals are targets. However, the feasibility of the attack is dependent on the attacker's knowledge about 1) the targeted individual and 2) the reference dataset. Within a population of over 2 million, where 8,173 individuals are targets, our attack achieves 31% recall and 17% precision. As a result, it is plausible that sharing of phenomic summary statistics may be accomplished with an acceptable level of privacy risk.
Collapse
Affiliation(s)
- Yongtai Liu
- Vanderbilt University, Nashville, Tennessee, USA
| | - Zhiyu Wan
- Vanderbilt University, Nashville, Tennessee, USA
| | - Weiyi Xia
- Vanderbilt University, Nashville, Tennessee, USA
| | | | | | | | - Abel Kho
- Northwestern University, Chicago, Illinois, USA
| | - David Carrell
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington, USA
| | | |
Collapse
|
516
|
Fuentes R, Letelier J, Tajer B, Valdivia LE, Mullins MC. Fishing forward and reverse: Advances in zebrafish phenomics. Mech Dev 2018; 154:296-308. [PMID: 30130581 PMCID: PMC6289646 DOI: 10.1016/j.mod.2018.08.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 08/06/2018] [Accepted: 08/17/2018] [Indexed: 12/15/2022]
Abstract
Understanding how the genome instructs the phenotypic characteristics of an organism is one of the major scientific endeavors of our time. Advances in genetics have progressively deciphered the inheritance, identity and biological relevance of genetically encoded information, contributing to the rise of several, complementary omic disciplines. One of them is phenomics, an emergent area of biology dedicated to the systematic multi-scale analysis of phenotypic traits. This discipline provides valuable gene function information to the rapidly evolving field of genetics. Current molecular tools enable genome-wide analyses that link gene sequence to function in multi-cellular organisms, illuminating the genome-phenome relationship. Among vertebrates, zebrafish has emerged as an outstanding model organism for high-throughput phenotyping and modeling of human disorders. Advances in both systematic mutagenesis and phenotypic analyses of embryonic and post-embryonic stages in zebrafish have revealed the function of a valuable collection of genes and the general structure of several complex traits. In this review, we summarize multiple large-scale genetic efforts addressing parental, embryonic, and adult phenotyping in the zebrafish. The genetic and quantitative tools available in the zebrafish model, coupled with the broad spectrum of phenotypes that can be assayed, make it a powerful model for phenomics, well suited for the dissection of genotype-phenotype associations in development, physiology, health and disease.
Collapse
Affiliation(s)
- Ricardo Fuentes
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joaquín Letelier
- Centro Andaluz de Biología del Desarrollo (CSIC/UPO/JA), Seville, Spain; Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Benjamin Tajer
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Leonardo E Valdivia
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.
| | - Mary C Mullins
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
517
|
Justice AC, Smith RV, Tate JP, McGinnis K, Xu K, Becker WC, Lee KY, Lynch K, Sun N, Concato J, Fiellin DA, Zhao H, Gelernter J, Kranzler HR, on behalf of the VA Million Veteran Program. AUDIT-C and ICD codes as phenotypes for harmful alcohol use: association with ADH1B polymorphisms in two US populations. Addiction 2018; 113:2214-2224. [PMID: 29972609 PMCID: PMC6226338 DOI: 10.1111/add.14374] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/31/2018] [Accepted: 06/28/2018] [Indexed: 01/28/2023]
Abstract
BACKGROUND AND AIMS Longitudinal electronic health record (EHR) data offer a large-scale, untapped source of phenotypical information on harmful alcohol use. Using established, alcohol-associated variants in the gene that encodes the enzyme alcohol dehydrogenase 1B (ADH1B) as criterion standards, we compared the individual and combined validity of three longitudinal EHR-based phenotypes of harmful alcohol use: Alcohol Use Disorders Identification Test-Consumption (AUDIT-C) trajectories; mean age-adjusted AUDIT-C; and diagnoses of alcohol use disorder (AUD). DESIGN With longitudinal EHR data from the Million Veteran Program (MVP) linked to genetic data, we used two population-specific polymorphisms in ADH1B that are associated strongly with AUD in African Americans (AAs) and European Americans (EAs): rs2066702 (Arg369Cys, AAs) and rs1229984 (Arg48His, EAs) as criterion measures. SETTING United States Department of Veterans Affairs Healthcare System. PARTICIPANTS A total of 167 721 veterans (57 677 AAs and 110 044 EAs; 92% male, mean age = 63 years) took part in this study. Data were collected from 1 October 2007 to 1 May 2017. MEASUREMENTS Using all AUDIT-C scores and AUD diagnostic codes recorded in the EHR, we calculated age-adjusted mean AUDIT-C values, longitudinal statistical trajectories of AUDIT-C scores and ICD-9/10 diagnostic groupings for AUD. FINDINGS A total of 19 793 AAs (34.3%) had one or two minor alleles at rs2066702 [minor allele frequency (MAF) = 0.190] and 6933 EAs (6.3%) had one or two minor alleles at rs1229984 (MAF = 0.032). In both populations, trajectories and age-adjusted mean AUDIT-C were correlated (r = 0.90) but, when considered separately, highest score (8+ versus 0) of age-adjusted mean AUDIT-C demonstrated a stronger association with the ADH1B variants [adjusted odds ratio (aOR) 0.54 in AAs and 0.37 in AAs] than did the highest trajectory (aOR 0.71 in AAs and 0.53 in EAs); combining AUDIT-C metrics did not improve discrimination. When age-adjusted mean AUDIT-C score and AUD diagnoses were considered together, age-adjusted mean AUDIT-C (8+ versus 0) was associated with lower odds of having the ADH1B minor allele than were AUD diagnostic codes: aOR = 0.59 versus 0.86 in AAs and 0.48 versus 0.68 in EAs. These independent associations combine to yield an even lower aOR of 0.51 for AAs and 0.33 for EAs. CONCLUSIONS The age-adjusted mean AUDIT-C score is associated more strongly with genetic polymorphisms of known risk for alcohol use disorder than are longitudinal trajectories of AUDIT-C or AUD diagnostic codes. AUD diagnostic codes modestly enhance this association.
Collapse
Affiliation(s)
- Amy C. Justice
- Yale School of Medicine, New Haven CT 06515,Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516,Yale School of Public Health, New Haven CT 06515
| | - Rachel V. Smith
- University of Louisville School of Nursing, Louisville, KY 40202
| | | | - Kathleen McGinnis
- Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
| | - Ke Xu
- Yale School of Medicine, New Haven CT 06515,Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
| | - William C. Becker
- Yale School of Medicine, New Haven CT 06515,Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
| | - Kuang-Yao Lee
- Department of Statistical Science, Temple University, Philadelphia, PA 19122
| | - Kevin Lynch
- VISN 4 MIRECC, Crescenz VAMC, Philadelphia, PA 19104,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | - Ning Sun
- Yale School of Medicine, New Haven CT 06515,Yale School of Public Health, New Haven CT 06515
| | - John Concato
- Yale School of Medicine, New Haven CT 06515,Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
| | - David A. Fiellin
- Yale School of Medicine, New Haven CT 06515,Yale School of Public Health, New Haven CT 06515
| | - Hongyu Zhao
- Yale School of Medicine, New Haven CT 06515,Yale School of Public Health, New Haven CT 06515
| | - Joel Gelernter
- Yale School of Medicine, New Haven CT 06515,Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516
| | - Henry R. Kranzler
- VISN 4 MIRECC, Crescenz VAMC, Philadelphia, PA 19104,University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104
| | | |
Collapse
|
518
|
Wilson JL, Racz R, Liu T, Adeniyi O, Sun J, Ramamoorthy A, Pacanowski M, Altman R. PathFX provides mechanistic insights into drug efficacy and safety for regulatory review and therapeutic development. PLoS Comput Biol 2018; 14:e1006614. [PMID: 30532240 PMCID: PMC6285459 DOI: 10.1371/journal.pcbi.1006614] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 10/31/2018] [Indexed: 12/14/2022] Open
Abstract
Failure to demonstrate efficacy and safety issues are important reasons that drugs do not reach the market. An incomplete understanding of how drugs exert their effects hinders regulatory and pharmaceutical industry projections of a drug's benefits and risks. Signaling pathways mediate drug response and while many signaling molecules have been characterized for their contribution to disease or their role in drug side effects, our knowledge of these pathways is incomplete. To better understand all signaling molecules involved in drug response and the phenotype associations of these molecules, we created a novel method, PathFX, a non-commercial entity, to identify these pathways and drug-related phenotypes. We benchmarked PathFX by identifying drugs' marketed disease indications and reported a sensitivity of 41%, a 2.7-fold improvement over similar approaches. We then used PathFX to strengthen signals for drug-adverse event pairs occurring in the FDA Adverse Event Reporting System (FAERS) and also identified opportunities for drug repurposing for new diseases based on interaction paths that associated a marketed drug to that disease. By discovering molecular interaction pathways, PathFX improved our understanding of drug associations to safety and efficacy phenotypes. The algorithm may provide a new means to improve regulatory and therapeutic development decisions.
Collapse
Affiliation(s)
- Jennifer L. Wilson
- Department of Bioengineering, Stanford University, Palo Alto California, United States of America
| | - Rebecca Racz
- Division of Applied Regulatory Science, US Food and Drug Administration, Silver Spring Maryland, United States of America
| | - Tianyun Liu
- Department of Bioengineering, Stanford University, Palo Alto California, United States of America
| | - Oluseyi Adeniyi
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring Maryland, United States of America
| | - Jielin Sun
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring Maryland, United States of America
| | - Anuradha Ramamoorthy
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring Maryland, United States of America
| | - Michael Pacanowski
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring Maryland, United States of America
| | - Russ Altman
- Department of Bioengineering, Stanford University, Palo Alto California, United States of America
- Department of Genetics, Stanford University, Palo Alto California, United States of America
| |
Collapse
|
519
|
Abstract
Data on disease burden are often used for assessing population health, evaluating the effectiveness of interventions, formulating health policies, and planning future resource allocation. We investigated whether Internet usage and social media data, specifically the search volume on Google, page view count on Wikipedia, and disease mentioning frequency on Twitter, correlated with the disease burden, measured by prevalence and treatment cost, for 1633 diseases over an 11-year period. We also applied least absolute shrinkage and selection operator to predict the burden of diseases. We found that Google search volume is relatively strongly correlated with the burdens for 39 of 1633 diseases, including viral hepatitis, diabetes mellitus, multiple sclerosis, and hemorrhoids. Wikipedia and Twitter data strongly correlated with the burdens of 15 and 7 diseases, respectively. However, an accurate analysis must consider each condition's characteristics, including acute/chronic nature, severity, familiarity to the public, and the presence of stigma.
Collapse
Affiliation(s)
| | | | - Sha Yu
- The University of North Carolina at Charlotte, USA
| | - Lixia Yao
- The University of North Carolina at Charlotte, USA; Mayo Clinic, USA
| |
Collapse
|
520
|
Verma M, Hontecillas R, Tubau-Juni N, Abedi V, Bassaganya-Riera J. Challenges in Personalized Nutrition and Health. Front Nutr 2018; 5:117. [PMID: 30555829 PMCID: PMC6281760 DOI: 10.3389/fnut.2018.00117] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 11/14/2018] [Indexed: 12/11/2022] Open
Affiliation(s)
- Meghna Verma
- Nutritional Immunology and Molecular Medicine Laboratory, Biocomplexity Institute of Virginia Tech, Blacksburg, VA, United States.,Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, Blacksburg, VA, United States
| | - Raquel Hontecillas
- Nutritional Immunology and Molecular Medicine Laboratory, Biocomplexity Institute of Virginia Tech, Blacksburg, VA, United States
| | - Nuria Tubau-Juni
- Nutritional Immunology and Molecular Medicine Laboratory, Biocomplexity Institute of Virginia Tech, Blacksburg, VA, United States
| | - Vida Abedi
- Nutritional Immunology and Molecular Medicine Laboratory, Biocomplexity Institute of Virginia Tech, Blacksburg, VA, United States.,Department of Biomedical and Translational Informatics, Geisinger Health System, Danville, PA, United States
| | - Josep Bassaganya-Riera
- Nutritional Immunology and Molecular Medicine Laboratory, Biocomplexity Institute of Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
521
|
Low YS, Alves VM, Fourches D, Sedykh A, Andrade CH, Muratov EN, Rusyn I, Tropsha A. Chemistry-Wide Association Studies (CWAS): A Novel Framework for Identifying and Interpreting Structure-Activity Relationships. J Chem Inf Model 2018; 58:2203-2213. [PMID: 30376324 DOI: 10.1021/acs.jcim.8b00450] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Quantitative structure-activity relationships (QSAR) models are often seen as a "black box" because they are considered difficult to interpret. Meanwhile, qualitative approaches, e.g., structural alerts (SA) or read-across, provide mechanistic insight, which is preferred for regulatory purposes, but predictive accuracy of such approaches is often low. Herein, we introduce the chemistry-wide association study (CWAS) approach, a novel framework that both addresses such deficiencies and combines advantages of statistical QSAR and alert-based approaches. The CWAS framework consists of the following steps: (i) QSAR model building for an end point of interest, (ii) identification of key chemical features, (iii) determination of communities of such features disproportionately co-occurring more frequently in the active than in the inactive class, and (iv) assembling these communities to form larger (and not necessarily chemically connected) novel structural alerts with high specificity. As a proof-of-concept, we have applied CWAS to model Ames mutagenicity and Stevens-Johnson Syndrome (SJS). For the well-studied Ames mutagenicity data set, we identified 76 important individual fragments and assembled co-occurring fragments into SA both replicative of known as well as representing novel mutagenicity alerts. For the SJS data set, we identified 29 important fragments and assembled co-occurring communities into SA including both known and novel alerts. In summary, we demonstrate that CWAS provides a new framework to interpret predictive QSAR models and derive refined structural alerts for more effective design and safety assessment of drugs and drug candidates.
