1
|
Sriram V, Woerner J, Ahn YY, Kim D. The interplay of sex and genotype in disease associations: a comprehensive network analysis in the UK Biobank. Hum Genomics 2025; 19:4. [PMID: 39825454 PMCID: PMC11740496 DOI: 10.1186/s40246-024-00710-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/17/2024] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Disease comorbidities and longer-term complications, arising from biologically related associations across phenotypes, can lead to increased risk of severe health outcomes. Given that many diseases exhibit sex-specific differences in their genetics, our objective was to determine whether genotype-by-sex (GxS) interactions similarly influence cross-phenotype associations. Through comparison of sex-stratified disease-disease networks (DDNs)-where nodes represent diseases and edges represent their relationships-we investigate sex differences in patterns of polygenicity and pleiotropy between diseases. RESULTS Using UK Biobank summary statistics, we built male- and female-specific DDNs for 103 diseases. This revealed that male and female diseasomes have similar topology and central diseases (e.g., hypertensive, chronic respiratory, and thyroid-based disorders), yet some phenotypes exhibit sex-specific influence in cross-phenotype associations. Multiple sclerosis and osteoarthritis are central only in the female DDN, while cardiometabolic diseases and skin cancer are more prominent in the male DDN. Edge comparison indicated similar shared genetics between the two graphs relative to a random model of disease association, though notable discrepancies in embedding distances and clustering patterns imply a more expansive genetic influence on multimorbidity risk for females than males. Analysis of pleiotropic contributions of two sexually-dimorphic single-nucleotide polymorphisms related to thyroid disorders further validated a distinct genetic architecture across sexes that influences associations, confirmed through examination of corresponding gene expression profiles from the GTEx Portal. CONCLUSIONS Our analysis affirms the presence of GxS interactions in cross-phenotype associations, emphasizing the need to investigate the role of sex in disease onset and its importance in biomedical discovery and precision medicine research.
Collapse
Affiliation(s)
- Vivek Sriram
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Richards Building B304, 3700 Hamilton Walk, Philadelphia, PA, 19104, USA
| | - Jakob Woerner
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Richards Building B304, 3700 Hamilton Walk, Philadelphia, PA, 19104, USA
| | - Yong-Yeol Ahn
- Center for Complex Networks and Systems Research, Luddy School of Informatics, Computing, and Engineering, Indiana University Bloomington, Bloomington, IN, 47405, USA.
| | - Dokyoon Kim
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Richards Building B304, 3700 Hamilton Walk, Philadelphia, PA, 19104, USA.
- Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
2
|
Hall M, Skinderhaug MK, Almaas E. Phenome-wide association network demonstrates close connection with individual disease trajectories from the HUNT study. PLoS One 2024; 19:e0311485. [PMID: 39729424 DOI: 10.1371/journal.pone.0311485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/16/2024] [Indexed: 12/29/2024] Open
Abstract
Disease networks offer a potential road map of connections between diseases. Several studies have created disease networks where diseases are connected either based on shared genes or Single Nucleotide Polymorphism (SNP) associations. However, it is still unclear to which degree SNP-based networks map to empirical, co-observed diseases within a different, general, adult study population spanning over a long time period. We created a SNP-based phenome-wide association network (PheNet) from a large population using the UK biobank phenome-wide association studies. Importantly, the SNP-associations are unbiased towards much studied diseases, adjusted for linkage disequilibrium, case/control imbalances, as well as relatedness. We map the PheNet to significantly co-occurring diseases in the Norwegian HUNT study population, and further, identify consecutively occurring diseases with significant ordering in occurrence, independent of age and gender in the PheNet. Our analysis reveals an overlap far larger than expected by chance between the two disease networks, with diseases typically connecting within their own category. Upon examining the sequential occurrence of diseases in the HUNT dataset, we find a giant component consisting of mostly cardiovascular disorders. This allows us to identify sequentially occurring diseases that are genetically linked and co-occur frequently, while also highlighting non-sequential diseases. Furthermore, we observe that survivors of severe cardiovascular diseases subsequently often face less severe conditions, but with a reduced time until their next fatal illness. The HUNT sub-PheNet showing both genetically and co-observed diseases offers an interesting framework to study groups of diseases and examine if they, in fact, are comorbidities. We find that the HUNT sub-PheNet offers the possibility to pinpoint exactly which mutation(s) constitute shared cause of the diseases. This could be of great benefit to both researchers and clinicians studying relationships between diseases.
Collapse
Affiliation(s)
- Martina Hall
- Department of Biotechnology and Food Science, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Marit K Skinderhaug
- Department of Biotechnology and Food Science, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Eivind Almaas
- Department of Biotechnology and Food Science, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- K. G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
3
|
Miyamoto H, Kondo Y, Itobayashi E, Uehara M, Hiraoka A, Kudo M, Kakizaki S, Kagawa T, Miuma S, Suzuki T, Sugi K, Suyama K, Beppu T, Toyoda H, Yoshiji H, Uojima H, Miyase S, Inoue K, Tamori A, Ito T, Shimose S, Suda G, Hayashi T, Onishi M, Narahara S, Watanabe T, Iwatsuki M, Fukushima S, Tanaka Y. Evaluation of the associations of interlukin-7 genetic variants with toxicity and efficacy of immune checkpoint inhibitors: A replication study of a Japanese population, based on the findings of a European genome-wide association study. Hepatol Res 2024; 54:1215-1225. [PMID: 38990762 DOI: 10.1111/hepr.14092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/05/2024] [Accepted: 06/24/2024] [Indexed: 07/13/2024]
Abstract
AIM Recent genome-wide association studies of European populations have identified rs16906115, a single-nucleotide polymorphism in the interleukin-7 gene, as a predictor of immune-related adverse events (irAEs) and the therapeutic efficacy of immune checkpoint inhibitors. We evaluated this single-nucleotide polymorphism in a Japanese population. METHODS From January 2021, we stored host DNA from individuals who received various types of immune checkpoint inhibitors. From this population, we categorized 510 participants into cases (grade ≥2 irAEs) and controls (received ≥3 immune checkpoint inhibitor doses, follow-up ≥12 weeks, no irAEs), and divided 339 hepatocellular carcinoma patients treated with atezolizumab/bevacizumab into responders and non-responders, evaluated using the modified response evaluation criteria in solid tumors. We compared the minor allele frequencies of rs16906115 between cases and controls, and responders and non-responders. RESULTS In the irAE prediction analysis of 234 cases and 276 controls, the minor allele frequency was 0.244 in the case group and 0.265 in the control group. This difference is not significant. In the analysis predicting the therapeutic efficacy for hepatocellular carcinoma patients, the responders had a significantly lower minor allele frequency of 0.220, compared with 0.300 for the non-responders (p = 0.022). Univariate and multivariate analyses identified the minor allele homozygosity as a significant predictor of treatment response, with odds ratios of 0.292 (p = 0.015) in the univariate analysis and 0.315 (p = 0.023) in the multivariate analysis. CONCLUSIONS In our Japanese cohort, no association was found between the rs16906115 minor allele and irAEs or treatment efficacy. The minor allele homozygosity may be associated with a negative therapeutic outcome. CLINICAL TRIAL REGISTRATION UMIN Clinical Trials Registry with the number UMIN000043798.
Collapse
Affiliation(s)
- Hideaki Miyamoto
- Department of Gastroenterology and Hepatology, Kumamoto University Hospital, Kumamoto, Japan
| | - Yasuteru Kondo
- Department of Gastroenterology and Hepatology, Kumamoto University Hospital, Kumamoto, Japan
- Department of Hepatology, Sendai Tokushukai Hospital, Miyagi, Japan
- Department of Hepatology, Sendai Kousei Hospital, Miyagi, Japan
| | - Ei Itobayashi
- Department of Gastroenterology, Asahi General Hospital, Chiba, Japan
| | - Masayoshi Uehara
- Department of Gastroenterology, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Atsushi Hiraoka
- Department of Gastroenterology, Ehime Prefectural Central Hospital, Ehime, Japan
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Hospital, Osaka, Japan
| | - Satoru Kakizaki
- Department of Clinical Research, NHO Takasaki General Medical Center, Gunma, Japan
| | - Tatehiro Kagawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Tokai University School of Medicine, Kanagawa, Japan
| | - Satoshi Miuma
- Department of Gastroenterology and Hepatology, Nagasaki University Hospital, Nagasaki, Japan
| | - Takanori Suzuki
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Science, Aichi, Japan
| | - Kazuhiro Sugi
- Department of Gastroenterology, National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Koichi Suyama
- Department of Medical Oncology, Toranomon Hospital, Tokyo, Japan
| | - Toru Beppu
- Department of Surgery, Yamaga City Medical Center, Kumamoto, Japan
| | - Hidenori Toyoda
- Department of Gastroenterology and Hepatology, Ogaki Municipal Hospital, Gifu, Japan
| | - Hitoshi Yoshiji
- Department of Gastroenterology, Nara Medical University, Nara, Japan
| | - Haruki Uojima
- Department of Gastroenterology, Kitasato University Hospital, Kanagawa, Japan
| | - Shiho Miyase
- Department of Gastroenterology and Hepatology, Kumamoto Shinto General Hospital, Kumamoto, Japan
| | - Kaori Inoue
- Department of Liver and Diabetes and Endocrinology, Saga University Hospital, Saga, Japan
| | - Akihiro Tamori
- Department of Hepatology, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Takanori Ito
- Department of Gastroenterology and Hepatology, Nagoya University Hospital, Aichi, Japan
| | - Shigeo Shimose
- Department of Gastroenterology, Kurume University Hospital, Fukuoka, Japan
| | - Goki Suda
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Hokkaido, Japan
| | - Tsuguru Hayashi
- Department of Hepatology, Sendai Kousei Hospital, Miyagi, Japan
| | - Masaya Onishi
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
- Department of Gastroenterology, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Satoshi Narahara
- Department of Gastroenterology and Hepatology, Kumamoto University Hospital, Kumamoto, Japan
| | - Takehisa Watanabe
- Department of Gastroenterology and Hepatology, Kumamoto University Hospital, Kumamoto, Japan
| | - Masaaki Iwatsuki
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoshi Fukushima
- Faculty of Life Sciences, Department of Dermatology and Plastic Surgery, Kumamoto University, Kumamoto, Japan
| | - Yasuhito Tanaka
- Department of Gastroenterology and Hepatology, Kumamoto University Hospital, Kumamoto, Japan
| |
Collapse
|
4
|
Upadhyay VR, Ramesh V, Kumar H, Somagond YM, Priyadarsini S, Kuniyal A, Prakash V, Sahoo A. Phenomics in Livestock Research: Bottlenecks and Promises of Digital Phenotyping and Other Quantification Techniques on a Global Scale. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:380-393. [PMID: 39012961 DOI: 10.1089/omi.2024.0109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Bottlenecks in moving genomics to real-life applications also include phenomics. This is true not only for genomics medicine and public health genomics but also in ecology and livestock phenomics. This expert narrative review explores the intricate relationship between genetic makeup and observable phenotypic traits across various biological levels in the context of livestock research. We unpack and emphasize the significance of precise phenotypic data in selective breeding outcomes and examine the multifaceted applications of phenomics, ranging from improvement to assessing welfare, reproductive traits, and environmental adaptation in livestock. As phenotypic traits exhibit strong correlations, their measurement alongside specific biological outcomes provides insights into performance, overall health, and clinical endpoints like morbidity and disease. In addition, automated assessment of livestock holds potential for monitoring the dynamic phenotypic traits across various species, facilitating a deeper comprehension of how they adapt to their environment and attendant stressors. A key challenge in genetic improvement in livestock is predicting individuals with optimal fitness without direct measurement. Temporal predictions from unmanned aerial systems can surpass genomic predictions, offering in-depth data on livestock. In the near future, digital phenotyping and digital biomarkers may further unravel the genetic intricacies of stress tolerance, adaptation and welfare aspects of animals enabling the selection of climate-resilient and productive livestock. This expert review thus delves into challenges associated with phenotyping and discusses technological advancements shaping the future of biological research concerning livestock.