Collapse
Affiliation(s)
- Yen S Low
- Laboratory for Molecular Modeling, UNC Eshelman School of Pharmacy , University of North Carolina , Chapel Hill , North Carolina 27599 , United States
| | - Vinicius M Alves
- Laboratory for Molecular Modeling, UNC Eshelman School of Pharmacy , University of North Carolina , Chapel Hill , North Carolina 27599 , United States.,Laboratory for Molecular Modeling and Design, Department of Pharmacy , Federal University of Goias , Goiania , Goias 74605-170 , Brazil
| | - Denis Fourches
- Department of Chemistry and Bioinformatics Research Center , North Carolina State University , Raleigh , North Carolina 27695 , United States
| | - Alexander Sedykh
- Sciome LLC , Research Triangle Park , North Carolina 27709 , United States
| | - Carolina Horta Andrade
- Laboratory for Molecular Modeling and Design, Department of Pharmacy , Federal University of Goias , Goiania , Goias 74605-170 , Brazil
| | - Eugene N Muratov
- Laboratory for Molecular Modeling, UNC Eshelman School of Pharmacy , University of North Carolina , Chapel Hill , North Carolina 27599 , United States.,Department of Chemical Technology , Odessa National Polytechnic University , Odessa 65000 , Ukraine
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences , Texas A&M University , College Station , Texas 77843 , United States
| | - Alexander Tropsha
- Laboratory for Molecular Modeling, UNC Eshelman School of Pharmacy , University of North Carolina , Chapel Hill , North Carolina 27599 , United States
| |
Collapse
|
522
|
Mahajan A, Taliun D, Thurner M, Robertson NR, Torres JM, Rayner NW, Steinthorsdottir V, Scott RA, Grarup N, Cook JP, Schmidt EM, Wuttke M, Sarnowski C, Mägi R, Nano J, Gieger C, Trompet S, Lecoeur C, Preuss M, Prins BP, Guo X, Bielak LF, DIAMANTE consortium, Bennett AJ, Bork-Jensen J, Brummett CM, Canouil M, Eckardt KU, Fischer K, Kardia SLR, Kronenberg F, Läll K, Liu CT, Locke AE, Luan J, Ntalla I, Nylander V, Schönherr S, Schurmann C, Yengo L, Bottinger EP, Brandslund I, Christensen C, Dedoussis G, Florez JC, ford I, Franco OH, Frayling TM, Giedraitis V, Hackinger S, Hattersley AT, Herder C, Ikram MA, Ingelsson M, Jørgensen ME, Jørgensen T, Kriebel J, Kuusisto J, Ligthart S, Lindgren CM, Linneberg A, Lyssenko V, Mamakou V, Meitinger T, Mohlke KL, Morris AD, Nadkarni G, Pankow JS, Peters A, Sattar N, Stančáková A, Strauch K, Taylor KD, Thorand B, Thorleifsson G, Thorsteinsdottir U, Tuomilehto J, Witte DR, Dupuis J, Peyser PA, Zeggini E, Loos RJF, Froguel P, Ingelsson E, Lind L, Groop L, Laakso M, Collins FS, Jukema JW, Palmer CNA, Grallert H, Metspalu A, Dehghan A, Köttgen A, Abecasis G, Meigs JB, Rotter JI, Marchini J, Pedersen O, Hansen T, et alMahajan A, Taliun D, Thurner M, Robertson NR, Torres JM, Rayner NW, Steinthorsdottir V, Scott RA, Grarup N, Cook JP, Schmidt EM, Wuttke M, Sarnowski C, Mägi R, Nano J, Gieger C, Trompet S, Lecoeur C, Preuss M, Prins BP, Guo X, Bielak LF, DIAMANTE consortium, Bennett AJ, Bork-Jensen J, Brummett CM, Canouil M, Eckardt KU, Fischer K, Kardia SLR, Kronenberg F, Läll K, Liu CT, Locke AE, Luan J, Ntalla I, Nylander V, Schönherr S, Schurmann C, Yengo L, Bottinger EP, Brandslund I, Christensen C, Dedoussis G, Florez JC, ford I, Franco OH, Frayling TM, Giedraitis V, Hackinger S, Hattersley AT, Herder C, Ikram MA, Ingelsson M, Jørgensen ME, Jørgensen T, Kriebel J, Kuusisto J, Ligthart S, Lindgren CM, Linneberg A, Lyssenko V, Mamakou V, Meitinger T, Mohlke KL, Morris AD, Nadkarni G, Pankow JS, Peters A, Sattar N, Stančáková A, Strauch K, Taylor KD, Thorand B, Thorleifsson G, Thorsteinsdottir U, Tuomilehto J, Witte DR, Dupuis J, Peyser PA, Zeggini E, Loos RJF, Froguel P, Ingelsson E, Lind L, Groop L, Laakso M, Collins FS, Jukema JW, Palmer CNA, Grallert H, Metspalu A, Dehghan A, Köttgen A, Abecasis G, Meigs JB, Rotter JI, Marchini J, Pedersen O, Hansen T, Langenberg C, Wareham NJ, Stefansson K, Gloyn AL, Morris AP, Boehnke M, McCarthy MI. Fine-mapping type 2 diabetes loci to single-variant resolution using high-density imputation and islet-specific epigenome maps. Nat Genet 2018; 50:1505-1513. [PMID: 30297969 PMCID: PMC6287706 DOI: 10.1038/s41588-018-0241-6] [Show More Authors] [Citation(s) in RCA: 1192] [Impact Index Per Article: 170.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 08/10/2018] [Indexed: 12/30/2022]
Abstract
We expanded GWAS discovery for type 2 diabetes (T2D) by combining data from 898,130 European-descent individuals (9% cases), after imputation to high-density reference panels. With these data, we (i) extend the inventory of T2D-risk variants (243 loci, 135 newly implicated in T2D predisposition, comprising 403 distinct association signals); (ii) enrich discovery of lower-frequency risk alleles (80 index variants with minor allele frequency <5%, 14 with estimated allelic odds ratio >2); (iii) substantially improve fine-mapping of causal variants (at 51 signals, one variant accounted for >80% posterior probability of association (PPA)); (iv) extend fine-mapping through integration of tissue-specific epigenomic information (islet regulatory annotations extend the number of variants with PPA >80% to 73); (v) highlight validated therapeutic targets (18 genes with associations attributable to coding variants); and (vi) demonstrate enhanced potential for clinical translation (genome-wide chip heritability explains 18% of T2D risk; individuals in the extremes of a T2D polygenic risk score differ more than ninefold in prevalence).
Collapse
Affiliation(s)
- Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| | - Daniel Taliun
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Matthias Thurner
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 7LE, UK
| | - Neil R Robertson
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 7LE, UK
| | - Jason M Torres
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| | - N William Rayner
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 7LE, UK
- Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | | | - Robert A Scott
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Niels Grarup
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - James P Cook
- Department of Biostatistics, University of Liverpool, Liverpool, L69 3GA, UK
| | - Ellen M Schmidt
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Matthias Wuttke
- Institute of Genetic Epidemiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany
| | - Chloé Sarnowski
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, 02118, USA
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu, 51010, Estonia
| | - Jana Nano
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015CN, The Netherlands
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Institute of Epidemiology 2, Helmholtz Zentrum München, German Research Center for Environmental Health, München-Neuherberg, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Stella Trompet
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden, 2300 RC, the Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, 2300 RC, the Netherlands
| | - Cécile Lecoeur
- CNRS-UMR8199, Lille University, Lille Pasteur Institute, Lille, 59000, France
| | - Michael Preuss
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, 10029, USA
| | - Bram Peter Prins
- Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Xiuqing Guo
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, California, 90502, US
| | - Lawrence F Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | | | - Amanda J Bennett
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 7LE, UK
| | - Jette Bork-Jensen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Chad M Brummett
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI, 48109, US
| | - Mickaël Canouil
- CNRS-UMR8199, Lille University, Lille Pasteur Institute, Lille, 59000, France
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care Charité, University Medicine Berlin, Berlin, 10117, Germany and German Chronic Kidney Disease study
| | - Krista Fischer
- Estonian Genome Center, University of Tartu, Tartu, 51010, Estonia
| | - Sharon LR Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Florian Kronenberg
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, 6020, Austria and German Chronic Kidney Disease study
| | - Kristi Läll
- Estonian Genome Center, University of Tartu, Tartu, 51010, Estonia
- Institute of Mathematical Statistics, University of Tartu, Tartu, Estonia
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, 02118, USA
| | - Adam E Locke
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, Division of Genomics & Bioinformatics, Washington University School of Medicine, St. Louis, MO, USA
| | - Jian'an Luan
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Ioanna Ntalla
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Vibe Nylander
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 7LE, UK
| | - Sebastian Schönherr
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, 6020, Austria and German Chronic Kidney Disease study
| | - Claudia Schurmann
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, 10029, USA
| | - Loïc Yengo
- CNRS-UMR8199, Lille University, Lille Pasteur Institute, Lille, 59000, France
| | - Erwin P Bottinger
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, 10029, USA
| | - Ivan Brandslund
- Institute of Regional Health Research, University of Southern Denmark, Odense, 5000, Denmark
- Department of Clinical Biochemistry, Vejle Hospital, Vejle, 7100, Denmark
| | | | - George Dedoussis
- Department of Nutrition and Dietetics, Harokopio University of Athens, Athens, 17671, Greece
| | - Jose C Florez
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, 02115, USA
- Diabetes Research Center (Diabetes Unit), Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, 02114, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute, Cambridge, MA, 02142, USA
| | - Ian ford
- Robertson Centre for Biostatistics, University of Glasgow, Glasgow, UK
| | - Oscar H Franco
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015CN, The Netherlands
| | - Timothy M Frayling
- Genetics of Complex Traits, University of Exeter Medical School, University of Exeter, Exeter, EX1 2LU, UK
| | - Vilmantas Giedraitis
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, SE-751 85, Sweden
| | - Sophie Hackinger
- Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Andrew T Hattersley
- University of Exeter Medical School, University of Exeter, Exeter, EX2 5DW, UK
| | - Christian Herder
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015CN, The Netherlands
| | - Martin Ingelsson
- Department of Public Health and Caring Sciences, Geriatrics, Uppsala University, Uppsala, SE-751 85, Sweden
| | - Marit E Jørgensen
- Steno Diabetes Center Copenhagen, Gentofte, 2820, Denmark
- National Institute of Public Health, Southern Denmark University, Copenhagen, 1353, Denmark
| | - Torben Jørgensen
- Research Centre for Prevention and Health, Capital Region of Denmark, Glostrup, 2600, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - Jennifer Kriebel
- Research Unit of Molecular Epidemiology, Institute of Epidemiology II, Helmholtz Zentrum München Research Center for Environmental Health, Neuherberg, 85764, Germany
- German Center for Diabetes Research (DZD), Neuherberg, 85764, Germany
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Symen Ligthart
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015CN, The Netherlands
| | - Cecilia M Lindgren
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Program in Medical and Population Genetics, Broad Institute, Cambridge, Massachusetts, 02142, USA
- Big Data Institute, Li Ka Shing Centre For Health Information and Discovery, University of Oxford, Oxford, OX37BN, UK
| | - Allan Linneberg
- Research Centre for Prevention and Health, Capital Region of Denmark, Glostrup, 2600, Denmark
- Department of Clinical Experimental Research, Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Valeriya Lyssenko
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University Diabetes Centre, Malmö, 20502, Sweden
| | - Vasiliki Mamakou
- Dromokaiteio Psychiatric Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Thomas Meitinger
- Institute of Human Genetics, Technische Universität München, Munich, 81675, Germany
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, 27599, USA
| | - Andrew D Morris
- Clinical Research Centre, Centre for Molecular Medicine, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
- The Usher Institute to the Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, EH16 4UX, UK
| | - Girish Nadkarni
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10069, USA
| | - James S Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, 55454, US
| | - Annette Peters
- German Center for Diabetes Research (DZD), Neuherberg, 85764, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, 81675, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Alena Stančáková
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, 85764, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität, Munich, 80802, Germany
| | - Kent D Taylor
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, California, 90502, US
| | - Barbara Thorand
- German Center for Diabetes Research (DZD), Neuherberg, 85764, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, 85764, Germany
| | | | - Unnur Thorsteinsdottir
- deCODE Genetics, Amgen inc., Reykjavik, 101, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | - Jaakko Tuomilehto
- Department of Health, National Institute for Health and Welfare, Helsinki, 00271, Finland
- Dasman Diabetes Institute, Dasman, 15462, Kuwait
- Department of Neuroscience and Preventive Medicine, Danube-University Krems, Krems, 3500, Austria
- Diabetes Research Group, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Daniel R Witte
- Department of Public Health, Aarhus University, Aarhus, Denmark
- Danish Diabetes Academy, Odense, Denmark