Collapse
Affiliation(s)
| | - Vikram Ramesh
- ICAR-National Research Centre on Mithun, Medziphema, Nagaland, India
| | - Harshit Kumar
- ICAR-National Research Centre on Mithun, Medziphema, Nagaland, India
| | - Y M Somagond
- ICAR-National Research Centre on Mithun, Medziphema, Nagaland, India
| | | | - Aruna Kuniyal
- ICAR-National Research Centre on Camel, Bikaner, Rajasthan, India
| | - Ved Prakash
- ICAR-National Research Centre on Camel, Bikaner, Rajasthan, India
| | - Artabandhu Sahoo
- ICAR-National Research Centre on Camel, Bikaner, Rajasthan, India
| |
Collapse
|
5
|
Jafari E, Blackman MH, Karnes JH, Van Driest SL, Crawford DC, Choi L, McDonough CW. Using electronic health records for clinical pharmacology research: Challenges and considerations. Clin Transl Sci 2024; 17:e13871. [PMID: 38943244 PMCID: PMC11213823 DOI: 10.1111/cts.13871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 07/01/2024] Open
Abstract
Electronic health records (EHRs) contain a vast array of phenotypic data on large numbers of individuals, often collected over decades. Due to the wealth of information, EHR data have emerged as a powerful resource to make first discoveries and identify disparities in our healthcare system. While the number of EHR-based studies has exploded in recent years, most of these studies are directed at associations with disease rather than pharmacotherapeutic outcomes, such as drug response or adverse drug reactions. This is largely due to challenges specific to deriving drug-related phenotypes from the EHR. There is great potential for EHR-based discovery in clinical pharmacology research, and there is a critical need to address specific challenges related to accurate and reproducible derivation of drug-related phenotypes from the EHR. This review provides a detailed evaluation of challenges and considerations for deriving drug-related data from EHRs. We provide an examination of EHR-based computable phenotypes and discuss cutting-edge approaches to map medication information for clinical pharmacology research, including medication-based computable phenotypes and natural language processing. We also discuss additional considerations such as data structure, heterogeneity and missing data, rare phenotypes, and diversity within the EHR. By further understanding the complexities associated with conducting clinical pharmacology research using EHR-based data, investigators will be better equipped to design thoughtful studies with more reproducible results. Progress in utilizing EHRs for clinical pharmacology research should lead to significant advances in our ability to understand differential drug response and predict adverse drug reactions.
Collapse
Affiliation(s)
- Eissa Jafari
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics and Precision Medicine, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
- Department of Pharmacy Practice, College of PharmacyJazan UniversityJazanSaudi Arabia
| | - Marisa H. Blackman
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jason H. Karnes
- Department of Pharmacy Practice and ScienceUniversity of Arizona R. Ken Coit College of PharmacyTucsonArizonaUSA
| | - Sara L. Van Driest
- Department of PediatricsVanderbilt University Medical Center (VUMC)NashvilleTennesseeUSA
- Present address:
All of US Research Program, National Institutes of HealthBethesdaMarylandUSA
| | - Dana C. Crawford
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational BiologyCase Western Reserve UniversityClevelandOhioUSA
- Department of Genetics and Genome Sciences, Cleveland Institute for Computational BiologyCase Western Reserve UniversityClevelandOhioUSA
| | - Leena Choi
- Department of Biostatistics and Biomedical InformaticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Caitrin W. McDonough
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics and Precision Medicine, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
6
|
Cao X, Zhang S, Sha Q. A novel method for multiple phenotype association studies based on genotype and phenotype network. PLoS Genet 2024; 20:e1011245. [PMID: 38728360 PMCID: PMC11111089 DOI: 10.1371/journal.pgen.1011245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 05/22/2024] [Accepted: 03/29/2024] [Indexed: 05/12/2024] Open
Abstract
Joint analysis of multiple correlated phenotypes for genome-wide association studies (GWAS) can identify and interpret pleiotropic loci which are essential to understand pleiotropy in diseases and complex traits. Meanwhile, constructing a network based on associations between phenotypes and genotypes provides a new insight to analyze multiple phenotypes, which can explore whether phenotypes and genotypes might be related to each other at a higher level of cellular and organismal organization. In this paper, we first develop a bipartite signed network by linking phenotypes and genotypes into a Genotype and Phenotype Network (GPN). The GPN can be constructed by a mixture of quantitative and qualitative phenotypes and is applicable to binary phenotypes with extremely unbalanced case-control ratios in large-scale biobank datasets. We then apply a powerful community detection method to partition phenotypes into disjoint network modules based on GPN. Finally, we jointly test the association between multiple phenotypes in a network module and a single nucleotide polymorphism (SNP). Simulations and analyses of 72 complex traits in the UK Biobank show that multiple phenotype association tests based on network modules detected by GPN are much more powerful than those without considering network modules. The newly proposed GPN provides a new insight to investigate the genetic architecture among different types of phenotypes. Multiple phenotypes association studies based on GPN are improved by incorporating the genetic information into the phenotype clustering. Notably, it might broaden the understanding of genetic architecture that exists between diagnoses, genes, and pleiotropy.
Collapse
Affiliation(s)
- Xuewei Cao
- Department of Mathematical Sciences, Michigan Technological University, Houghton, Michigan, United States of America
| | - Shuanglin Zhang
- Department of Mathematical Sciences, Michigan Technological University, Houghton, Michigan, United States of America
| | - Qiuying Sha
- Department of Mathematical Sciences, Michigan Technological University, Houghton, Michigan, United States of America
| |
Collapse
|
7
|
Woerner J, Sriram V, Nam Y, Verma A, Kim D. Uncovering genetic associations in the human diseasome using an endophenotype-augmented disease network. Bioinformatics 2024; 40:btae126. [PMID: 38527901 PMCID: PMC10963079 DOI: 10.1093/bioinformatics/btae126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/17/2024] [Indexed: 03/27/2024] Open
Abstract
MOTIVATION Many diseases, particularly cardiometabolic disorders, exhibit complex multimorbidities with one another. An intuitive way to model the connections between phenotypes is with a disease-disease network (DDN), where nodes represent diseases and edges represent associations, such as shared single-nucleotide polymorphisms (SNPs), between pairs of diseases. To gain further genetic understanding of molecular contributors to disease associations, we propose a novel version of the shared-SNP DDN (ssDDN), denoted as ssDDN+, which includes connections between diseases derived from genetic correlations with intermediate endophenotypes. We hypothesize that a ssDDN+ can provide complementary information to the disease connections in a ssDDN, yielding insight into the role of clinical laboratory measurements in disease interactions. RESULTS Using PheWAS summary statistics from the UK Biobank, we constructed a ssDDN+ revealing hundreds of genetic correlations between diseases and quantitative traits. Our augmented network uncovers genetic associations across different disease categories, connects relevant cardiometabolic diseases, and highlights specific biomarkers that are associated with cross-phenotype associations. Out of the 31 clinical measurements under consideration, HDL-C connects the greatest number of diseases and is strongly associated with both type 2 diabetes and heart failure. Triglycerides, another blood lipid with known genetic causes in non-mendelian diseases, also adds a substantial number of edges to the ssDDN. This work demonstrates how association with clinical biomarkers can better explain the shared genetics between cardiometabolic disorders. Our study can facilitate future network-based investigations of cross-phenotype associations involving pleiotropy and genetic heterogeneity, potentially uncovering sources of missing heritability in multimorbidities. AVAILABILITY AND IMPLEMENTATION The generated ssDDN+ can be explored at https://hdpm.biomedinfolab.com/ddn/biomarkerDDN.
Collapse
Affiliation(s)
- Jakob Woerner
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Vivek Sriram
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Yonghyun Nam
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Anurag Verma
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Dokyoon Kim
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA 19104, United States
| |
Collapse
|
8
|
Ouyang D, Huang C, Liu H, Xie W, Chen C, Su B, Guo L. Comprehensive analysis of genetic associations and single-cell expression profiles reveals potential links between migraine and multiple diseases: a phenome-wide association study. Front Neurol 2024; 15:1301208. [PMID: 38385040 PMCID: PMC10879407 DOI: 10.3389/fneur.2024.1301208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 01/19/2024] [Indexed: 02/23/2024] Open
Abstract
Migraine is a common neurological disorder that affects more than one billion people worldwide. Recent genome-wide association studies have identified 123 genetic loci associated with migraine risk. However, the biological mechanisms underlying migraine and its relationships with other complex diseases remain unclear. We performed a phenome-wide association study (PheWAS) using UK Biobank data to investigate associations between migraine and 416 phenotypes. Mendelian randomization was employed using the IVW method. For loci associated with multiple diseases, pleiotropy was tested using MR-Egger. Single-cell RNA sequencing data was analyzed to profile the expression of 73 migraine susceptibility genes across brain cell types. qPCR was used to validate the expression of selected genes in microglia. PheWAS identified 15 disorders significantly associated with migraine, with one association detecting potential pleiotropy. Single-cell analysis revealed elevated expression of seven susceptibility genes (including ZEB2, RUNX1, SLC24A3, ANKDD1B, etc.) in brain glial cells. And qPCR confirmed the upregulation of these genes in LPS-treated microglia. This multimodal analysis provides novel insights into the link between migraine and other diseases. The single-cell profiling suggests the involvement of specific brain cells and molecular pathways. Validation of gene expression in microglia supports their potential role in migraine pathology. Overall, this study uncovers pleiotropic relationships and the biological underpinnings of migraine susceptibility.
Collapse
Affiliation(s)
- Di Ouyang
- Nanjing University of Chinese Medicine, Nanjing, China
- Traditional Chinese Medicine Hospital of Yulin, Yulin, China
| | - Chunying Huang
- Traditional Chinese Medicine Hospital of Yulin, Yulin, China
| | - Huihua Liu
- Traditional Chinese Medicine Hospital of Yulin, Yulin, China
| | | | | | - Ben Su
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lizhong Guo
- Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
9
|
Venkateswaran V, Boulier K, Ding Y, Johnson R, Bhattacharya A, Pasaniuc B. Polygenic scores for tobacco use provide insights into systemic health risks in a diverse EHR-linked biobank in Los Angeles. Transl Psychiatry 2024; 14:38. [PMID: 38238290 PMCID: PMC10796315 DOI: 10.1038/s41398-024-02743-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 12/19/2023] [Accepted: 01/08/2024] [Indexed: 01/22/2024] Open
Abstract
Tobacco use is a major risk factor for many diseases and is heavily influenced by environmental factors with significant underlying genetic contributions. Here, we evaluated the predictive performance, risk stratification, and potential systemic health effects of tobacco use disorder (TUD) predisposing germline variants using a European- ancestry-derived polygenic score (PGS) in 24,202 participants from the multi-ancestry, hospital-based UCLA ATLAS biobank. Among genetically inferred ancestry groups (GIAs), TUD-PGS was significantly associated with TUD in European American (EA) (OR: 1.20, CI: [1.16, 1.24]), Hispanic/Latin American (HL) (OR:1.19, CI: [1.11, 1.28]), and East Asian American (EAA) (OR: 1.18, CI: [1.06, 1.31]) GIAs but not in African American (AA) GIA (OR: 1.04, CI: [0.93, 1.17]). Similarly, TUD-PGS offered strong risk stratification across PGS quantiles in EA and HL GIAs and inconsistently in EAA and AA GIAs. In a cross-ancestry phenome-wide association meta-analysis, TUD-PGS was associated with cardiometabolic, respiratory, and psychiatric phecodes (17 phecodes at P < 2.7E-05). In individuals with no history of smoking, the top TUD-PGS associations with obesity and alcohol-related disorders (P = 3.54E-07, 1.61E-06) persist. Mendelian Randomization (MR) analysis provides evidence of a causal association between adiposity measures and tobacco use. Inconsistent predictive performance of the TUD-PGS across GIAs motivates the inclusion of multiple ancestry populations at all levels of genetic research of tobacco use for equitable clinical translation of TUD-PGS. Phenome associations suggest that TUD-predisposed individuals may require comprehensive tobacco use prevention and management approaches to address underlying addictive tendencies.
Collapse
Affiliation(s)
- Vidhya Venkateswaran
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Oral Biology, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Office of the Director and National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Kristin Boulier
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Medicine, Division of Cardiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yi Ding
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Ruth Johnson
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Computer Science, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Arjun Bhattacharya
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Institute for Data Science in Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Bogdan Pasaniuc
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Computational Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Institute of Precision Health, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
10
|
Roy DG, Singh L, Chaturvedi HK, Chinnaswamy S. Gender-dependent multiple cross-phenotype association of interferon lambda genetic variants with peripheral blood profiles in healthy individuals. Mol Genet Genomic Med 2024; 12:e2292. [PMID: 37795763 PMCID: PMC10767428 DOI: 10.1002/mgg3.2292] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/08/2023] [Accepted: 09/19/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Type III interferons (IFN), also called as lambda IFNs (IFN-λs), are antiviral and immunomodulatory cytokines that are evolutionarily important in humans. Given their central roles in innate immunity, they could be influencing other aspects of human biology. This study aimed to examine the association of genetic variants that control the expression and/or activity of IFN-λ3 and IFN-λ4 with multiple phenotypes in blood profiles of healthy individuals. METHODS In a cohort of about 550 self-declared healthy individuals, after applying several exclusion criteria to determine their health status, we measured 30 blood parameters, including cellular, biochemical, and metabolic profiles. We genotyped them at rs12979860 and rs28416813 using competitive allele-specific PCR assays and tested their association with the blood profiles under dominant and recessive models for the minor allele. IFN-λ4 variants rs368234815 and rs117648444 were also genotyped or inferred. RESULTS We saw no association in the combined cohort under either of the models for any of the phenotypes. When we stratified the cohort based on gender, we saw a significant association only in males with monocyte (p = 1 × 10-3 ) and SGOT (p = 7 × 10-3 ) levels under the dominant model and with uric acid levels (p = 0.01) under the recessive model. When we tested the IFN-λ4 activity modifying variant within groupings based on absence or presence of one or two copies of IFN-λ4 and on different activity levels of IFN-λ4, we found significant (p < 0.05) association with several phenotypes like monocyte, triglyceride, VLDL, ALP, and uric acid levels, only in males. All the above significant associations did not show any confounding when we tested for the same with up to ten different demographic and lifestyle variables. CONCLUSIONS These results show that lambda interferons can have pleiotropic effects. However, gender seems to be an effect modifier, with males being more sensitive than females to the effect.
Collapse
Affiliation(s)
- Debarati Guha Roy
- Infectious Disease GeneticsNational Institute of Biomedical GenomicsKalyaniIndia
- Regional Centre for BiotechnologyFaridabadIndia
| | - Lucky Singh
- ICMR‐National Institute of Medical StatisticsNew DelhiIndia
| | | | - Sreedhar Chinnaswamy
- Infectious Disease GeneticsNational Institute of Biomedical GenomicsKalyaniIndia
- Regional Centre for BiotechnologyFaridabadIndia
| |
Collapse
|
11
|
Cao Q, Du X, Jiang XY, Tian Y, Gao CH, Liu ZY, Xu T, Tao XX, Lei M, Wang XQ, Ye LL, Duan DD. Phenome-wide association study and precision medicine of cardiovascular diseases in the post-COVID-19 era. Acta Pharmacol Sin 2023; 44:2347-2357. [PMID: 37532784 PMCID: PMC10692238 DOI: 10.1038/s41401-023-01119-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 05/29/2023] [Indexed: 08/04/2023]
Abstract
SARS-CoV-2 infection causes injuries of not only the lungs but also the heart and endothelial cells in vasculature of multiple organs, and induces systemic inflammation and immune over-reactions, which makes COVID-19 a disease phenome that simultaneously affects multiple systems. Cardiovascular diseases (CVD) are intrinsic risk and causative factors for severe COVID-19 comorbidities and death. The wide-spread infection and reinfection of SARS-CoV-2 variants and the long-COVID may become a new common threat to human health and propose unprecedented impact on the risk factors, pathophysiology, and pharmacology of many diseases including CVD for a long time. COVID-19 has highlighted the urgent demand for precision medicine which needs new knowledge network to innovate disease taxonomy for more precise diagnosis, therapy, and prevention of disease. A deeper understanding of CVD in the setting of COVID-19 phenome requires a paradigm shift from the current phenotypic study that focuses on the virus or individual symptoms to phenomics of COVID-19 that addresses the inter-connectedness of clinical phenotypes, i.e., clinical phenome. Here, we summarize the CVD manifestations in the full clinical spectrum of COVID-19, and the phenome-wide association study of CVD interrelated to COVID-19. We discuss the underlying biology for CVD in the COVID-19 phenome and the concept of precision medicine with new phenomic taxonomy that addresses the overall pathophysiological responses of the body to the SARS-CoV-2 infection. We also briefly discuss the unique taxonomy of disease as Zheng-hou patterns in traditional Chinese medicine, and their potential implications in precision medicine of CVD in the post-COVID-19 era.
Collapse
Affiliation(s)
- Qian Cao
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Xin Du
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Xiao-Yan Jiang
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Yuan Tian
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Chen-Hao Gao
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Zi-Yu Liu
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Ting Xu
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Xing-Xing Tao
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Ming Lei
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Xiao-Qiang Wang
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Lingyu Linda Ye
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China.