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, 02118, USA
- National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts, 01702, USA
| | - Patricia A Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Eleftheria Zeggini
- Department of Human Genetics, Wellcome Trust Sanger Institute, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, 10029, USA
- Mindich Child Health and Development Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Philippe Froguel
- CNRS-UMR8199, Lille University, Lille Pasteur Institute, Lille, 59000, France
- Department of Genomics of Common Disease, School of Public Health, Imperial College London, London, W12 0NN, UK
| | - Erik Ingelsson
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, 94305, US
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, 75185, Sweden
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, SE-751 85, Sweden
| | - Leif Groop
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University Diabetes Centre, Malmö, 20502, Sweden
- Finnish Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, 70210, Finland
| | - Francis S Collins
- Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, 2300 RC, the Netherlands
| | - Colin N A Palmer
- Pat Macpherson Centre for Pharmacogenetics and Pharmacogenomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Harald Grallert
- Research Unit of Molecular Epidemiology, Institute of Epidemiology II, Helmholtz Zentrum München Research Center for Environmental Health, Neuherberg, 85764, Germany
- German Center for Diabetes Research (DZD), Neuherberg, 85764, Germany
- Clinical Cooparation Group Type 2 Diabetes, Helmholtz Zentrum München, Ludwig-Maximillians University Munich, Germany
- Clinical Cooparation Group Nutrigenomics and Type 2 Diabetes, Helmholtz Zentrum München, Technical University Munich, Germany
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu, 51010, Estonia
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, 3015CN, The Netherlands
- Department of Epidemiology and Biostatistics, Imperial College London, London, W2 1PG, UK
- MRC-PHE Centre for Environment and Health, Imperial College London, London, W2 1PG, UK
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany
| | - Goncalo Abecasis
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - James B Meigs
- General Medicine Division, Massachusetts General Hospital and Department of Medicine, Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Jerome I Rotter
- Departments of Pediatrics and Medicine, The Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, California, 90502, US
| | - Jonathan Marchini
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Department of Statistics, University of Oxford, Oxford, OX1 3TG, UK
| | - Oluf Pedersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
- Faculty of Health Sciences, University of Southern Denmark, Odense, 5000, Denmark
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Kari Stefansson
- deCODE Genetics, Amgen inc., Reykjavik, 101, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, 101, Iceland
| | - Anna L Gloyn
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 7LE, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, OX3 7LE, UK
| | - Andrew P Morris
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Department of Biostatistics, University of Liverpool, Liverpool, L69 3GA, UK
- Estonian Genome Center, University of Tartu, Tartu, 51010, Estonia
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, Michigan, 48109, USA
| | - Mark I McCarthy
- Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 7LE, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, OX3 7LE, UK
| |
Collapse
|
523
|
Cronin RM, Conway D, Condon D, Jerome RN, Byrne DW, Harris PA. Patient and healthcare provider views on a patient-reported outcomes portal. J Am Med Inform Assoc 2018; 25:1470-1480. [PMID: 30239733 PMCID: PMC6213079 DOI: 10.1093/jamia/ocy111] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/09/2018] [Accepted: 07/24/2018] [Indexed: 12/20/2022] Open
Abstract
Background Over the past decade, public interest in managing health-related information for personal understanding and self-improvement has rapidly expanded. This study explored aspects of how patient-provided health information could be obtained through an electronic portal and presented to inform and engage patients while also providing information for healthcare providers. Methods We invited participants using ResearchMatch from 2 cohorts: (1) self-reported healthy volunteers (no medical conditions) and (2) individuals with a self-reported diagnosis of anxiety and/or depression. Participants used a secure web application (dashboard) to complete the PROMIS® domain survey(s) and then complete a feedback survey. A community engagement studio with 5 healthcare providers assessed perspectives on the feasibility and features of a portal to collect and display patient provided health information. We used bivariate analyses and regression analyses to determine differences between cohorts. Results A total of 480 participants completed the study (239 healthy, 241 anxiety and/or depression). While participants from the tw2o cohorts had significantly different PROMIS scores (p < .05), both cohorts welcomed the concept of a patient-centric dashboard, saw value in sharing results with their healthcare provider, and wanted to view results over time. However, factors needing consideration before widespread use included personalization for the patient and their health issues, integration with existing information (eg electronic health records), and integration into clinician workflow. Conclusions Our findings demonstrated a strong desire among healthy people, patients with chronic diseases, and healthcare providers for a self-assessment portal that can collect patient-reported outcome metrics and deliver personalized feedback.
Collapse
Affiliation(s)
- Robert M Cronin
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Douglas Conway
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - David Condon
- Department of Medical Social Sciences, Northwestern University, Chicago, IL, USA
| | - Rebecca N Jerome
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Daniel W Byrne
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Paul A Harris
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
524
|
Genomic and Phenomic Research in the 21st Century. Trends Genet 2018; 35:29-41. [PMID: 30342790 DOI: 10.1016/j.tig.2018.09.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 09/24/2018] [Accepted: 09/25/2018] [Indexed: 02/06/2023]
Abstract
The field of human genomics has changed dramatically over time. Initial genomic studies were predominantly restricted to rare disorders in small families. Over the past decade, researchers changed course from family-based studies and instead focused on common diseases and traits in populations of unrelated individuals. With further advancements in biobanking, computer science, electronic health record (EHR) data, and more affordable high-throughput genomics, we are experiencing a new paradigm in human genomic research. Rapidly changing technologies and resources now make it possible to study thousands of diseases simultaneously at the genomic level. This review will focus on these advancements as scientists begin to incorporate phenome-wide strategies in human genomic research to understand the etiology of human diseases and develop new drugs to treat them.
Collapse
|
525
|
Yu KH, Miron O, Palmer N, Lemos DR, Fox K, Kou SC, Sahin M, Kohane IS. Data-driven analyses revealed the comorbidity landscape of tuberous sclerosis complex. Neurology 2018; 91:974-976. [PMID: 30333165 DOI: 10.1212/wnl.0000000000006546] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 08/27/2018] [Indexed: 11/15/2022] Open
Affiliation(s)
- Kun-Hsing Yu
- From Harvard Medical School (K.-H.Y., O.M., N.P., I.S.K.), Boston; Harvard University (K.-H.Y., S.C.K.), Cambridge; Brigham and Women's Hospital (D.R.L.), Boston, MA; Aetna Inc. (K.F.), Hartford, CT; and Boston Children's Hospital (M.S., I.S.K.), MA
| | - Oren Miron
- From Harvard Medical School (K.-H.Y., O.M., N.P., I.S.K.), Boston; Harvard University (K.-H.Y., S.C.K.), Cambridge; Brigham and Women's Hospital (D.R.L.), Boston, MA; Aetna Inc. (K.F.), Hartford, CT; and Boston Children's Hospital (M.S., I.S.K.), MA
| | - Nathan Palmer
- From Harvard Medical School (K.-H.Y., O.M., N.P., I.S.K.), Boston; Harvard University (K.-H.Y., S.C.K.), Cambridge; Brigham and Women's Hospital (D.R.L.), Boston, MA; Aetna Inc. (K.F.), Hartford, CT; and Boston Children's Hospital (M.S., I.S.K.), MA
| | - Dario R Lemos
- From Harvard Medical School (K.-H.Y., O.M., N.P., I.S.K.), Boston; Harvard University (K.-H.Y., S.C.K.), Cambridge; Brigham and Women's Hospital (D.R.L.), Boston, MA; Aetna Inc. (K.F.), Hartford, CT; and Boston Children's Hospital (M.S., I.S.K.), MA
| | - Kathe Fox
- From Harvard Medical School (K.-H.Y., O.M., N.P., I.S.K.), Boston; Harvard University (K.-H.Y., S.C.K.), Cambridge; Brigham and Women's Hospital (D.R.L.), Boston, MA; Aetna Inc. (K.F.), Hartford, CT; and Boston Children's Hospital (M.S., I.S.K.), MA
| | - S C Kou
- From Harvard Medical School (K.-H.Y., O.M., N.P., I.S.K.), Boston; Harvard University (K.-H.Y., S.C.K.), Cambridge; Brigham and Women's Hospital (D.R.L.), Boston, MA; Aetna Inc. (K.F.), Hartford, CT; and Boston Children's Hospital (M.S., I.S.K.), MA
| | - Mustafa Sahin
- From Harvard Medical School (K.-H.Y., O.M., N.P., I.S.K.), Boston; Harvard University (K.-H.Y., S.C.K.), Cambridge; Brigham and Women's Hospital (D.R.L.), Boston, MA; Aetna Inc. (K.F.), Hartford, CT; and Boston Children's Hospital (M.S., I.S.K.), MA
| | - Isaac S Kohane
- From Harvard Medical School (K.-H.Y., O.M., N.P., I.S.K.), Boston; Harvard University (K.-H.Y., S.C.K.), Cambridge; Brigham and Women's Hospital (D.R.L.), Boston, MA; Aetna Inc. (K.F.), Hartford, CT; and Boston Children's Hospital (M.S., I.S.K.), MA.
| |
Collapse
|
526
|
Diogo D, Tian C, Franklin CS, Alanne-Kinnunen M, March M, Spencer CCA, Vangjeli C, Weale ME, Mattsson H, Kilpeläinen E, Sleiman PMA, Reilly DF, McElwee J, Maranville JC, Chatterjee AK, Bhandari A, Nguyen KDH, Estrada K, Reeve MP, Hutz J, Bing N, John S, MacArthur DG, Salomaa V, Ripatti S, Hakonarson H, Daly MJ, Palotie A, Hinds DA, Donnelly P, Fox CS, Day-Williams AG, Plenge RM, Runz H. Phenome-wide association studies across large population cohorts support drug target validation. Nat Commun 2018; 9:4285. [PMID: 30327483 PMCID: PMC6191429 DOI: 10.1038/s41467-018-06540-3] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 09/05/2018] [Indexed: 12/12/2022] Open
Abstract
Phenome-wide association studies (PheWAS) have been proposed as a possible aid in drug development through elucidating mechanisms of action, identifying alternative indications, or predicting adverse drug events (ADEs). Here, we select 25 single nucleotide polymorphisms (SNPs) linked through genome-wide association studies (GWAS) to 19 candidate drug targets for common disease indications. We interrogate these SNPs by PheWAS in four large cohorts with extensive health information (23andMe, UK Biobank, FINRISK, CHOP) for association with 1683 binary endpoints in up to 697,815 individuals and conduct meta-analyses for 145 mapped disease endpoints. Our analyses replicate 75% of known GWAS associations (P < 0.05) and identify nine study-wide significant novel associations (of 71 with FDR < 0.1). We describe associations that may predict ADEs, e.g., acne, high cholesterol, gout, and gallstones with rs738409 (p.I148M) in PNPLA3 and asthma with rs1990760 (p.T946A) in IFIH1. Our results demonstrate PheWAS as a powerful addition to the toolkit for drug discovery.
Collapse
Affiliation(s)
| | - Chao Tian
- 23andMe Inc, Mountain View, CA, 94041, USA
| | | | - Mervi Alanne-Kinnunen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014, Helsinki, Finland
| | - Michael March
- The Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | | | | | - Hannele Mattsson
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014, Helsinki, Finland
- National Institute for Health and Welfare, FI-00271, Helsinki, Finland
| | - Elina Kilpeläinen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014, Helsinki, Finland
| | - Patrick M A Sleiman
- The Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | - Joshua McElwee
- Merck Sharp & Dohme, Boston, MA, 02115, USA
- Nimbus Therapeutics, Cambridge, MA, 02139, USA
| | - Joseph C Maranville
- Merck Sharp & Dohme, Boston, MA, 02115, USA
- Celgene, Cambridge, MA, 02140, USA
| | - Arnaub K Chatterjee
- Merck Sharp & Dohme, Boston, MA, 02115, USA
- McKinsey & Co., Boston, MA, 02210, USA
| | - Aman Bhandari
- Merck Sharp & Dohme, Boston, MA, 02115, USA
- Vertex Pharmaceuticals, Boston, MA, 02210, USA
| | | | - Karol Estrada
- Biogen, Research and Early Development, Cambridge, MA, 02142, USA
| | | | | | - Nan Bing
- Pfizer, Cambridge, MA, 02139, USA
| | - Sally John
- Biogen, Research and Early Development, Cambridge, MA, 02142, USA
| | - Daniel G MacArthur
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Veikko Salomaa
- National Institute for Health and Welfare, FI-00271, Helsinki, Finland
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014, Helsinki, Finland
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Hakon Hakonarson
- The Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mark J Daly
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014, Helsinki, Finland
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Psychiatric & Neurodevelopmental Genetics Unit, Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | | | | | | | - Aaron G Day-Williams
- Merck Sharp & Dohme, Boston, MA, 02115, USA
- Biogen, Research and Early Development, Cambridge, MA, 02142, USA
| | - Robert M Plenge
- Merck Sharp & Dohme, Boston, MA, 02115, USA
- Celgene, Cambridge, MA, 02140, USA
| | - Heiko Runz
- Merck Sharp & Dohme, Boston, MA, 02115, USA.