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, China.
- Key Laboratory of Autoimmune Diseases and Precision Medicie, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750001, China.
| | - Dayue Darrel Duan
- Center for Phenomics of Traditional Chinese Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China.
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, China.
- Key Laboratory of Autoimmune Diseases and Precision Medicie, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750001, China.
- The Department of Pharmacology, University of Nevada Reno School of Medicine, Reno, NV, 89557, USA.
| |
Collapse
|
12
|
McCoy JCS, Spicer JI, Ibbini Z, Tills O. Phenomics as an approach to Comparative Developmental Physiology. Front Physiol 2023; 14:1229500. [PMID: 37645563 PMCID: PMC10461620 DOI: 10.3389/fphys.2023.1229500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/24/2023] [Indexed: 08/31/2023] Open
Abstract
The dynamic nature of developing organisms and how they function presents both opportunity and challenge to researchers, with significant advances in understanding possible by adopting innovative approaches to their empirical study. The information content of the phenotype during organismal development is arguably greater than at any other life stage, incorporating change at a broad range of temporal, spatial and functional scales and is of broad relevance to a plethora of research questions. Yet, effectively measuring organismal development, and the ontogeny of physiological regulations and functions, and their responses to the environment, remains a significant challenge. "Phenomics", a global approach to the acquisition of phenotypic data at the scale of the whole organism, is uniquely suited as an approach. In this perspective, we explore the synergies between phenomics and Comparative Developmental Physiology (CDP), a discipline of increasing relevance to understanding sensitivity to drivers of global change. We then identify how organismal development itself provides an excellent model for pushing the boundaries of phenomics, given its inherent complexity, comparably smaller size, relative to adult stages, and the applicability of embryonic development to a broad suite of research questions using a diversity of species. Collection, analysis and interpretation of whole organismal phenotypic data are the largest obstacle to capitalising on phenomics for advancing our understanding of biological systems. We suggest that phenomics within the context of developing organismal form and function could provide an effective scaffold for addressing grand challenges in CDP and phenomics.
Collapse
Affiliation(s)
| | | | | | - Oliver Tills
- School of Biological and Marine Sciences, University of Plymouth, Plymouth, United Kingdom
| |
Collapse
|
13
|
Woerner J, Sriram V, Nam Y, Verma A, Kim D. Uncovering genetic associations in the human diseasome using an endophenotype-augmented disease network. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.11.23289852. [PMID: 37293013 PMCID: PMC10246076 DOI: 10.1101/2023.05.11.23289852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Many diseases exhibit complex multimorbidities with one another. An intuitive way to model the connections between phenotypes is with a disease-disease network (DDN), where nodes represent diseases and edges represent associations, such as shared single-nucleotide polymorphisms (SNPs), between pairs of diseases. To gain further genetic understanding of molecular contributors to disease associations, we propose a novel version of the shared-SNP DDN (ssDDN), denoted as ssDDN+, which includes connections between diseases derived from genetic correlations with endophenotypes. We hypothesize that a ssDDN+ can provide complementary information to the disease connections in a ssDDN, yielding insight into the role of clinical laboratory measurements in disease interactions. Using PheWAS summary statistics from the UK Biobank, we constructed a ssDDN+ revealing hundreds of genetic correlations between disease phenotypes and quantitative traits. Our augmented network uncovers genetic associations across different disease categories, connects relevant cardiometabolic diseases, and highlights specific biomarkers that are associated with cross-phenotype associations. Out of the 31 clinical measurements under consideration, HDL-C connects the greatest number of diseases and is strongly associated with both type 2 diabetes and diabetic retinopathy. Triglycerides, another blood lipid with known genetics causes in non-mendelian diseases, also adds a substantial number of edges to the ssDDN. Our study can facilitate future network-based investigations of cross-phenotype associations involving pleiotropy and genetic heterogeneity, potentially uncovering sources of missing heritability in multimorbidities.
Collapse
Affiliation(s)
- Jakob Woerner
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vivek Sriram
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yonghyun Nam
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anurag Verma
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Dokyoon Kim
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
14
|
Kerchberger VE, Peterson JF, Wei WQ. Scanning the medical phenome to identify new diagnoses after recovery from COVID-19 in a US cohort. J Am Med Inform Assoc 2023; 30:233-244. [PMID: 36005898 PMCID: PMC9452157 DOI: 10.1093/jamia/ocac159] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/29/2022] [Accepted: 08/23/2022] [Indexed: 01/20/2023] Open
Abstract
OBJECTIVE COVID-19 survivors are at risk for long-term health effects, but assessing the sequelae of COVID-19 at large scales is challenging. High-throughput methods to efficiently identify new medical problems arising after acute medical events using the electronic health record (EHR) could improve surveillance for long-term consequences of acute medical problems like COVID-19. MATERIALS AND METHODS We augmented an existing high-throughput phenotyping method (PheWAS) to identify new diagnoses occurring after an acute temporal event in the EHR. We then used the temporal-informed phenotypes to assess development of new medical problems among COVID-19 survivors enrolled in an EHR cohort of adults tested for COVID-19 at Vanderbilt University Medical Center. RESULTS The study cohort included 186 105 adults tested for COVID-19 from March 5, 2020 to November 1, 2021; of which 30 088 (16.2%) tested positive. Median follow-up after testing was 412 days (IQR 274-528). Our temporal-informed phenotyping was able to distinguish phenotype chapters based on chronicity of their constituent diagnoses. PheWAS with temporal-informed phenotypes identified increased risk for 43 diagnoses among COVID-19 survivors during outpatient follow-up, including multiple new respiratory, cardiovascular, neurological, and pregnancy-related conditions. Findings were robust to sensitivity analyses, and several phenotypic associations were supported by changes in outpatient vital signs or laboratory tests from the pretesting to postrecovery period. CONCLUSION Temporal-informed PheWAS identified new diagnoses affecting multiple organ systems among COVID-19 survivors. These findings can inform future efforts to enable longitudinal health surveillance for survivors of COVID-19 and other acute medical conditions using the EHR.
Collapse
Affiliation(s)
- Vern Eric Kerchberger
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Josh F Peterson
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
15
|
Guan S, Yu YN, Li B, Gu H, Chen L, Wang N, Wang B, Liu X, Liu J, Wang Z. Discovery of Drug-Responsive Phenomic Alteration-Related Driver Genes in the Treatment of Coronary Heart Disease. Pharmgenomics Pers Med 2023; 16:201-217. [PMID: 36945217 PMCID: PMC10024908 DOI: 10.2147/pgpm.s398522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 02/25/2023] [Indexed: 03/17/2023] Open
Abstract
Background The Xueyu Zheng (XYZ) phenome is central to coronary heart disease (CHD), but efforts to detect genetic associations in the XYZ phenome have been disappointing. Methods The phenomic alteration-related genes (PARGs) for the XYZ phenome were screened using |ρ| > 0.4 and p < 0.05 after treatment with Danhong injection at day 14 and day 30. Then, the driver genes for the Protein-Protein Interaction (PPI) networks of the PARGs established using STRING 11.0 were detected using a personalized network control algorithm (PNC). Finally, the molecular correlations of the driver genes with the XYZ phenome were analyzed with the Gene Ontology (GO) biological processes and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways from a holistic viewpoint. Results A total of 525 and 309 PARGs in the XYZ phenome at day 14 and day 30 were identified. These genes were separately enriched in 48 and 35 pathways. Furthermore, five driver genes were detected. These genes were mainly correlated with endoplasmic reticulum stress-mediated apoptosis and autophagy regulation, which could suppress atherosclerosis progression. Conclusion Our study detected the drug-responsive PARGs of the XYZ phenome in CHD and provides an exemplary strategy to investigate the genetic associations among this common phenome and its component symptoms in patients with CHD. Trial Registration ClinicalTrials.gov, NCT01681316; registered on September 7, 2012.
Collapse
Affiliation(s)
- Shuang Guan
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Ya-Nan Yu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Bing Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Hao Gu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Lin Chen
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Nian Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Bo Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Xi Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Jun Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
| | - Zhong Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, People’s Republic of China
- Correspondence: Zhong Wang; Jun Liu, Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, No. 16 Nanxiaojie, Dongzhimennei, Beijing, People’s Republic of China, Email ;
| |
Collapse
|
16
|
Liang X, Cao X, Sha Q, Zhang S. HCLC-FC: A novel statistical method for phenome-wide association studies. PLoS One 2022; 17:e0276646. [PMID: 36350801 PMCID: PMC9645610 DOI: 10.1371/journal.pone.0276646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 10/11/2022] [Indexed: 11/11/2022] Open
Abstract
The emergence of genetic data coupled to longitudinal electronic medical records (EMRs) offers the possibility of phenome-wide association studies (PheWAS). In PheWAS, the whole phenome can be divided into numerous phenotypic categories according to the genetic architecture across phenotypes. Currently, statistical analyses for PheWAS are mainly univariate analyses, which test the association between one genetic variant and one phenotype at a time. In this article, we derived a novel and powerful multivariate method for PheWAS. The proposed method involves three steps. In the first step, we apply the bottom-up hierarchical clustering method to partition a large number of phenotypes into disjoint clusters within each phenotypic category. In the second step, the clustering linear combination method is used to combine test statistics within each category based on the phenotypic clusters and obtain p-values from each phenotypic category. In the third step, we propose a new false discovery rate (FDR) control approach. We perform extensive simulation studies to compare the performance of our method with that of other existing methods. The results show that our proposed method controls FDR very well and outperforms other methods we compared with. We also apply the proposed approach to a set of EMR-based phenotypes across more than 300,000 samples from the UK Biobank. We find that the proposed approach not only can well-control FDR at a nominal level but also successfully identify 1,244 significant SNPs that are reported to be associated with some phenotypes in the GWAS catalog. Our open-access tools and instructions on how to implement HCLC-FC are available at https://github.com/XiaoyuLiang/HCLCFC.