- Biogen, Research and Early Development, Cambridge, MA, 02142, USA.
| |
Collapse
|
527
|
Stanaway IB, Hall TO, Rosenthal EA, Palmer M, Naranbhai V, Knevel R, Namjou-Khales B, Carroll RJ, Kiryluk K, Gordon AS, Linder J, Howell KM, Mapes BM, Lin FTJ, Joo YY, Hayes MG, Gharavi AG, Pendergrass SA, Ritchie MD, de Andrade M, Croteau-Chonka DC, Raychaudhuri S, Weiss ST, Lebo M, Amr SS, Carrell D, Larson EB, Chute CG, Rasmussen-Torvik LJ, Roy-Puckelwartz MJ, Sleiman P, Hakonarson H, Li R, Karlson EW, Peterson JF, Kullo IJ, Chisholm R, Denny JC, Jarvik GP, Crosslin DR. The eMERGE genotype set of 83,717 subjects imputed to ~40 million variants genome wide and association with the herpes zoster medical record phenotype. Genet Epidemiol 2018; 43:63-81. [PMID: 30298529 PMCID: PMC6375696 DOI: 10.1002/gepi.22167] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 08/10/2018] [Accepted: 08/28/2018] [Indexed: 12/30/2022]
Abstract
The Electronic Medical Records and Genomics (eMERGE) network is a network of medical centers with electronic medical records linked to existing biorepository samples for genomic discovery and genomic medicine research. The network sought to unify the genetic results from 78 Illumina and Affymetrix genotype array batches from 12 contributing medical centers for joint association analysis of 83,717 human participants. In this report, we describe the imputation of eMERGE results and methods to create the unified imputed merged set of genome‐wide variant genotype data. We imputed the data using the Michigan Imputation Server, which provides a missing single‐nucleotide variant genotype imputation service using the minimac3 imputation algorithm with the Haplotype Reference Consortium genotype reference set. We describe the quality control and filtering steps used in the generation of this data set and suggest generalizable quality thresholds for imputation and phenotype association studies. To test the merged imputed genotype set, we replicated a previously reported chromosome 6 HLA‐B herpes zoster (shingles) association and discovered a novel zoster‐associated loci in an epigenetic binding site near the terminus of chromosome 3 (3p29).
Collapse
Affiliation(s)
- Ian B Stanaway
- Department of Biomedical Informatics Medical Education, School of Medicine, University of Washington, Seattle, Washington
| | - Taryn O Hall
- Department of Biomedical Informatics Medical Education, School of Medicine, University of Washington, Seattle, Washington
| | - Elisabeth A Rosenthal
- Division of Medical Genetics, School of Medicine, University of Washington, Seattle, Washington
| | - Melody Palmer
- Division of Medical Genetics, School of Medicine, University of Washington, Seattle, Washington
| | - Vivek Naranbhai
- Department of Biomedical Informatics Medical Education, School of Medicine, University of Washington, Seattle, Washington.,Harvard Medical School, Harvard University, Cambridge, Massachusetts
| | - Rachel Knevel
- Harvard Medical School, Harvard University, Cambridge, Massachusetts
| | - Bahram Namjou-Khales
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Robert J Carroll
- Departments of Biomedical Informatics and Medicine, Vanderbilt University, Nashville, Tennessee
| | - Krzysztof Kiryluk
- Department of Medicine, Columbia University, New York City, New York
| | - Adam S Gordon
- Division of Medical Genetics, School of Medicine, University of Washington, Seattle, Washington
| | - Jodell Linder
- Vanderbilt Institute for Clinical and Translational Research, School of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Kayla Marie Howell
- Vanderbilt Institute for Clinical and Translational Research, School of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Brandy M Mapes
- Vanderbilt Institute for Clinical and Translational Research, School of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Frederick T J Lin
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | | | - M Geoffrey Hayes
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Ali G Gharavi
- Department of Medicine, Columbia University, New York City, New York
| | | | - Marylyn D Ritchie
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | - Soumya Raychaudhuri
- Harvard Medical School, Harvard University, Cambridge, Massachusetts.,Program in Medical and Population Genetics, Broad Institute of Massachusetts Technical Institute and Harvard University, Cambridge, Massachusetts
| | - Scott T Weiss
- Harvard Medical School, Harvard University, Cambridge, Massachusetts
| | - Matt Lebo
- Harvard Medical School, Harvard University, Cambridge, Massachusetts
| | - Sami S Amr
- Harvard Medical School, Harvard University, Cambridge, Massachusetts
| | - David Carrell
- Kaiser Permanente Washington Health Research Institute (Formerly Group Health Cooperative-Seattle), Kaiser Permanente, Seattle, Washington
| | - Eric B Larson
- Kaiser Permanente Washington Health Research Institute (Formerly Group Health Cooperative-Seattle), Kaiser Permanente, Seattle, Washington
| | - Christopher G Chute
- Schools of Medicine, Public Health, and Nursing, Johns Hopkins University, Baltimore, Maryland
| | | | | | - Patrick Sleiman
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | | | - Rongling Li
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Elizabeth W Karlson
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Josh F Peterson
- Departments of Biomedical Informatics and Medicine, Vanderbilt University, Nashville, Tennessee
| | | | - Rex Chisholm
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Joshua Charles Denny
- Departments of Biomedical Informatics and Medicine, Vanderbilt University, Nashville, Tennessee
| | - Gail P Jarvik
- Division of Medical Genetics, School of Medicine, University of Washington, Seattle, Washington
| | -
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - David R Crosslin
- Department of Biomedical Informatics Medical Education, School of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
528
|
Jerome RN, Pulley JM, Roden DM, Shirey-Rice JK, Bastarache LA, R Bernard G, B Ekstrom L, Lancaster WJ, Denny JC. Using Human 'Experiments of Nature' to Predict Drug Safety Issues: An Example with PCSK9 Inhibitors. Drug Saf 2018; 41:303-311. [PMID: 29185237 DOI: 10.1007/s40264-017-0616-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION When a new drug enters the market, its full array of side effects remains to be defined. Current surveillance approaches targeting these effects remain largely reactive. There is a need for development of methods to predict specific safety events that should be sought for a given new drug during development and postmarketing activities. OBJECTIVE We present here a safety signal identification approach applied to a new set of drug entities, inhibitors of the serine protease proprotein convertase subtilisin/kexin type 9 (PCSK9). METHODS Using phenome-wide association study (PheWAS) methods, we analyzed available genotype and clinical data from 29,722 patients, leveraging the known effects of changes in PCSK9 to identify novel phenotypes in which this protein and its inhibitors may have impact. RESULTS PheWAS revealed a significantly reduced risk of hypercholesterolemia (odds ratio [OR] 0.68, p = 7.6 × 10-4) in association with a known loss-of-function variant in PCSK9, R46L. Similarly, laboratory data indicated significantly reduced beta mean low-density lipoprotein cholesterol (- 14.47 mg/dL, p = 2.58 × 10-23) in individuals carrying the R46L variant. The R46L variant was also associated with an increased risk of spina bifida (OR 5.90, p = 2.7 × 10-4), suggesting that further investigation of potential connections between inhibition of PCSK9 and neural tube defects may be warranted. CONCLUSION This novel methodology provides an opportunity to put in place new mechanisms to assess the safety and long-term tolerability of PCSK9 inhibitors specifically, and other new agents in general, as they move into human testing and expanded clinical use.
Collapse
Affiliation(s)
- Rebecca N Jerome
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Jill M Pulley
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dan M Roden
- Office of Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jana K Shirey-Rice
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lisa A Bastarache
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Gordon R Bernard
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA
- Office of Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Leeland B Ekstrom
- Vanderbilt Institute for Clinical and Translational Research, Vanderbilt University Medical Center, Nashville, TN, USA
- Nashville Biosciences, Nashville, TN, USA
| | - William J Lancaster
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Joshua C Denny
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
529
|
Salisbury-Ruf CT, Bertram CC, Vergeade A, Lark DS, Shi Q, Heberling ML, Fortune NL, Okoye GD, Jerome WG, Wells QS, Fessel J, Moslehi J, Chen H, Roberts LJ, Boutaud O, Gamazon ER, Zinkel SS. Bid maintains mitochondrial cristae structure and function and protects against cardiac disease in an integrative genomics study. eLife 2018; 7:40907. [PMID: 30281024 PMCID: PMC6234033 DOI: 10.7554/elife.40907] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/27/2018] [Indexed: 01/07/2023] Open
Abstract
Bcl-2 family proteins reorganize mitochondrial membranes during apoptosis, to form pores and rearrange cristae. In vitro and in vivo analysis integrated with human genetics reveals a novel homeostatic mitochondrial function for Bcl-2 family protein Bid. Loss of full-length Bid results in apoptosis-independent, irregular cristae with decreased respiration. Bid-/- mice display stress-induced myocardial dysfunction and damage. A gene-based approach applied to a biobank, validated in two independent GWAS studies, reveals that decreased genetically determined BID expression associates with myocardial infarction (MI) susceptibility. Patients in the bottom 5% of the expression distribution exhibit >4 fold increased MI risk. Carrier status with nonsynonymous variation in Bid’s membrane binding domain, BidM148T, associates with MI predisposition. Furthermore, Bid but not BidM148T associates with Mcl-1Matrix, previously implicated in cristae stability; decreased MCL-1 expression associates with MI. Our results identify a role for Bid in homeostatic mitochondrial cristae reorganization, that we link to human cardiac disease. Cells contain specialized structures called mitochondria, which help to convert fuel into energy. These tiny energy factories have a unique double membrane, with a smooth outer and a folded inner lining. The folds, called cristae, provide a scaffold for the molecular machinery that produces chemical energy that the cell can use. The cristae are dynamic, and can change shape, condensing to increase energy output. Mitochondria also play a role in cell death. In certain situations, cristae can widen and release the proteins held within their folds. This can trigger a program of self-destruction in the cell. A family of proteins called Bcl-2 control such a ‘programmed cell death’ through the release of mitochondrial proteins. Some family members, including a protein called Bid, can reorganize cristae to regulate this cell-death program. When cells die, Bid proteins that had been split move to the mitochondria. But, even when cells are healthy, Bid molecules that are intact are always there, suggesting that this form of the protein may have another purpose. To investigate this further, Salisbury-Ruf, Bertram et al. used mice with Bid, and mice that lacked the protein. Without Bid, cells – including heart cells – struggled to work properly and used less oxygen than their normal counterparts. A closer look using electron microscopy revealed abnormalities in the cristae. However, adding ‘intact’ Bid proteins back in to the deficient cells restored them to normal. Moreover, without Bid, the mice hearts were less able to respond to an increased demand for energy. This decreased their performance and caused the formation of scars in the heart muscle called fibrosis, similar to a pattern observed in human patients following a heart attack. DNA data from an electronic health record database revealed a link between low levels of Bid genes and heart attack in humans, which was confirmed in further studies. In addition, a specific mutation in the Bid gene was found to affect its ability to regulate the formation of proper cristae. Combining evidence from mice with human genetics revealed new information about heart diseases. Mitochondrial health may be affected by a combination of specific variations in genes and changes in the Bid protein, which could affect heart attack risk. Understanding more about this association could help to identify and potentially reduce certain risk factors for heart attack.
Collapse
Affiliation(s)
- Christi T Salisbury-Ruf
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| | - Clinton C Bertram
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States
| | - Aurelia Vergeade
- Department of Pharmacology, Vanderbilt University, Nashville, United States
| | - Daniel S Lark
- Molecular Physiology and Biophysics, Vanderbilt University, Nashville, United States
| | - Qiong Shi
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States
| | - Marlene L Heberling
- Department of Biological Sciences, Vanderbilt University, Nashville, United States
| | - Niki L Fortune
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States
| | - G Donald Okoye
- Division of Cardiovascular Medicine and Cardio-oncology Program, Vanderbilt University Medical Center, Nashville, United States
| | - W Gray Jerome
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, United States
| | - Quinn S Wells
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States
| | - Josh Fessel
- Department of Medicine, Vanderbilt University Medical Center, Nashville, United States
| | - Javid Moslehi
- Division of Cardiovascular Medicine and Cardio-oncology Program, Vanderbilt University Medical Center, Nashville, United States
| | - Heidi Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, United States
| | - L Jackson Roberts
- Department of Pharmacology, Vanderbilt University, Nashville, United States.,Department of Medicine, Vanderbilt University Medical Center, Nashville, United States
| | - Olivier Boutaud
- Department of Pharmacology, Vanderbilt University, Nashville, United States
| | - Eric R Gamazon
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, United States.,Clare Hall, University of Cambridge, Cambridge, United Kingdom
| | - Sandra S Zinkel
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, United States.,Department of Medicine, Vanderbilt University Medical Center, Nashville, United States
| |
Collapse
|
530
|
Gkoutos GV, Schofield PN, Hoehndorf R. The anatomy of phenotype ontologies: principles, properties and applications. Brief Bioinform 2018; 19:1008-1021. [PMID: 28387809 PMCID: PMC6169674 DOI: 10.1093/bib/bbx035] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 02/05/2017] [Indexed: 12/14/2022] Open
Abstract
The past decade has seen an explosion in the collection of genotype data in domains as diverse as medicine, ecology, livestock and plant breeding. Along with this comes the challenge of dealing with the related phenotype data, which is not only large but also highly multidimensional. Computational analysis of phenotypes has therefore become critical for our ability to understand the biological meaning of genomic data in the biological sciences. At the heart of computational phenotype analysis are the phenotype ontologies. A large number of these ontologies have been developed across many domains, and we are now at a point where the knowledge captured in the structure of these ontologies can be used for the integration and analysis of large interrelated data sets. The Phenotype And Trait Ontology framework provides a method for formal definitions of phenotypes and associated data sets and has proved to be key to our ability to develop methods for the integration and analysis of phenotype data. Here, we describe the development and products of the ontological approach to phenotype capture, the formal content of phenotype ontologies and how their content can be used computationally.