Collapse
Affiliation(s)
- Xiaoyu Liang
- Department of Preventive Medicine, Division of Biostatistics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Xuewei Cao
- Department of Mathematical Sciences, Michigan Technological University, Houghton, Michigan, United States of America
| | - Qiuying Sha
- Department of Mathematical Sciences, Michigan Technological University, Houghton, Michigan, United States of America
| | - Shuanglin Zhang
- Department of Mathematical Sciences, Michigan Technological University, Houghton, Michigan, United States of America
| |
Collapse
|
17
|
Akwo EA, Chen HC, Liu G, Triozzi JL, Tao R, Yu Z, Chung CP, Giri A, Ikizler TA, Stein CM, Siew ED, Feng Q, Robinson-Cohen C, Hung AM, the VA Million Veteran Program 12. Phenome-Wide Association Study of UMOD Gene Variants and Differential Associations With Clinical Outcomes Across Populations in the Million Veteran Program a Multiethnic Biobank. Kidney Int Rep 2022; 7:1802-1818. [PMID: 35967117 PMCID: PMC9366371 DOI: 10.1016/j.ekir.2022.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/22/2022] [Accepted: 05/09/2022] [Indexed: 11/19/2022] Open
Abstract
Introduction Common variants in the UMOD gene are considered an evolutionary adaptation against urinary tract infections (UTIs) and have been implicated in kidney stone formation, chronic kidney disease (CKD), and hypertension. However, differences in UMOD variant-phenotype associations across population groups are unclear. Methods We tested associations between UMOD/PDILT variants and up to 1528 clinical diagnosis codes mapped to phenotype groups in the Million Veteran Program (MVP), using published phenome-wide association study (PheWAS) methodology. Associations were tested using logistic regression adjusted for age, sex, and 10 principal components of ancestry. Bonferroni correction for multiple comparisons was applied. Results Among 648,593 veterans, mean (SD) age was 62 (14) years; 9% were female, 19% Black, and 8% Hispanic. In White patients, the rs4293393 UMOD risk variant associated with increased uromodulin was associated with increased odds of CKD (odds ratio [OR]: 1.22, 95% CI: 1.20-1.24, P = 5.90 × 10-111), end-stage kidney disease (OR: 1.17, 95% CI: 1.11-1.24, P = 2.40 × 10-09), and hypertension (OR: 1.03, 95% CI: 1.05-1.05, P = 2.11 × 10-06) and significantly lower odds of UTIs (OR: 0.94, 95% CI: 0.92-0.96, P = 1.21 × 10-10) and kidney calculus (OR: 0.85, 95% CI: 0.83-0.86, P = 4.27 × 10-69). Similar findings were observed across UMOD/PDILT variants. The rs77924615 PDILT variant had stronger associations with acute cystitis in White female (OR: 0.73, 95% CI: 0.59-0.91, P = 4.98 × 10-03) versus male (OR: 0.99, 95% CI: 0.89-1.11, P = 8.80 × 10-01) (P interaction = 0.01) patients. In Black patients, the rs77924615 PDILT variant was significantly associated with pyelonephritis (OR: 0.65, 95% CI: 0.54-0.79, P = 1.05 × 10-05), whereas associations with UMOD promoter variants were attenuated. Conclusion Robust associations were observed between UMOD/PDILT variants linked with increased uromodulin expression and lower odds of UTIs and calculus and increased odds of CKD and hypertension. However, these associations varied significantly across ancestry groups and sex.
Collapse
Affiliation(s)
- Elvis A. Akwo
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Center for Kidney Disease, Nashville, Tennessee, USA
- VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Hua-Chang Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ge Liu
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jefferson L. Triozzi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ran Tao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Genetics Institute, Nashville, Tennessee, USA
| | - Zhihong Yu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Cecilia P. Chung
- VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
- Vanderbilt Genetics Institute, Nashville, Tennessee, USA
- Division of Rheumatology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ayush Giri
- Vanderbilt Genetics Institute, Nashville, Tennessee, USA
- Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - T. Alp Ikizler
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Center for Kidney Disease, Nashville, Tennessee, USA
- VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - C. Michael Stein
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Edward D. Siew
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Center for Kidney Disease, Nashville, Tennessee, USA
- VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - QiPing Feng
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Cassianne Robinson-Cohen
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Center for Kidney Disease, Nashville, Tennessee, USA
- VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Adriana M. Hung
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Center for Kidney Disease, Nashville, Tennessee, USA
- VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - the VA Million Veteran Program12
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Center for Kidney Disease, Nashville, Tennessee, USA
- VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Genetics Institute, Nashville, Tennessee, USA
- Division of Rheumatology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
18
|
Zhang X, Lucas AM, Veturi Y, Drivas TG, Bone WP, Verma A, Chung WK, Crosslin D, Denny JC, Hebbring S, Jarvik GP, Kullo I, Larson EB, Rasmussen-Torvik LJ, Schaid DJ, Smoller JW, Stanaway IB, Wei WQ, Weng C, Ritchie MD. Large-scale genomic analyses reveal insights into pleiotropy across circulatory system diseases and nervous system disorders. Nat Commun 2022; 13:3428. [PMID: 35701404 PMCID: PMC9198016 DOI: 10.1038/s41467-022-30678-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 05/10/2022] [Indexed: 01/18/2023] Open
Abstract
Clinical and epidemiological studies have shown that circulatory system diseases and nervous system disorders often co-occur in patients. However, genetic susceptibility factors shared between these disease categories remain largely unknown. Here, we characterized pleiotropy across 107 circulatory system and 40 nervous system traits using an ensemble of methods in the eMERGE Network and UK Biobank. Using a formal test of pleiotropy, five genomic loci demonstrated statistically significant evidence of pleiotropy. We observed region-specific patterns of direction of genetic effects for the two disease categories, suggesting potential antagonistic and synergistic pleiotropy. Our findings provide insights into the relationship between circulatory system diseases and nervous system disorders which can provide context for future prevention and treatment strategies.
Collapse
Affiliation(s)
- Xinyuan Zhang
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Anastasia M Lucas
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yogasudha Veturi
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Theodore G Drivas
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - William P Bone
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Anurag Verma
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Wendy K Chung
- Department of Pediatrics and Medicine, Columbia University, New York, NY, 10032, USA
| | - David Crosslin
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA, 98109, USA
| | - Joshua C Denny
- Department of Medicine, Vanderbilt University, Nashville, TN, 37235, USA
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, 37230, USA
| | - Scott Hebbring
- Center for Human Genetics, Marshfield Clinic, Marshfield, WI, 54449, USA
| | - Gail P Jarvik
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA, 98109, USA
| | - Iftikhar Kullo
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, 55905, USA
| | - Eric B Larson
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, 98101, USA
| | - Laura J Rasmussen-Torvik
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Daniel J Schaid
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jordan W Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Ian B Stanaway
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA, 98109, USA
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN, 37230, USA
| | - Chunhua Weng
- Department of Biomedical Informatics, Columbia University, New York, NY, 10032, USA
| | - Marylyn D Ritchie
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
19
|
Dumancas GG, Harrison D, Rico JA, Zamora PRFC, Liwag AG, Villaruz JF, Guanzon MLVV, Ferraris HFD, Jalandoni PJB, Padernal WF, Villareal BNL, Maculada RA, Fernandez RMA, Villa FR, de Castro R. Are phenome-wide association studies feasible in a developing country? Trends Genet 2022; 38:885-888. [PMID: 35660028 DOI: 10.1016/j.tig.2022.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/02/2022] [Accepted: 05/03/2022] [Indexed: 11/18/2022]
Abstract
Phenome-wide association studies (PheWASs), a powerful approach that examines phenotypes associated with a genetic marker, have been used extensively in highly developed countries. Although there may be a clear need for PheWAS in a developing country such as the Philippines, limitations related to resources and practicality would make conducting them a challenge.
Collapse
Affiliation(s)
- Gerard G Dumancas
- Department of Chemistry, Loyola Science Center, The University of Scranton, Scranton, PA 18510, USA; Balik Scientist Program, Department of Science and Technology-Philippine Council for Health Research and Development, Bicutan, Taguig City, 1631, Philippines.
| | - Destiny Harrison
- Department of Mathematics and Physical Sciences, Louisiana State University - Alexandria, Alexandria, LA 71302, USA
| | - Jonathan Adam Rico
- Center for Informatics, University of San Agustin, Gen. Luna St, Iloilo City, 5000, Philippines
| | | | - Aretha G Liwag
- West Visayas State University, Luna St, Lapaz, Iloilo City, 5000, Philippines
| | - Joselito F Villaruz
- West Visayas State University, Luna St, Lapaz, Iloilo City, 5000, Philippines
| | - Ma Luz Vicenta V Guanzon
- Corazon Locsin Montelibano Memorial Regional Hospital, Lacson St, Bacolod, Negros Occidental, 6100, Philippines
| | - Hans Francis D Ferraris
- Corazon Locsin Montelibano Memorial Regional Hospital, Lacson St, Bacolod, Negros Occidental, 6100, Philippines
| | | | | | | | | | | | | | - Romulo de Castro
- Center for Informatics, University of San Agustin, Gen. Luna St, Iloilo City, 5000, Philippines; Balik Scientist Program, Department of Science and Technology-Philippine Council for Health Research and Development, Bicutan, Taguig City, 1631, Philippines
| |
Collapse
|
20
|
Haleem A, Klees S, Schmitt AO, Gültas M. Deciphering Pleiotropic Signatures of Regulatory SNPs in Zea mays L. Using Multi-Omics Data and Machine Learning Algorithms. Int J Mol Sci 2022; 23:5121. [PMID: 35563516 PMCID: PMC9100765 DOI: 10.3390/ijms23095121] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/28/2022] [Accepted: 05/02/2022] [Indexed: 01/25/2023] Open
Abstract
Maize is one of the most widely grown cereals in the world. However, to address the challenges in maize breeding arising from climatic anomalies, there is a need for developing novel strategies to harness the power of multi-omics technologies. In this regard, pleiotropy is an important genetic phenomenon that can be utilized to simultaneously enhance multiple agronomic phenotypes in maize. In addition to pleiotropy, another aspect is the consideration of the regulatory SNPs (rSNPs) that are likely to have causal effects in phenotypic development. By incorporating both aspects in our study, we performed a systematic analysis based on multi-omics data to reveal the novel pleiotropic signatures of rSNPs in a global maize population. For this purpose, we first applied Random Forests and then Markov clustering algorithms to decipher the pleiotropic signatures of rSNPs, based on which hierarchical network models are constructed to elucidate the complex interplay among transcription factors, rSNPs, and phenotypes. The results obtained in our study could help to understand the genetic programs orchestrating multiple phenotypes and thus could provide novel breeding targets for the simultaneous improvement of several agronomic traits.