Collapse
Affiliation(s)
| | | | - Robert Hoehndorf
- Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, King Abdullah University of Science and Technology, Thuwal
| |
Collapse
|
531
|
Rhoades SD, Bastarache L, Denny JC, Hughey JJ. Pulling the covers in electronic health records for an association study with self-reported sleep behaviors. Chronobiol Int 2018; 35:1702-1712. [PMID: 30183400 DOI: 10.1080/07420528.2018.1508152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The electronic health record (EHR) contains rich histories of clinical care, but has not traditionally been mined for information related to sleep habits. Here, we performed a retrospective EHR study based on a cohort of 3,652 individuals with self-reported sleep behaviors documented from visits to the sleep clinic. These individuals were obese (mean body mass index 33.6 kg/m2) and had a high prevalence of sleep apnea (60.5%), however we found sleep behaviors largely concordant with prior prospective cohort studies. In our cohort, average wake time was 1 hour later and average sleep duration was 40 minutes longer on weekends than on weekdays (p < 10-12). Sleep duration varied considerably as a function of age and tended to be longer in females and in whites. Additionally, through phenome-wide association analyses, we found an association of long weekend sleep with depression, and an unexpectedly large number of associations of long weekday sleep with mental health and neurological disorders (q < 0.05). We then sought to replicate previously published genetic associations with morning/evening preference on a subset of our cohort with extant genotyping data (n = 555). While those findings did not replicate in our cohort, a polymorphism (rs3754214) in high linkage disequilibrium with a previously published polymorphism near TARS2 was associated with long sleep duration (p < 0.01). Collectively, our results highlight the potential of the EHR for uncovering the correlates of human sleep in real-world populations.
Collapse
Affiliation(s)
- Seth D Rhoades
- a Department of Biomedical Informatics , Vanderbilt University Medical Center , Nashville , Tennessee , USA
| | - Lisa Bastarache
- a Department of Biomedical Informatics , Vanderbilt University Medical Center , Nashville , Tennessee , USA
| | - Joshua C Denny
- a Department of Biomedical Informatics , Vanderbilt University Medical Center , Nashville , Tennessee , USA.,b Department of Medicine , Vanderbilt University Medical Center , Nashville , Tennessee , USA
| | - Jacob J Hughey
- a Department of Biomedical Informatics , Vanderbilt University Medical Center , Nashville , Tennessee , USA.,c Department of Biological Sciences , Vanderbilt University , Nashville , Tennessee , USA
| |
Collapse
|
532
|
Cai T, Zhang Y, Ho YL, Link N, Sun J, Huang J, Cai TA, Damrauer S, Ahuja Y, Honerlaw J, Huang J, Costa L, Schubert P, Hong C, Gagnon D, Sun YV, Gaziano JM, Wilson P, Cho K, Tsao P, O’Donnell CJ, Liao KP. Association of Interleukin 6 Receptor Variant With Cardiovascular Disease Effects of Interleukin 6 Receptor Blocking Therapy: A Phenome-Wide Association Study. JAMA Cardiol 2018; 3:849-857. [PMID: 30090940 PMCID: PMC6233652 DOI: 10.1001/jamacardio.2018.2287] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 06/13/2018] [Indexed: 12/30/2022]
Abstract
Importance Electronic health record (EHR) biobanks containing clinical and genomic data on large numbers of individuals have great potential to inform drug discovery. Individuals with interleukin 6 receptor (IL6R) single-nucleotide polymorphisms (SNPs) who are not receiving IL6R blocking therapy have biomarker profiles similar to those treated with IL6R blockers. This gene-drug pair provides an example to test whether associations of IL6R SNPs with a broad range of phenotypes can inform which diseases may benefit from treatment with IL6R blockade. Objective To determine whether screening for clinical associations with the IL6R SNP in a phenome-wide association study (PheWAS) using EHR biobank data can identify drug effects from IL6R clinical trials. Design, Setting, and Participants Diagnosis codes and routine laboratory measurements were extracted from the VA Million Veteran Program (MVP); diagnosis codes were mapped to phenotype groups using published PheWAS methods. A PheWAS was performed by fitting logistic regression models for testing associations of the IL6R SNPs with 1342 phenotype groups and by fitting linear regression models for testing associations of the IL6R SNP with 26 routine laboratory measurements. Significance was reported using a false discovery rate of 0.05 or less. Findings were replicated in 2 independent cohorts using UK Biobank and Vanderbilt University Biobank data. The Million Veteran Program included 332 799 US veterans; the UK Biobank, 408 455 individuals from the general population of the United Kingdom; and the Vanderbilt University Biobank, 13 835 patients from a tertiary care center. Exposures IL6R SNPs (rs2228145; rs4129267). Main Outcomes and Measures Phenotypes defined by International Classification of Diseases, Ninth Revision codes. Results Of the 332 799 veterans included in the main cohort, 305 228 (91.7%) were men, and the mean (SD) age was 66.1 (13.6) years. The IL6R SNP was most strongly associated with a reduced risk of aortic aneurysm phenotypes (odds ratio, 0.87-0.90; 95% CI, 0.84-0.93) in the MVP. We observed known off-target effects of IL6R blockade from clinical trials (eg, higher hemoglobin level). The reduced risk for aortic aneurysms among those with the IL6R SNP in the MVP was replicated in the Vanderbilt University Biobank, and the reduced risk for coronary heart disease was replicated in the UK Biobank. Conclusions and Relevance In this proof-of-concept study, we demonstrated application of the PheWAS using large EHR biobanks to inform drug effects. The findings of an association of the IL6R SNP with reduced risk for aortic aneurysms correspond with the newest indication for IL6R blockade, giant cell arteritis, of which a major complication is aortic aneurysm.
Collapse
Affiliation(s)
- Tianxi Cai
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Yichi Zhang
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Yuk-Lam Ho
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Nicholas Link
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Jiehuan Sun
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Jie Huang
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Brigham and Women’s Hospital, Boston, Massachusetts
| | - Tianrun A. Cai
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Brigham and Women’s Hospital, Boston, Massachusetts
| | - Scott Damrauer
- Corporal Michael Crescenz Veterans Affairs Medical Center, Perlman School of Medicine, University of Pennsylvania, Philadelphia
| | - Yuri Ahuja
- Harvard Medical School, Boston, Massachusetts
| | | | - Jie Huang
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Lauren Costa
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Petra Schubert
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
| | - Chuan Hong
- Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - David Gagnon
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Boston University School of Public Health, Boston, Massachusetts
| | - Yan V. Sun
- Emory University Schools of Medicine and Public Health, Atlanta, Georgia
| | - J. Michael Gaziano
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Brigham and Women’s Hospital, Boston, Massachusetts
| | - Peter Wilson
- Emory University Schools of Medicine and Public Health, Atlanta, Georgia
- Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
| | - Kelly Cho
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Brigham and Women’s Hospital, Boston, Massachusetts
| | - Philip Tsao
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California
- Department of Medicine, Stanford University of Medicine, Stanford, California
| | - Christopher J. O’Donnell
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Associate Editor, JAMA Cardiology
| | - Katherine P. Liao
- Veterans Affairs Boston Healthcare System, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Brigham and Women’s Hospital, Boston, Massachusetts
| |
Collapse
|
533
|
A study paradigm integrating prospective epidemiologic cohorts and electronic health records to identify disease biomarkers. Nat Commun 2018; 9:3522. [PMID: 30166544 PMCID: PMC6117367 DOI: 10.1038/s41467-018-05624-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 07/13/2018] [Indexed: 01/05/2023] Open
Abstract
Defining the full spectrum of human disease associated with a biomarker is necessary to advance the biomarker into clinical practice. We hypothesize that associating biomarker measurements with electronic health record (EHR) populations based on shared genetic architectures would establish the clinical epidemiology of the biomarker. We use Bayesian sparse linear mixed modeling to calculate SNP weightings for 53 biomarkers from the Atherosclerosis Risk in Communities study. We use the SNP weightings to computed predicted biomarker values in an EHR population and test associations with 1139 diagnoses. Here we report 116 associations meeting a Bonferroni level of significance. A false discovery rate (FDR)-based significance threshold reveals more known and undescribed associations across a broad range of biomarkers, including biometric measures, plasma proteins and metabolites, functional assays, and behaviors. We confirm an inverse association between LDL-cholesterol level and septicemia risk in an independent epidemiological cohort. This approach efficiently discovers biomarker-disease associations. Biomarker identification requires prohibitively large cohorts with gene expression and phenotype data. The approach introduced here learns polygenic predictors of expression from genetic and expression data, used to infer biomarker levels in patients with genetic and disease information.
Collapse
|
534
|
An integrated clinical program and crowdsourcing strategy for genomic sequencing and Mendelian disease gene discovery. NPJ Genom Med 2018; 3:21. [PMID: 30131872 PMCID: PMC6089983 DOI: 10.1038/s41525-018-0060-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 04/06/2018] [Accepted: 07/06/2018] [Indexed: 12/18/2022] Open
Abstract
Despite major progress in defining the genetic basis of Mendelian disorders, the molecular etiology of many cases remains unknown. Patients with these undiagnosed disorders often have complex presentations and require treatment by multiple health care specialists. Here, we describe an integrated clinical diagnostic and research program using whole-exome and whole-genome sequencing (WES/WGS) for Mendelian disease gene discovery. This program employs specific case ascertainment parameters, a WES/WGS computational analysis pipeline that is optimized for Mendelian disease gene discovery with variant callers tuned to specific inheritance modes, an interdisciplinary crowdsourcing strategy for genomic sequence analysis, matchmaking for additional cases, and integration of the findings regarding gene causality with the clinical management plan. The interdisciplinary gene discovery team includes clinical, computational, and experimental biomedical specialists who interact to identify the genetic etiology of the disease, and when so warranted, to devise improved or novel treatments for affected patients. This program effectively integrates the clinical and research missions of an academic medical center and affords both diagnostic and therapeutic options for patients suffering from genetic disease. It may therefore be germane to other academic medical institutions engaged in implementing genomic medicine programs.
Collapse
|
535
|
Prokop JW, May T, Strong K, Bilinovich SM, Bupp C, Rajasekaran S, Worthey EA, Lazar J. Genome sequencing in the clinic: the past, present, and future of genomic medicine. Physiol Genomics 2018; 50:563-579. [PMID: 29727589 PMCID: PMC6139636 DOI: 10.1152/physiolgenomics.00046.2018] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Genomic sequencing has undergone massive expansion in the past 10 yr, from a rarely used research tool into an approach that has broad applications in a clinical setting. From rare disease to cancer, genomics is transforming our knowledge of biology. The transition from targeted gene sequencing, to whole exome sequencing, to whole genome sequencing has only been made possible due to rapid advancements in technologies and informatics that have plummeted the cost per base of DNA sequencing and analysis. The tools of genomics have resolved the etiology of disease for previously undiagnosable conditions, identified cancer driver gene variants, and have impacted the understanding of pathophysiology for many diseases. However, this expansion of use has also highlighted research's current voids in knowledge. The lack of precise animal models for gene-to-function association, lack of tools for analysis of genomic structural changes, skew in populations used for genetic studies, publication biases, and the "Unknown Proteome" all contribute to voids needing filled for genomics to work in a fast-paced clinical setting. The future will hold the tools to fill in these voids, with new data sets and the continual development of new technologies allowing for expansion of genomic medicine, ushering in the days to come for precision medicine. In this review we highlight these and other points in hopes of advancing and guiding precision medicine into the future for optimal success.
Collapse
Affiliation(s)
- Jeremy W Prokop
- HudsonAlpha Institute for Biotechnology , Huntsville, Alabama
- Department of Pediatrics and Human Development, Michigan State University , East Lansing, Michigan
- Department of Pharmacology and Toxicology, Michigan State University , East Lansing, Michigan
| | - Thomas May
- HudsonAlpha Institute for Biotechnology , Huntsville, Alabama
- Institute for Health and Aging, University of California San Francisco , San Francisco, California
- Elson S. Floyd College of Medicine, Washington State University , Spokane, Washington
| | - Kim Strong
- HudsonAlpha Institute for Biotechnology , Huntsville, Alabama
| | - Stephanie M Bilinovich
- Department of Pediatrics and Human Development, Michigan State University , East Lansing, Michigan
| | - Caleb Bupp
- Department of Pediatrics and Human Development, Michigan State University , East Lansing, Michigan
- Department of Genetics, Helen DeVos Children's Hospital, Spectrum Health, Grand Rapids, Michigan
| | - Surender Rajasekaran
- Department of Pediatrics and Human Development, Michigan State University , East Lansing, Michigan
- Department of Pediatric Critical Care Medicine, Helen DeVos Children's Hospital, Spectrum Health, Grand Rapids, Michigan
| | | | - Jozef Lazar
- HudsonAlpha Institute for Biotechnology , Huntsville, Alabama
| |
Collapse
|
536
|
Feng Q, Wei WQ, Chung CP, Levinson RT, Sundermann AC, Mosley JD, Bastarache L, Ferguson JF, Cox NJ, Roden DM, Denny JC, Linton MF, Edwards DRV, Stein CM. Relationship between very low low-density lipoprotein cholesterol concentrations not due to statin therapy and risk of type 2 diabetes: A US-based cross-sectional observational study using electronic health records. PLoS Med 2018; 15:e1002642. [PMID: 30153257 PMCID: PMC6112635 DOI: 10.1371/journal.pmed.1002642] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 07/25/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Observations from statin clinical trials and from Mendelian randomization studies suggest that low low-density lipoprotein cholesterol (LDL-C) concentrations may be associated with increased risk of type 2 diabetes mellitus (T2DM). Despite the findings from statin clinical trials and genetic studies, there is little direct evidence implicating low LDL-C concentrations in increased risk of T2DM. METHODS AND FINDINGS We used de-identified electronic health records (EHRs) at Vanderbilt University Medical Center to compare the risk of T2DM in a cross-sectional study among individuals with very low (≤60 mg/dl, N = 8,943) and normal (90-130 mg/dl, N = 71,343) LDL-C levels calculated using the Friedewald formula. LDL-C levels associated with statin use, hospitalization, or a serum albumin level < 3 g/dl were excluded. We used a 2-phase approach: in 1/3 of the sample (discovery) we used T2DM phenome-wide association study codes (phecodes) to identify cases and controls, and in the remaining 2/3 (validation) we identified T2DM cases and controls using a validated algorithm. The analysis plan for the validation phase was constructed at the time of the design of that component of the study. The prevalence of T2DM in the very low and normal LDL-C groups was compared using logistic regression with adjustment for age, race, sex, body mass index (BMI), high-density lipoprotein cholesterol, triglycerides, and duration of care. Secondary analyses included prespecified stratification by sex, race, BMI, and LDL-C level. In the discovery cohort, phecodes related to T2DM were significantly more frequent in the very low LDL-C group. In the validation cohort (N = 33,039 after applying the T2DM algorithm to identify cases and controls), the risk of T2DM was increased in the very low compared to normal LDL-C group (odds ratio [OR] 2.06, 95% CI 1.80-2.37; P < 2 × 10-16). The findings remained significant in sensitivity analyses. The association between low LDL-C levels and T2DM was significant in males (OR 2.43, 95% CI 2.00-2.95; P < 2 × 10-16) and females (OR 1.74, 95% CI 1.42-2.12; P = 6.88 × 10-8); in normal weight (OR 2.18, 95% CI 1.59-2.98; P = 1.1× 10-6), overweight (OR 2.17, 95% CI 1.65-2.83; P = 1.73× 10-8), and obese (OR 2.00, 95% CI 1.65-2.41; P = 8 × 10-13) categories; and in individuals with LDL-C < 40 mg/dl (OR 2.31, 95% CI 1.71-3.10; P = 3.01× 10-8) and LDL-C 40-60 mg/dl (OR 1.99, 95% CI 1.71-2.32; P < 2.0× 10-16). The association was significant in individuals of European ancestry (OR 2.67, 95% CI 2.25-3.17; P < 2 × 10-16) but not in those of African ancestry (OR 1.09, 95% CI 0.81-1.46; P = 0.56). A limitation was that we only compared groups with very low and normal LDL-C levels; also, since this was not an inception cohort, we cannot exclude the possibility of reverse causation. CONCLUSIONS Very low LDL-C concentrations occurring in the absence of statin treatment were significantly associated with T2DM risk in a large EHR population; this increased risk was present in both sexes and all BMI categories, and in individuals of European ancestry but not of African ancestry. Longitudinal cohort studies to assess the relationship between very low LDL-C levels not associated with lipid-lowering therapy and risk of developing T2DM will be important.
Collapse
Affiliation(s)
- QiPing Feng
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Cecilia P Chung
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America.,Division of Rheumatology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Rebecca T Levinson
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Alexandra C Sundermann
- Vanderbilt Epidemiology Center, Institute for Medicine and Public Health, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Jonathan D Mosley
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Lisa Bastarache
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Jane F Ferguson
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Nancy J Cox
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Dan M Roden
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America.,Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America.,Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Joshua C Denny
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - MacRae F Linton
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America.,Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Digna R Velez Edwards
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America.,Vanderbilt Epidemiology Center, Institute for Medicine and Public Health, Vanderbilt University, Nashville, Tennessee, United States of America
| | - C Michael Stein
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America.,Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, United States of America
| |
Collapse
|
537
|
Abstract
Biomedical data science has experienced an explosion of new data over the past decade. Abundant genetic and genomic data are increasingly available in large, diverse data sets due to the maturation of modern molecular technologies. Along with these molecular data, dense, rich phenotypic data are also available on comprehensive clinical data sets from health care provider organizations, clinical trials, population health registries, and epidemiologic studies. The methods and approaches for interrogating these large genetic/genomic and clinical data sets continue to evolve rapidly, as our understanding of the questions and challenges continue to emerge. In this review, the state-of-the-art methodologies for genetic/genomic analysis along with complex phenomics will be discussed. This field is changing and adapting to the novel data types made available, as well as technological advances in computation and machine learning. Thus, I will also discuss the future challenges in this exciting and innovative space. The promises of precision medicine rely heavily on the ability to marry complex genetic/genomic data with clinical phenotypes in meaningful ways.
Collapse
Affiliation(s)
- Marylyn D. Ritchie
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
538
|
SUSTain. PROCEEDINGS OF THE 24TH ACM SIGKDD INTERNATIONAL CONFERENCE ON KNOWLEDGE DISCOVERY & DATA MINING 2018; 2018:2080-2089. [PMID: 33680534 PMCID: PMC7935718 DOI: 10.1145/3219819.3219999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
This paper presents a new method, which we call SUSTain, that extends real-valued matrix and tensor factorizations to data where values are integers. Such data are common when the values correspond to event counts or ordinal measures. The conventional approach is to treat integer data as real, and then apply real-valued factorizations. However, doing so fails to preserve important characteristics of the original data, thereby making it hard to interpret the results. Instead, our approach extracts factor values from integer datasets as scores that are constrained to take values from a small integer set. These scores are easy to interpret: a score of zero indicates no feature contribution and higher scores indicate distinct levels of feature importance. At its core, SUSTain relies on: a) a problem partitioning into integer-constrained subproblems, so that they can be optimally solved in an efficient manner; and b) organizing the order of the subproblems’ solution, to promote reuse of shared intermediate results. We propose two variants, SUSTainM and SUSTainT, to handle both matrix and tensor inputs, respectively. We evaluate SUSTain against several state-of-the-art baselines on both synthetic and real Electronic Health Record (EHR) datasets. Comparing to those baselines, SUSTain shows either significantly better fit or orders of magnitude speedups that achieve a comparable fit (up to 425× faster). We apply SUSTain to EHR datasets to extract patient phenotypes (i.e., clinically meaningful patient clusters). Furthermore, 87% of them were validated as clinically meaningful phenotypes related to heart failure by a cardiologist.
Collapse
|
539
|
Li X, Meng X, Spiliopoulou A, Timofeeva M, Wei WQ, Gifford A, Shen X, He Y, Varley T, McKeigue P, Tzoulaki I, Wright AF, Joshi P, Denny JC, Campbell H, Theodoratou E. MR-PheWAS: exploring the causal effect of SUA level on multiple disease outcomes by using genetic instruments in UK Biobank. Ann Rheum Dis 2018; 77:1039-1047. [PMID: 29437585 PMCID: PMC6029646 DOI: 10.1136/annrheumdis-2017-212534] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/12/2018] [Accepted: 01/21/2018] [Indexed: 02/05/2023]
Abstract
OBJECTIVES We aimed to investigate the role of serum uric acid (SUA) level in a broad spectrum of disease outcomes using data for 120 091 individuals from UK Biobank. METHODS We performed a phenome-wide association study (PheWAS) to identify disease outcomes associated with SUA genetic risk loci. We then implemented conventional Mendelianrandomisation (MR) analysis to investigate the causal relevance between SUA level and disease outcomes identified from PheWAS. We next applied MR Egger analysis to detect and account for potential pleiotropy, which conventional MR analysis might mistake for causality, and used the HEIDI (heterogeneity in dependent instruments) test to remove cross-phenotype associations that were likely due to genetic linkage. RESULTS Our PheWAS identified 25 disease groups/outcomes associated with SUA genetic risk loci after multiple testing correction (P<8.57e-05). Our conventional MR analysis implicated a causal role of SUA level in three disease groups: inflammatory polyarthropathies (OR=1.22, 95% CI 1.11 to 1.34), hypertensive disease (OR=1.08, 95% CI 1.03 to 1.14) and disorders of metabolism (OR=1.07, 95% CI 1.01 to 1.14); and four disease outcomes: gout (OR=4.88, 95% CI 3.91 to 6.09), essential hypertension (OR=1.08, 95% CI 1.03 to 1.14), myocardial infarction (OR=1.16, 95% CI 1.03 to 1.30) and coeliac disease (OR=1.41, 95% CI 1.05 to 1.89). After balancing pleiotropic effects in MR Egger analysis, only gout and its encompassing disease group of inflammatory polyarthropathies were considered to be causally associated with SUA level. Our analysis highlighted a locus (ATXN2/S2HB3) that may influence SUA level and multiple cardiovascular and autoimmune diseases via pleiotropy. CONCLUSIONS Elevated SUA level is convincing to cause gout and inflammatory polyarthropathies, and might act as a marker for the wider range of diseases with which it associates. Our findings support further investigation on the clinical relevance of SUA level with cardiovascular, metabolic, autoimmune and respiratory diseases.
Collapse
Affiliation(s)
- Xue Li
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Xiangrui Meng
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Athina Spiliopoulou
- Centre for Population Health Sciences, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Maria Timofeeva
- Colon Cancer Genetics Group, Medical Research Council Human Genetics Unit, Medical Research Council, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Aliya Gifford
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Xia Shen
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Yazhou He
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
- West China School of Medicine, West China Hospital, Sichuan University, Sichuan, China
| | - Tim Varley
- Public Health and Intelligence, NHS National Services Scotland, Edinburgh, UK
| | - Paul McKeigue
- Centre for Population Health Sciences, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Ioanna Tzoulaki
- Department Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- MRC-PHE Centre for Environment, School of Public Health, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Alan F Wright
- Medical Research Council Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Peter Joshi
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Joshua C Denny
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Evropi Theodoratou
- Centre for Global Health Research, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
- Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
540
|
Discovering hidden knowledge through auditing clinical diagnostic knowledge bases. J Biomed Inform 2018; 84:75-81. [PMID: 29940263 DOI: 10.1016/j.jbi.2018.06.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 06/19/2018] [Accepted: 06/21/2018] [Indexed: 11/21/2022]
Abstract
OBJECTIVE Evaluate potential for data mining auditing techniques to identify hidden concepts in diagnostic knowledge bases (KB). Improving completeness enhances KB applications such as differential diagnosis and patient case simulation. MATERIALS AND METHODS Authors used unsupervised (Pearson's correlation - PC, Kendall's correlation - KC, and a heuristic algorithm - HA) methods to identify existing and discover new finding-finding interrelationships ("properties") in the INTERNIST-1/QMR KB. Authors estimated KB maintenance efficiency gains (effort reduction) of the approaches. RESULTS The methods discovered new properties at 95% CI rates of [0.1%, 5.4%] (PC), [2.8%, 12.5%] (KC), and [5.6%, 18.8%] (HA). Estimated manual effort reduction for HA-assisted determination of new properties was approximately 50-fold. CONCLUSION Data mining can provide an efficient supplement to ensuring the completeness of finding-finding interdependencies in diagnostic knowledge bases. Authors' findings should be applicable to other diagnostic systems that record finding frequencies within diseases (e.g., DXplain, ISABEL).