Collapse
Affiliation(s)
- Ataul Haleem
- Breeding Informatics Group, Department of Animal Sciences, Georg-August University, Margarethe von Wrangell-Weg 7, 37075 Göttingen, Germany; (A.H.); (S.K.); (A.O.S.)
- Faculty of Agriculture, South Westphalia University of Applied Sciences, Lübecker Ring 2, 59494 Soest, Germany
| | - Selina Klees
- Breeding Informatics Group, Department of Animal Sciences, Georg-August University, Margarethe von Wrangell-Weg 7, 37075 Göttingen, Germany; (A.H.); (S.K.); (A.O.S.)
- Center for Integrated Breeding Research (CiBreed), Georg-August University, Carl-Sprengel-Weg 1, 37075 Göttingen, Germany
| | - Armin Otto Schmitt
- Breeding Informatics Group, Department of Animal Sciences, Georg-August University, Margarethe von Wrangell-Weg 7, 37075 Göttingen, Germany; (A.H.); (S.K.); (A.O.S.)
- Center for Integrated Breeding Research (CiBreed), Georg-August University, Carl-Sprengel-Weg 1, 37075 Göttingen, Germany
| | - Mehmet Gültas
- Faculty of Agriculture, South Westphalia University of Applied Sciences, Lübecker Ring 2, 59494 Soest, Germany
- Center for Integrated Breeding Research (CiBreed), Georg-August University, Carl-Sprengel-Weg 1, 37075 Göttingen, Germany
| |
Collapse
|
21
|
Villa O, Stuhr NL, Yen CA, Crimmins EM, Arpawong TE, Curran SP. Genetic variation in ALDH4A1 is associated with muscle health over the lifespan and across species. eLife 2022; 11:74308. [PMID: 35470798 PMCID: PMC9106327 DOI: 10.7554/elife.74308] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
The influence of genetic variation on the aging process, including the incidence and severity of age-related diseases, is complex. Here, we define the evolutionarily conserved mitochondrial enzyme ALH-6/ALDH4A1 as a predictive biomarker for age-related changes in muscle health by combining Caenorhabditis elegans genetics and a gene-wide association scanning (GeneWAS) from older human participants of the US Health and Retirement Study (HRS). In a screen for mutations that activate oxidative stress responses, specifically in the muscle of C. elegans, we identified 96 independent genetic mutants harboring loss-of-function alleles of alh-6, exclusively. Each of these genetic mutations mapped to the ALH-6 polypeptide and led to the age-dependent loss of muscle health. Intriguingly, genetic variants in ALDH4A1 show associations with age-related muscle-related function in humans. Taken together, our work uncovers mitochondrial alh-6/ALDH4A1 as a critical component to impact normal muscle aging across species and a predictive biomarker for muscle health over the lifespan. Ageing is inevitable, but what makes one person ‘age well’ and another decline more quickly remains largely unknown. While many aspects of ageing are clearly linked to genetics, the specific genes involved often remain unidentified. Sarcopenia is an age-related condition affecting the muscles. It involves a gradual loss of muscle mass that becomes faster with age, and is associated with loss of mobility, decreased quality of life, and increased risk of death. Around half of all people aged 80 and over suffer from sarcopenia. Several lifestyle factors, especially poor diet and lack of exercise, are associated with the condition, but genetics is also involved: the condition accelerates more quickly in some people than others, and even fit, physically active individuals can be affected. To study the genetics of conditions like sarcopenia, researchers often use animals like flies or worms, which have short generation times but share genetic similarities with humans. For example, the worm Caenorhabditis elegans has equivalents of several human muscle genes, including the gene alh-6. In worms, alh-6 is important for maintaining energy supply to the muscles, and mutating it not only leads to muscle damage but also to premature ageing. Given this insight, Villa, Stuhr, Yen et al. wanted to determine if variation in the human version of alh-6, ALDH4A1, also contributes to individual differences in muscle ageing and decline in humans. Evaluating variation in this gene required a large amount of genetic data from older adults. These were taken from a continuous study that follows >35,000 older adults. Importantly, the study collects not only information on gene sequences but also measures of muscle health and performance over time for each individual. Analysis of these genetic data revealed specific small variations in the DNA of ALDH4A1, all of which associated with reduced muscle health. Follow-up experiments in worms used genetic engineering techniques to test how variation in the worm alh-6 gene could influence age-related health. The resulting mutant worms developed muscle problems much earlier than their normal counterparts, supporting the role of alh-6/ALDH4A1 in determining muscle health across the lifespan of both worms and humans. These results have identified a key influencer of muscle health during ageing in worms, and emphasize the importance of validating effects of genetic variation among humans during this process. Villa, Stuhr, Yen et al. hope that this study will help researchers find more genetic ‘markers’ of muscle health, and ultimately allow us to predict an individual’s risk of sarcopenia based on their genetic make-up.
Collapse
Affiliation(s)
- Osvaldo Villa
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, United States
| | - Nicole L Stuhr
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, United States.,Dornsife College of Letters, Arts, and Science, Department of Molecular and Computational Biology, University of Southern California, Los Angeles, United States
| | - Chia-An Yen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, United States.,Dornsife College of Letters, Arts, and Science, Department of Molecular and Computational Biology, University of Southern California, Los Angeles, United States
| | - Eileen M Crimmins
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, United States
| | - Thalida Em Arpawong
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, United States
| | - Sean P Curran
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, United States.,Dornsife College of Letters, Arts, and Science, Department of Molecular and Computational Biology, University of Southern California, Los Angeles, United States.,Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, United States
| |
Collapse
|
22
|
Maturation and application of phenome-wide association studies. Trends Genet 2022; 38:353-363. [PMID: 34991903 DOI: 10.1016/j.tig.2021.12.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 11/12/2021] [Accepted: 12/02/2021] [Indexed: 12/12/2022]
Abstract
In the past 10 years since its introduction, phenome-wide association studies (PheWAS) have uncovered novel genotype-phenotype relationships. Along the way, PheWAS have evolved in many aspects as a study design with the expanded availability of large data repositories with genome-wide data linked to detailed phenotypic data. Advancement in methods, including algorithms, software, and publicly available integrated resources, makes it feasible to more fully realize the potential of PheWAS, overcoming the previous computational and analytical limitations. We review here the most recent improvements and notable applications of PheWAS since the second half of the decade from its inception. We also note the challenges that remain embedded along the entire PheWAS analytical pipeline that necessitate further development of tools and resources to further advance the understanding of the complex genetic architecture underlying human diseases and traits.
Collapse
|
23
|
Doran S, Arif M, Lam S, Bayraktar A, Turkez H, Uhlen M, Boren J, Mardinoglu A. Multi-omics approaches for revealing the complexity of cardiovascular disease. Brief Bioinform 2021; 22:bbab061. [PMID: 33725119 PMCID: PMC8425417 DOI: 10.1093/bib/bbab061] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/20/2021] [Accepted: 02/05/2021] [Indexed: 02/06/2023] Open
Abstract
The development and progression of cardiovascular disease (CVD) can mainly be attributed to the narrowing of blood vessels caused by atherosclerosis and thrombosis, which induces organ damage that will result in end-organ dysfunction characterized by events such as myocardial infarction or stroke. It is also essential to consider other contributory factors to CVD, including cardiac remodelling caused by cardiomyopathies and co-morbidities with other diseases such as chronic kidney disease. Besides, there is a growing amount of evidence linking the gut microbiota to CVD through several metabolic pathways. Hence, it is of utmost importance to decipher the underlying molecular mechanisms associated with these disease states to elucidate the development and progression of CVD. A wide array of systems biology approaches incorporating multi-omics data have emerged as an invaluable tool in establishing alterations in specific cell types and identifying modifications in signalling events that promote disease development. Here, we review recent studies that apply multi-omics approaches to further understand the underlying causes of CVD and provide possible treatment strategies by identifying novel drug targets and biomarkers. We also discuss very recent advances in gut microbiota research with an emphasis on how diet and microbial composition can impact the development of CVD. Finally, we present various biological network analyses and other independent studies that have been employed for providing mechanistic explanation and developing treatment strategies for end-stage CVD, namely myocardial infarction and stroke.