Collapse
|
541
|
Alexander MR, Norlander AE, Elijovich F, Atreya RV, Gaye A, Gnecco JS, Laffer CL, Galindo CL, Madhur MS. Human monocyte transcriptional profiling identifies IL-18 receptor accessory protein and lactoferrin as novel immune targets in hypertension. Br J Pharmacol 2018; 176:2015-2027. [PMID: 29774543 DOI: 10.1111/bph.14364] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 03/30/2018] [Accepted: 04/30/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Monocytes play a critical role in hypertension. The purpose of our study was to use an unbiased approach to determine whether hypertensive individuals on conventional therapy exhibit an altered monocyte gene expression profile and to perform validation studies of selected genes to identify novel therapeutic targets for hypertension. EXPERIMENTAL APPROACH Next generation RNA sequencing identified differentially expressed genes in a small discovery cohort of normotensive and hypertensive individuals. Several of these genes were further investigated for association with hypertension in multiple validation cohorts using qRT-PCR, regression analysis, phenome-wide association study and case-control analysis of a missense polymorphism. KEY RESULTS We identified 60 genes that were significantly differentially expressed in hypertensive monocytes, many of which are related to IL-1β. Uni- and multivariate regression analyses of the expression of these genes with mean arterial pressure (MAP) revealed four genes that significantly correlated with MAP in normotensive and/or hypertensive individuals. Of these, lactoferrin (LTF), peptidoglycan recognition protein 1 and IL-18 receptor accessory protein (IL18RAP) remained significantly elevated in peripheral monocytes of hypertensive individuals in a separate validation cohort. Interestingly, IL18RAP expression associated with MAP in a cohort of African Americans. Furthermore, homozygosity for a missense single nucleotide polymorphism in LTF that decreases antimicrobial function and increases protein levels (rs1126478) was over-represented in patients with hypertension relative to controls (odds ratio 1.16). CONCLUSIONS AND IMPLICATIONS These data demonstrate that monocytes exhibit enhanced pro-inflammatory gene expression in hypertensive individuals and identify IL18RAP and LTF as potential novel mediators of human hypertension. LINKED ARTICLES This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc.
Collapse
Affiliation(s)
- Matthew R Alexander
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Allison E Norlander
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Fernando Elijovich
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ravi V Atreya
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Amadou Gaye
- Metabolic, Cardiovascular and Inflammatory Disease Genomics Branch, National Human Genome Research Institute, Bethesda, MD, USA
| | - Juan S Gnecco
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN, USA
| | - Cheryl L Laffer
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cristi L Galindo
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Meena S Madhur
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.,Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
542
|
Mosley JD, Levinson RT, Brittain EL, Gupta DK, Farber-Eger E, Shaffer CM, Denny JC, Roden DM, Wells QS. Clinical Features Associated With Nascent Left Ventricular Diastolic Dysfunction in a Population Aged 40 to 55 Years. Am J Cardiol 2018; 121:1552-1557. [PMID: 29627106 PMCID: PMC5975107 DOI: 10.1016/j.amjcard.2018.02.042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/15/2018] [Accepted: 02/26/2018] [Indexed: 11/26/2022]
Abstract
Diastolic dysfunction (DD), an abnormality in cardiac left ventricular (LV) chamber compliance, is associated with increased morbidity and mortality. Although DD has been extensively studied in older populations, co-morbidity patterns are less well characterized in middle-aged subjects. We screened 156,434 subjects with transthoracic echocardiogram reports available through Vanderbilt's electronic heath record and identified 6,612 subjects 40 to 55 years old with an LV ejection fraction ≥50% and diastolic function staging. We tested 452 incident and prevalent clinical diagnoses for associations with early-stage DD (n = 1,676) versus normal function. There were 44 co-morbid diagnoses associated with grade 1 DD including hypertension (odds ratio [OR] = 2.02, 95% confidence interval [CI] 1.78 to 2.28, p <5.3 × 10-29), type 2 diabetes (OR 1.96, 95% CI 1.68 to 2.29, p = 2.1 × 10-17), tachycardia (OR 1.38, 95% CI 0.53 to 2.19, p = 2.9 × 10-6), obesity (OR 1.76, 95% CI 1.51 to 2.06, p = 1.7 × 10-12), and clinical end points, including end-stage renal disease (OR 3.29, 95% CI 2.19 to 4.96, p = 1.2 × 10-8) and stroke (OR 1.5, 95% CI 1.12 to 2.02, p = 6.9 × 10-3). Among the 60 incident diagnoses associated with DD, heart failure with preserved ejection fraction (OR 4.63, 95% CI 3.39 to 6.32, p = 6.3 × 10-22) had the most significant association. Among subjects with normal diastolic function and blood pressure at baseline, a blood pressure measurement in the hypertensive range at the time of the second echocardiogram was associated with progression to stage 1 DD (p = 0.04). In conclusion, DD was common among subjects 40 to 55 years old and was associated with a heavy burden of co-morbid disease.
Collapse
Affiliation(s)
- Jonathan D Mosley
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee.
| | - Rebecca T Levinson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Evan L Brittain
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Deepak K Gupta
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Eric Farber-Eger
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Christian M Shaffer
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Josh C Denny
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Dan M Roden
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Pharmacology, Vanderbilt University, Nashville, Tennessee
| | - Quinn S Wells
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Pharmacology, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
543
|
Gusarova V, O'Dushlaine C, Teslovich TM, Benotti PN, Mirshahi T, Gottesman O, Van Hout CV, Murray MF, Mahajan A, Nielsen JB, Fritsche L, Wulff AB, Gudbjartsson DF, Sjögren M, Emdin CA, Scott RA, Lee WJ, Small A, Kwee LC, Dwivedi OP, Prasad RB, Bruse S, Lopez AE, Penn J, Marcketta A, Leader JB, Still CD, Kirchner HL, Mirshahi UL, Wardeh AH, Hartle CM, Habegger L, Fetterolf SN, Tusie-Luna T, Morris AP, Holm H, Steinthorsdottir V, Sulem P, Thorsteinsdottir U, Rotter JI, Chuang LM, Damrauer S, Birtwell D, Brummett CM, Khera AV, Natarajan P, Orho-Melander M, Flannick J, Lotta LA, Willer CJ, Holmen OL, Ritchie MD, Ledbetter DH, Murphy AJ, Borecki IB, Reid JG, Overton JD, Hansson O, Groop L, Shah SH, Kraus WE, Rader DJ, Chen YDI, Hveem K, Wareham NJ, Kathiresan S, Melander O, Stefansson K, Nordestgaard BG, Tybjærg-Hansen A, Abecasis GR, Altshuler D, Florez JC, Boehnke M, McCarthy MI, Yancopoulos GD, Carey DJ, Shuldiner AR, Baras A, Dewey FE, Gromada J. Genetic inactivation of ANGPTL4 improves glucose homeostasis and is associated with reduced risk of diabetes. Nat Commun 2018; 9:2252. [PMID: 29899519 PMCID: PMC5997992 DOI: 10.1038/s41467-018-04611-z] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/10/2018] [Indexed: 01/05/2023] Open
Abstract
Angiopoietin-like 4 (ANGPTL4) is an endogenous inhibitor of lipoprotein lipase that modulates lipid levels, coronary atherosclerosis risk, and nutrient partitioning. We hypothesize that loss of ANGPTL4 function might improve glucose homeostasis and decrease risk of type 2 diabetes (T2D). We investigate protein-altering variants in ANGPTL4 among 58,124 participants in the DiscovEHR human genetics study, with follow-up studies in 82,766 T2D cases and 498,761 controls. Carriers of p.E40K, a variant that abolishes ANGPTL4 ability to inhibit lipoprotein lipase, have lower odds of T2D (odds ratio 0.89, 95% confidence interval 0.85-0.92, p = 6.3 × 10-10), lower fasting glucose, and greater insulin sensitivity. Predicted loss-of-function variants are associated with lower odds of T2D among 32,015 cases and 84,006 controls (odds ratio 0.71, 95% confidence interval 0.49-0.99, p = 0.041). Functional studies in Angptl4-deficient mice confirm improved insulin sensitivity and glucose homeostasis. In conclusion, genetic inactivation of ANGPTL4 is associated with improved glucose homeostasis and reduced risk of T2D.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Anubha Mahajan
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Jonas B Nielsen
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, University of Michigan, Ann Arbor, 48109, MI, USA
- Department of Human Genetics, University of Michigan, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Lars Fritsche
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Anders Berg Wulff
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen, 2100, Denmark
| | | | - Marketa Sjögren
- Department of Clinical Sciences, Malmö, Lund University, Malmö, 221, Sweden
| | - Connor A Emdin
- Program in Medical and Population Genetics, Broad Institute, Cambridge, 02142, MA, USA
| | - Robert A Scott
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Wen-Jane Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, 40705, Taiwan
- Department of Social Work, Tunghai University, Taichung, 40704, Taiwan
| | - Aeron Small
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104, USA
| | - Lydia C Kwee
- Division of Cardiology, Department of Medicine; Molecular Physiology Institute, School of Medicine, Duke University, Durham, 27710, NC, USA
| | - Om Prakash Dwivedi
- Finnish Institute of Molecular Medicine (FIMM), Helsinki University, Helsinki, 00170, Finland
| | - Rashmi B Prasad
- Department of Clinical Sciences, Clinical Research Centre, Lund University, Malmö, 221, Sweden
| | - Shannon Bruse
- Regeneron Genetics Center, Tarrytown, 10591, NY, USA
| | | | - John Penn
- Regeneron Genetics Center, Tarrytown, 10591, NY, USA
| | | | | | | | | | | | | | | | | | | | - Teresa Tusie-Luna
- Instituto de Investigaciones Biomédicas, UNAM, Coyoacán, 04510, Mexico City, Mexico
- Unidad de Biología Molecular y Medicina Genómica, UNAM/INCMNSZ Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, 14080, Mexico
| | - Andrew P Morris
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
- Department of Biostatistics, University of Liverpool, Liverpool, L69 7ZX, UK
- Estonian Genome Center, University of Tartu, Tartu, 50090, Estonia
| | - Hilma Holm
- deCODE Genetics/Amgen, Inc., Reykjavik, 101, Iceland
| | | | - Patrick Sulem
- deCODE Genetics/Amgen, Inc., Reykjavik, 101, Iceland
| | | | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Departments of Pediatrics and Medicine, LABioMed at Harbor-UCLA Medical Center, Torrance, 90502, CA, USA
| | - Lee-Ming Chuang
- Division of Endocrinology & Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, 10617, Taiwan
- Institute of Preventive Medicine, School of Public Health, National Taiwan University, Taipei, 10617, Taiwan
| | - Scott Damrauer
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Department of Surgery, Corporal Michael Crescenz VA Medical Center, Philadelphia, 19104, PA, USA
| | - David Birtwell
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104, USA
| | - Chad M Brummett
- Department of Anesthesiology, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Amit V Khera
- Program in Medical and Population Genetics, Broad Institute, Cambridge, 02142, MA, USA
- Center for Human Genetic Research, Cardiovascular Research Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, 02114, MA, USA
| | - Pradeep Natarajan
- Program in Medical and Population Genetics, Broad Institute, Cambridge, 02142, MA, USA
- Center for Human Genetic Research, Cardiovascular Research Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, 02114, MA, USA
| | | | - Jason Flannick
- Program in Medical and Population Genetics, Broad Institute, Cambridge, 02142, MA, USA
- Center for Human Genetic Research, Massachusetts General Hospital, Harvard Medical School, Boston, 02114, MA, USA
| | - Luca A Lotta
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Cristen J Willer
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, University of Michigan, Ann Arbor, 48109, MI, USA
- Department of Human Genetics, University of Michigan, University of Michigan, Ann Arbor, 48109, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Oddgeir L Holmen
- HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, 7601, Norway
| | | | | | | | | | | | | | - Ola Hansson
- Finnish Institute of Molecular Medicine (FIMM), Helsinki University, Helsinki, 00170, Finland
- Department of Clinical Sciences, Clinical Research Centre, Lund University, Malmö, 221, Sweden
| | - Leif Groop
- Finnish Institute of Molecular Medicine (FIMM), Helsinki University, Helsinki, 00170, Finland
- Department of Clinical Sciences, Clinical Research Centre, Lund University, Malmö, 221, Sweden
| | - Svati H Shah
- Division of Cardiology, Department of Medicine; Molecular Physiology Institute, School of Medicine, Duke University, Durham, 27710, NC, USA
| | - William E Kraus
- Division of Cardiology, Department of Medicine; Molecular Physiology Institute, School of Medicine, Duke University, Durham, 27710, NC, USA
| | - Daniel J Rader
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 19104, USA
| | - Yii-Der I Chen
- Institute for Translational Genomics and Population Sciences, Departments of Pediatrics and Medicine, LABioMed at Harbor-UCLA Medical Center, Torrance, 90502, CA, USA
| | - Kristian Hveem
- HUNT Research Centre, Department of Public Health and General Practice, Norwegian University of Science and Technology, Levanger, 7601, Norway
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health, Norwegian University of Science and Technology, Trondheim, 7491, Norway
- Department of Medicine, Levanger Hospital, Nord-Trøndelag Health Trust, Levanger, 7601, Norway
| | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Sekar Kathiresan
- Center for Human Genetic Research, Cardiovascular Research Center and Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, 02114, MA, USA
| | - Olle Melander
- Department of Clinical Sciences, Malmö, Lund University, Malmö, 221, Sweden
| | | | - Børge G Nordestgaard
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, 2730, Denmark
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, 2730, Denmark
- The Copenhagen City Heart Study, Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, 2400, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Anne Tybjærg-Hansen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen, 2100, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, 2730, Denmark
- The Copenhagen City Heart Study, Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, 2400, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Goncalo R Abecasis
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, 48109, MI, USA
| | - David Altshuler
- Program in Medical and Population Genetics, Broad Institute, Cambridge, 02142, MA, USA
- Department of Molecular Biology, Diabetes Unit, and Center for Human Genetic Research, Massachusetts General Hospital, Boston, 02114, MA, USA
- Departments of Genetics and Medicine, Harvard Medical School, Boston, 02115, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA
| | - Jose C Florez
- Diabetes Unit and Center for Human Genetic Research, Massachusetts General Hospital, Boston, 02115, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute, Cambridge, 02142, MA, USA
- Department of Medicine, Harvard Medical School, Boston, 02115, MA, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, 48109, MI, USA
| | - Mark I McCarthy
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, OX3 7LE, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, OX4 2PG, UK
| | | | | | | | - Aris Baras
- Regeneron Genetics Center, Tarrytown, 10591, NY, USA.