Collapse
Affiliation(s)
- Stephen Doran
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, SE1 9RT, United Kingdom
| | - Muhammad Arif
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Simon Lam
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, SE1 9RT, United Kingdom
| | - Abdulahad Bayraktar
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, SE1 9RT, United Kingdom
| | - Hasan Turkez
- Department of Medical Biology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Mathias Uhlen
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Jan Boren
- Institute of Medicine, Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital Gothenburg, Sweden
| | - Adil Mardinoglu
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, SE1 9RT, United Kingdom
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
24
|
Kafkas Ş, Althubaiti S, Gkoutos GV, Hoehndorf R, Schofield PN. Linking common human diseases to their phenotypes; development of a resource for human phenomics. J Biomed Semantics 2021; 12:17. [PMID: 34425897 PMCID: PMC8383460 DOI: 10.1186/s13326-021-00249-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/30/2021] [Indexed: 11/11/2022] Open
Abstract
Background In recent years a large volume of clinical genomics data has become available due to rapid advances in sequencing technologies. Efficient exploitation of this genomics data requires linkage to patient phenotype profiles. Current resources providing disease-phenotype associations are not comprehensive, and they often do not have broad coverage of the disease terminologies, particularly ICD-10, which is still the primary terminology used in clinical settings. Methods We developed two approaches to gather disease-phenotype associations. First, we used a text mining method that utilizes semantic relations in phenotype ontologies, and applies statistical methods to extract associations between diseases in ICD-10 and phenotype ontology classes from the literature. Second, we developed a semi-automatic way to collect ICD-10–phenotype associations from existing resources containing known relationships. Results We generated four datasets. Two of them are independent datasets linking diseases to their phenotypes based on text mining and semi-automatic strategies. The remaining two datasets are generated from these datasets and cover a subset of ICD-10 classes of common diseases contained in UK Biobank. We extensively validated our text mined and semi-automatically curated datasets by: comparing them against an expert-curated validation dataset containing disease–phenotype associations, measuring their similarity to disease–phenotype associations found in public databases, and assessing how well they could be used to recover gene–disease associations using phenotype similarity. Conclusion We find that our text mining method can produce phenotype annotations of diseases that are correct but often too general to have significant information content, or too specific to accurately reflect the typical manifestations of the sporadic disease. On the other hand, the datasets generated from integrating multiple knowledgebases are more complete (i.e., cover more of the required phenotype annotations for a given disease). We make all data freely available at 10.5281/zenodo.4726713. Supplementary Information The online version contains supplementary material available at (10.1186/s13326-021-00249-x).
Collapse
Affiliation(s)
- Şenay Kafkas
- Computational Bioscience Research Center (CBRC), Computer, Electrical, and Mathematical Sciences & Engineering Division, King Abdullah University of Science and Technology, 4700 KAUST, Thuwal, 23955, Saudi Arabia
| | - Sara Althubaiti
- Computational Bioscience Research Center (CBRC), Computer, Electrical, and Mathematical Sciences & Engineering Division, King Abdullah University of Science and Technology, 4700 KAUST, Thuwal, 23955, Saudi Arabia
| | - Georgios V Gkoutos
- Health Data Research UK, Midlands site, Edgbaston, Birmingham, B15 2TT, United Kingdom.,Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom
| | - Robert Hoehndorf
- Computational Bioscience Research Center (CBRC), Computer, Electrical, and Mathematical Sciences & Engineering Division, King Abdullah University of Science and Technology, 4700 KAUST, Thuwal, 23955, Saudi Arabia.
| | - Paul N Schofield
- Department of Physiology, Development & Neuroscience, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom
| |
Collapse
|
25
|
Wang L, Zhang X, Meng X, Koskeridis F, Georgiou A, Yu L, Campbell H, Theodoratou E, Li X. Methodology in phenome-wide association studies: a systematic review. J Med Genet 2021; 58:720-728. [PMID: 34272311 DOI: 10.1136/jmedgenet-2021-107696] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 05/27/2021] [Indexed: 11/04/2022]
Abstract
Phenome-wide association study (PheWAS) has been increasingly used to identify novel genetic associations across a wide spectrum of phenotypes. This systematic review aims to summarise the PheWAS methodology, discuss the advantages and challenges of PheWAS, and provide potential implications for future PheWAS studies. Medical Literature Analysis and Retrieval System Online (MEDLINE) and Excerpta Medica Database (EMBASE) databases were searched to identify all published PheWAS studies up until 24 April 2021. The PheWAS methodology incorporating how to perform PheWAS analysis and which software/tool could be used, were summarised based on the extracted information. A total of 1035 studies were identified and 195 eligible articles were finally included. Among them, 137 (77.0%) contained 10 000 or more study participants, 164 (92.1%) defined the phenome based on electronic medical records data, 140 (78.7%) used genetic variants as predictors, and 73 (41.0%) conducted replication analysis to validate PheWAS findings and almost all of them (94.5%) received consistent results. The methodology applied in these PheWAS studies was dissected into several critical steps, including quality control of the phenome, selecting predictors, phenotyping, statistical analysis, interpretation and visualisation of PheWAS results, and the workflow for performing a PheWAS was established with detailed instructions on each step. This study provides a comprehensive overview of PheWAS methodology to help practitioners achieve a better understanding of the PheWAS design, to detect understudied or overstudied outcomes, and to direct their research by applying the most appropriate software and online tools for their study data structure.
Collapse
Affiliation(s)
- Lijuan Wang
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaomeng Zhang
- Centre for Global Health, The University of Edinburgh Usher Institute of Population Health Sciences and Informatics, Edinburgh, UK
| | - Xiangrui Meng
- Vanke School of Public Health, Tsinghua University, Beijing, China
| | - Fotios Koskeridis
- Department of Hygiene and Epidemiology, University of Ioannina, Ioannina, Epirus, Greece
| | - Andrea Georgiou
- Department of Hygiene and Epidemiology, University of Ioannina, Ioannina, Epirus, Greece
| | - Lili Yu
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Harry Campbell
- Centre for Global Health, The University of Edinburgh Usher Institute of Population Health Sciences and Informatics, Edinburgh, UK
| | - Evropi Theodoratou
- Centre for Global Health, The University of Edinburgh Usher Institute of Population Health Sciences and Informatics, Edinburgh, UK.,Cancer Research UK Edinburgh Centre, The University of Edinburgh MRC Institute of Genetics and Molecular Medicine, Edinburgh, UK
| | - Xue Li
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
26
|
Zhao B, Luo H, Huang X, Wei C, Di J, Tian Y, Fu X, Li B, Liu GE, Fang L, Zhang S, Tian K. Integration of a single-step genome-wide association study with a multi-tissue transcriptome analysis provides novel insights into the genetic basis of wool and weight traits in sheep. Genet Sel Evol 2021; 53:56. [PMID: 34193030 PMCID: PMC8247193 DOI: 10.1186/s12711-021-00649-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 06/22/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Genetic improvement of wool and growth traits is a major goal in the sheep industry, but their underlying genetic architecture remains elusive. To improve our understanding of these mechanisms, we conducted a weighted single-step genome-wide association study (WssGWAS) and then integrated the results with large-scale transcriptome data for five wool traits and one growth trait in Merino sheep: mean fibre diameter (MFD), coefficient of variation of the fibre diameter (CVFD), crimp number (CN), mean staple length (MSL), greasy fleece weight (GFW), and live weight (LW). RESULTS Our dataset comprised 7135 individuals with phenotype data, among which 1217 had high-density (HD) genotype data (n = 372,534). The genotypes of 707 of these animals were imputed from the Illumina Ovine single nucleotide polymorphism (SNP) 54 BeadChip to the HD Array. The heritability of these traits ranged from 0.05 (CVFD) to 0.36 (MFD), and between-trait genetic correlations ranged from - 0.44 (CN vs. LW) to 0.77 (GFW vs. LW). By integrating the GWAS signals with RNA-seq data from 500 samples (representing 87 tissue types from 16 animals), we detected tissues that were relevant to each of the six traits, e.g. liver, muscle and the gastrointestinal (GI) tract were the most relevant tissues for LW, and leukocytes and macrophages were the most relevant cells for CN. For the six traits, 54 quantitative trait loci (QTL) were identified covering 81 candidate genes on 21 ovine autosomes. Multiple candidate genes showed strong tissue-specific expression, e.g. BNC1 (associated with MFD) and CHRNB1 (LW) were specifically expressed in skin and muscle, respectively. By conducting phenome-wide association studies (PheWAS) in humans, we found that orthologues of several of these candidate genes were significantly (FDR < 0.05) associated with similar traits in humans, e.g. BNC1 was significantly associated with MFD in sheep and with hair colour in humans, and CHRNB1 was significantly associated with LW in sheep and with body mass index in humans. CONCLUSIONS Our findings provide novel insights into the biological and genetic mechanisms underlying wool and growth traits, and thus will contribute to the genetic improvement and gene mapping of complex traits in sheep.
Collapse
Affiliation(s)
- Bingru Zhao
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Hanpeng Luo
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xixia Huang
- College of Animal Science, Xinjiang Agricultural University, Urumqi, China
| | - Chen Wei
- College of Animal Science, Xinjiang Agricultural University, Urumqi, China
| | - Jiang Di
- Key Laboratory of Genetics Breeding and Reproduction of the Fine Wool Sheep & Cashmere Goat in Xinjiang, Institute of Animal Science, Xinjiang Academy of Animal Sciences, Urumqi, China
| | - Yuezhen Tian
- Key Laboratory of Genetics Breeding and Reproduction of the Fine Wool Sheep & Cashmere Goat in Xinjiang, Institute of Animal Science, Xinjiang Academy of Animal Sciences, Urumqi, China
| | - Xuefeng Fu
- Key Laboratory of Genetics Breeding and Reproduction of the Fine Wool Sheep & Cashmere Goat in Xinjiang, Institute of Animal Science, Xinjiang Academy of Animal Sciences, Urumqi, China
| | - Bingjie Li
- Scotland's Rural College (SRUC), Roslin Institute Building, Midlothian, EH25 9RG, UK
| | - George E Liu
- Animal Genomics and Improvement Laboratory, Henry A. Wallace Beltsville Agricultural Research Center, Agricultural Research Service, Agricultural Research Service, USDA, Beltsville, MD, USA
| | - Lingzhao Fang
- MRC Human Genetics Unit at the Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK.