| | | | | |
Collapse
|
544
|
Uneven Distribution of Mutational Variance Across the Transcriptome of Drosophila serrata Revealed by High-Dimensional Analysis of Gene Expression. Genetics 2018; 209:1319-1328. [PMID: 29884746 DOI: 10.1534/genetics.118.300757] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/31/2018] [Indexed: 11/18/2022] Open
Abstract
There are essentially an infinite number of traits that could be measured on any organism, and almost all individual traits display genetic variation, yet substantial genetic variance in a large number of independent traits is not plausible under basic models of selection and mutation. One mechanism that may be invoked to explain the observed levels of genetic variance in individual traits is that pleiotropy results in fewer dimensions of phenotypic space with substantial genetic variance. Multivariate genetic analyses of small sets of functionally related traits have shown that standing genetic variance is often concentrated in relatively few dimensions. It is unknown if a similar concentration of genetic variance occurs at a phenome-wide scale when many traits of disparate function are considered, or if the genetic variance generated by new mutations is also unevenly distributed across phenotypic space. Here, we used a Bayesian sparse factor model to characterize the distribution of mutational variance of 3385 gene expression traits of Drosophila serrata after 27 generations of mutation accumulation, and found that 46% of the estimated mutational variance was concentrated in just 21 dimensions with significant mutational heritability. We show that the extent of concentration of mutational variance into such a small subspace has the potential to substantially bias the response to selection of these traits.
Collapse
|
545
|
Fritsche LG, Gruber SB, Wu Z, Schmidt EM, Zawistowski M, Moser SE, Blanc VM, Brummett CM, Kheterpal S, Abecasis GR, Mukherjee B. Association of Polygenic Risk Scores for Multiple Cancers in a Phenome-wide Study: Results from The Michigan Genomics Initiative. Am J Hum Genet 2018; 102:1048-1061. [PMID: 29779563 DOI: 10.1016/j.ajhg.2018.04.001] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 03/26/2018] [Indexed: 12/11/2022] Open
Abstract
Health systems are stewards of patient electronic health record (EHR) data with extraordinarily rich depth and breadth, reflecting thousands of diagnoses and exposures. Measures of genomic variation integrated with EHRs offer a potential strategy to accurately stratify patients for risk profiling and discover new relationships between diagnoses and genomes. The objective of this study was to evaluate whether polygenic risk scores (PRS) for common cancers are associated with multiple phenotypes in a phenome-wide association study (PheWAS) conducted in 28,260 unrelated, genotyped patients of recent European ancestry who consented to participate in the Michigan Genomics Initiative, a longitudinal biorepository effort within Michigan Medicine. PRS for 12 cancer traits were calculated using summary statistics from the NHGRI-EBI catalog. A total of 1,711 synthetic case-control studies was used for PheWAS analyses. There were 13,490 (47.7%) patients with at least one cancer diagnosis in this study sample. PRS exhibited strong association for several cancer traits they were designed for, including female breast cancer, prostate cancer, melanoma, basal cell carcinoma, squamous cell carcinoma, and thyroid cancer. Phenome-wide significant associations were observed between PRS and many non-cancer diagnoses. To differentiate PRS associations driven by the primary trait from associations arising through shared genetic risk profiles, the idea of "exclusion PRS PheWAS" was introduced. Further analysis of temporal order of the diagnoses improved our understanding of these secondary associations. This comprehensive PheWAS used PRS instead of a single variant.
Collapse
Affiliation(s)
- Lars G Fritsche
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, 7491 Trondheim, Sør-Trøndelag, Norway
| | - Stephen B Gruber
- USC Norris Comprehensive Cancer Center, Los Angeles, CA 90033, USA
| | - Zhenke Wu
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; Michigan Institute for Data Science, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ellen M Schmidt
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Matthew Zawistowski
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Stephanie E Moser
- Division of Pain Medicine, Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Victoria M Blanc
- Central Biorepository, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Chad M Brummett
- Division of Pain Medicine, Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sachin Kheterpal
- Division of Pain Medicine, Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor, MI 48109, USA
| | - Gonçalo R Abecasis
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Bhramar Mukherjee
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; Michigan Institute for Data Science, University of Michigan, Ann Arbor, MI 48109, USA; Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
546
|
Dahir KM, Tilden DR, Warner JL, Bastarache L, Smith DK, Gifford A, Ramirez AH, Simmons JS, Black MM, Newman JH, Denny JC. Rare Variants in the Gene ALPL That Cause Hypophosphatasia Are Strongly Associated With Ovarian and Uterine Disorders. J Clin Endocrinol Metab 2018; 103:2234-2243. [PMID: 29659871 PMCID: PMC6456921 DOI: 10.1210/jc.2017-02676] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 04/02/2018] [Indexed: 11/19/2022]
Abstract
Context Mutations in alkaline phosphatase (AlkP), liver/bone/kidney (ALPL), which encodes tissue-nonspecific isozyme AlkP, cause hypophosphatasia (HPP). HPP is suspected by a low-serum AlkP. We hypothesized that some patients with bone or dental disease have undiagnosed HPP, caused by ALPL variants. Objective Our objective was to discover the prevalence of these gene variants in the Vanderbilt University DNA Biobank (BioVU) and to assess phenotypic associations. Design We identified subjects in BioVU, a repository of DNA, that had at least one of three known, rare HPP disease-causing variants in ALPL: rs199669988, rs121918007, and/or rs121918002. To evaluate for phenotypic associations, we conducted a sequential phenome-wide association study of ALPL variants and then performed a de-identified manual record review to refine the phenotype. Results Out of 25,822 genotyped individuals, we identified 52 women and 53 men with HPP disease-causing variants in ALPL, 7/1000. None had a clinical diagnosis of HPP. For patients with ALPL variants, the average serum AlkP levels were in the lower range of normal or lower. Forty percent of men and 62% of women had documented bone and/or dental disease, compatible with the diagnosis of HPP. Forty percent of the female patients had ovarian pathology or other gynecological abnormalities compared with 15% seen in controls. Conclusions Variants in the ALPL gene cause bone and dental disease in patients with and without the standard biomarker, low plasma AlkP. ALPL gene variants are more prevalent than currently reported and underdiagnosed. Gynecologic disease appears to be associated with HPP-causing variants in ALPL.
Collapse
Affiliation(s)
- Kathryn M Dahir
- Division of Endocrinology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Daniel R Tilden
- Department of Internal Medicine and Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jeremy L Warner
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lisa Bastarache
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Derek K Smith
- Departments of Biostatistics and Oral Maxillofacial Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Aliya Gifford
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Andrea H Ramirez
- Division of Endocrinology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jill S Simmons
- Division of Pediatric Endocrinology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Margo M Black
- Division of Pediatric Endocrinology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - John H Newman
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Josh C Denny
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee
- Division of General Internal Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
547
|
Hemani G, Zheng J, Elsworth B, Wade KH, Haberland V, Baird D, Laurin C, Burgess S, Bowden J, Langdon R, Tan VY, Yarmolinsky J, Shihab HA, Timpson NJ, Evans DM, Relton C, Martin RM, Davey Smith G, Gaunt TR, Haycock PC. The MR-Base platform supports systematic causal inference across the human phenome. eLife 2018. [PMID: 29846171 DOI: 10.7554/elife.34408s] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
Results from genome-wide association studies (GWAS) can be used to infer causal relationships between phenotypes, using a strategy known as 2-sample Mendelian randomization (2SMR) and bypassing the need for individual-level data. However, 2SMR methods are evolving rapidly and GWAS results are often insufficiently curated, undermining efficient implementation of the approach. We therefore developed MR-Base (<ext-link ext-link-type="uri" xlink:href="http://www.mrbase.org">http://www.mrbase.org</ext-link>): a platform that integrates a curated database of complete GWAS results (no restrictions according to statistical significance) with an application programming interface, web app and R packages that automate 2SMR. The software includes several sensitivity analyses for assessing the impact of horizontal pleiotropy and other violations of assumptions. The database currently comprises 11 billion single nucleotide polymorphism-trait associations from 1673 GWAS and is updated on a regular basis. Integrating data with software ensures more rigorous application of hypothesis-driven analyses and allows millions of potential causal relationships to be efficiently evaluated in phenome-wide association studies.
Collapse
Affiliation(s)
- Gibran Hemani
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Jie Zheng
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Benjamin Elsworth
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Kaitlin H Wade
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Valeriia Haberland
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Denis Baird
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Charles Laurin
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Stephen Burgess
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Jack Bowden
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Ryan Langdon
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Vanessa Y Tan
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - James Yarmolinsky
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Hashem A Shihab
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Nicholas J Timpson
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - David M Evans
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom.,University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia
| | - Caroline Relton
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Richard M Martin
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - George Davey Smith
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Tom R Gaunt
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Philip C Haycock
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
548
|
Hemani G, Zheng J, Elsworth B, Wade KH, Haberland V, Baird D, Laurin C, Burgess S, Bowden J, Langdon R, Tan VY, Yarmolinsky J, Shihab HA, Timpson NJ, Evans DM, Relton C, Martin RM, Davey Smith G, Gaunt TR, Haycock PC. The MR-Base platform supports systematic causal inference across the human phenome. eLife 2018; 7:e34408. [PMID: 29846171 PMCID: PMC5976434 DOI: 10.7554/elife.34408] [Citation(s) in RCA: 4609] [Impact Index Per Article: 658.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 03/28/2018] [Indexed: 12/21/2022] Open
Abstract
Results from genome-wide association studies (GWAS) can be used to infer causal relationships between phenotypes, using a strategy known as 2-sample Mendelian randomization (2SMR) and bypassing the need for individual-level data. However, 2SMR methods are evolving rapidly and GWAS results are often insufficiently curated, undermining efficient implementation of the approach. We therefore developed MR-Base (http://www.mrbase.org): a platform that integrates a curated database of complete GWAS results (no restrictions according to statistical significance) with an application programming interface, web app and R packages that automate 2SMR. The software includes several sensitivity analyses for assessing the impact of horizontal pleiotropy and other violations of assumptions. The database currently comprises 11 billion single nucleotide polymorphism-trait associations from 1673 GWAS and is updated on a regular basis. Integrating data with software ensures more rigorous application of hypothesis-driven analyses and allows millions of potential causal relationships to be efficiently evaluated in phenome-wide association studies.
Collapse
Affiliation(s)
- Gibran Hemani
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | - Jie Zheng
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | - Benjamin Elsworth
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | - Kaitlin H Wade
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | - Valeriia Haberland
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | - Denis Baird
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | - Charles Laurin
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | - Stephen Burgess
- Department of Public Health and Primary CareUniversity of CambridgeCambridgeUnited Kingdom
| | - Jack Bowden
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | - Ryan Langdon
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | - Vanessa Y Tan
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | - James Yarmolinsky
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | - Hashem A Shihab
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | - Nicholas J Timpson
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | - David M Evans
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
- University of Queensland Diamantina InstituteTranslational Research InstituteBrisbaneAustralia
| | - Caroline Relton
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | - Richard M Martin
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | - George Davey Smith
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | - Tom R Gaunt
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | - Philip C Haycock
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| |
Collapse
|
549
|
|
550
|
Bloodworth MH, Rusznak M, Bastarache L, Wang J, Denny JC, Peebles RS. Association of ST2 polymorphisms with atopy, asthma, and leukemia. J Allergy Clin Immunol 2018; 142:991-993.e3. [PMID: 29787780 DOI: 10.1016/j.jaci.2018.03.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 02/01/2018] [Accepted: 03/19/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Melissa H Bloodworth
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Mark Rusznak
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Lisa Bastarache
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Janey Wang
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tenn
| | - Joshua C Denny
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, Tenn
| | - R Stokes Peebles
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, Tenn; Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tenn.
| |
Collapse
|