| | - Shengli Zhang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics, Breeding, and Reproduction, Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China.
| | - Kechuan Tian
- Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China.
| |
Collapse
|
27
|
Zhao L, Batta I, Matloff W, O'Driscoll C, Hobel S, Toga AW. Neuroimaging PheWAS (Phenome-Wide Association Study): A Free Cloud-Computing Platform for Big-Data, Brain-Wide Imaging Association Studies. Neuroinformatics 2021; 19:285-303. [PMID: 32822005 DOI: 10.1007/s12021-020-09486-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Large-scale, case-control genome-wide association studies (GWASs) have revealed genetic variations associated with diverse neurological and psychiatric disorders. Recent advances in neuroimaging and genomic databases of large healthy and diseased cohorts have empowered studies to characterize effects of the discovered genetic factors on brain structure and function, implicating neural pathways and genetic mechanisms in the underlying biology. However, the unprecedented scale and complexity of the imaging and genomic data requires new advanced biomedical data science tools to manage, process and analyze the data. In this work, we introduce Neuroimaging PheWAS (phenome-wide association study): a web-based system for searching over a wide variety of brain-wide imaging phenotypes to discover true system-level gene-brain relationships using a unified genotype-to-phenotype strategy. This design features a user-friendly graphical user interface (GUI) for anonymous data uploading, study definition and management, and interactive result visualizations as well as a cloud-based computational infrastructure and multiple state-of-art methods for statistical association analysis and multiple comparison correction. We demonstrated the potential of Neuroimaging PheWAS with a case study analyzing the influences of the apolipoprotein E (APOE) gene on various brain morphological properties across the brain in the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort. Benchmark tests were performed to evaluate the system's performance using data from UK Biobank. The Neuroimaging PheWAS system is freely available. It simplifies the execution of PheWAS on neuroimaging data and provides an opportunity for imaging genetics studies to elucidate routes at play for specific genetic variants on diseases in the context of detailed imaging phenotypic data.
Collapse
Affiliation(s)
- Lu Zhao
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Ishaan Batta
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - William Matloff
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Caroline O'Driscoll
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Samuel Hobel
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
28
|
Ahluwalia TS, Prins BP, Abdollahi M, Armstrong NJ, Aslibekyan S, Bain L, Jefferis B, Baumert J, Beekman M, Ben-Shlomo Y, Bis JC, Mitchell BD, de Geus E, Delgado GE, Marek D, Eriksson J, Kajantie E, Kanoni S, Kemp JP, Lu C, Marioni RE, McLachlan S, Milaneschi Y, Nolte IM, Petrelis AM, Porcu E, Sabater-Lleal M, Naderi E, Seppälä I, Shah T, Singhal G, Standl M, Teumer A, Thalamuthu A, Thiering E, Trompet S, Ballantyne CM, Benjamin EJ, Casas JP, Toben C, Dedoussis G, Deelen J, Durda P, Engmann J, Feitosa MF, Grallert H, Hammarstedt A, Harris SE, Homuth G, Hottenga JJ, Jalkanen S, Jamshidi Y, Jawahar MC, Jess T, Kivimaki M, Kleber ME, Lahti J, Liu Y, Marques-Vidal P, Mellström D, Mooijaart SP, Müller-Nurasyid M, Penninx B, Revez JA, Rossing P, Räikkönen K, Sattar N, Scharnagl H, Sennblad B, Silveira A, Pourcain BS, Timpson NJ, Trollor J, van Dongen J, Van Heemst D, Visvikis-Siest S, Vollenweider P, Völker U, Waldenberger M, Willemsen G, Zabaneh D, Morris RW, Arnett DK, Baune BT, Boomsma DI, Chang YPC, Deary IJ, Deloukas P, Eriksson JG, Evans DM, Ferreira MA, Gaunt T, Gudnason V, Hamsten A, Heinrich J, Hingorani A, Humphries SE, Jukema JW, Koenig W, et alAhluwalia TS, Prins BP, Abdollahi M, Armstrong NJ, Aslibekyan S, Bain L, Jefferis B, Baumert J, Beekman M, Ben-Shlomo Y, Bis JC, Mitchell BD, de Geus E, Delgado GE, Marek D, Eriksson J, Kajantie E, Kanoni S, Kemp JP, Lu C, Marioni RE, McLachlan S, Milaneschi Y, Nolte IM, Petrelis AM, Porcu E, Sabater-Lleal M, Naderi E, Seppälä I, Shah T, Singhal G, Standl M, Teumer A, Thalamuthu A, Thiering E, Trompet S, Ballantyne CM, Benjamin EJ, Casas JP, Toben C, Dedoussis G, Deelen J, Durda P, Engmann J, Feitosa MF, Grallert H, Hammarstedt A, Harris SE, Homuth G, Hottenga JJ, Jalkanen S, Jamshidi Y, Jawahar MC, Jess T, Kivimaki M, Kleber ME, Lahti J, Liu Y, Marques-Vidal P, Mellström D, Mooijaart SP, Müller-Nurasyid M, Penninx B, Revez JA, Rossing P, Räikkönen K, Sattar N, Scharnagl H, Sennblad B, Silveira A, Pourcain BS, Timpson NJ, Trollor J, van Dongen J, Van Heemst D, Visvikis-Siest S, Vollenweider P, Völker U, Waldenberger M, Willemsen G, Zabaneh D, Morris RW, Arnett DK, Baune BT, Boomsma DI, Chang YPC, Deary IJ, Deloukas P, Eriksson JG, Evans DM, Ferreira MA, Gaunt T, Gudnason V, Hamsten A, Heinrich J, Hingorani A, Humphries SE, Jukema JW, Koenig W, Kumari M, Kutalik Z, Lawlor DA, Lehtimäki T, März W, Mather KA, Naitza S, Nauck M, Ohlsson C, Price JF, Raitakari O, Rice K, Sachdev PS, Slagboom E, Sørensen TIA, Spector T, Stacey D, Stathopoulou MG, Tanaka T, Wannamethee SG, Whincup P, Rotter JI, Dehghan A, Boerwinkle E, Psaty BM, Snieder H, Alizadeh BZ. Genome-wide association study of circulating interleukin 6 levels identifies novel loci. Hum Mol Genet 2021; 30:393-409. [PMID: 33517400 PMCID: PMC8098112 DOI: 10.1093/hmg/ddab023] [Show More Authors] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/02/2020] [Accepted: 01/04/2021] [Indexed: 12/13/2022] Open
Abstract
Interleukin 6 (IL-6) is a multifunctional cytokine with both pro- and anti-inflammatory properties with a heritability estimate of up to 61%. The circulating levels of IL-6 in blood have been associated with an increased risk of complex disease pathogenesis. We conducted a two-staged, discovery and replication meta genome-wide association study (GWAS) of circulating serum IL-6 levels comprising up to 67 428 (ndiscovery = 52 654 and nreplication = 14 774) individuals of European ancestry. The inverse variance fixed effects based discovery meta-analysis, followed by replication led to the identification of two independent loci, IL1F10/IL1RN rs6734238 on chromosome (Chr) 2q14, (Pcombined = 1.8 × 10-11), HLA-DRB1/DRB5 rs660895 on Chr6p21 (Pcombined = 1.5 × 10-10) in the combined meta-analyses of all samples. We also replicated the IL6R rs4537545 locus on Chr1q21 (Pcombined = 1.2 × 10-122). Our study identifies novel loci for circulating IL-6 levels uncovering new immunological and inflammatory pathways that may influence IL-6 pathobiology.
Collapse
Affiliation(s)
- Tarunveer S Ahluwalia
- Steno Diabetes Center Copenhagen, Gentofte DK2820, Denmark.,Department of Biology, The Bioinformatics Center, University of Copenhagen, Copenhagen DK2200, Denmark
| | - Bram P Prins
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, The Netherlands
| | - Mohammadreza Abdollahi
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, The Netherlands
| | | | - Stella Aslibekyan
- Department of Epidemiology, University of Alabama at Birmingham School of Public Health, Birmingham, Alabama 35233, USA
| | - Lisa Bain
- QIMR Berghofer Medical Research Institute, Brisbane 4006, Australia
| | - Barbara Jefferis
- Department of Primary Care & Population Health, UCL Institute of Epidemiology & Health Care, University College London, London NW3 2PF, UK
| | - Jens Baumert
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Marian Beekman
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden 2300 RC, The Netherlands
| | - Yoav Ben-Shlomo
- Population Health Sciences, University of Bristol, Bristol BS8 2PS, UK
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98101, USA
| | - Braxton D Mitchell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21202, USA
| | - Eco de Geus
- Department of Biological Psychology, Behaviour and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081 BT, The Netherlands.,Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam 1105 AZ, The Netherlands
| | - Graciela E Delgado
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany
| | - Diana Marek
- SIB Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Joel Eriksson
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, Centre for Bone and Arthritis Research (CBAR), University of Gothenburg, Gothenburg 41345, Sweden
| | - Eero Kajantie
- Chronic Disease Prevention Unit, National Institute for Health and Welfare, PO Box 30, Helsinki 00271, Finland.,Hospital for Children and Adolescents, Helsinki University Central Hospital and University of Helsinki, Helsinki 00014, Finland
| | - Stavroula Kanoni
- William Harvey Research Institute, Barts & the London Medical School, Queen Mary University of London, London EC1M 6BQ, UK
| | - John P Kemp
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland 4102, Australia.,MRC Integrative Epidemiology Unit, Population Health Sciences, University of Bristol, Bristol BS8 2BN, UK
| | - Chen Lu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK.,Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Stela McLachlan
- Usher Institute, University of Edinburgh, Edinburgh EH8 9AG, UK
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit, Amsterdam 1081 HJ, The Netherlands
| | - Ilja M Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, The Netherlands
| | | | - Eleonora Porcu
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato (CA) 09042, Italy
| | - Maria Sabater-Lleal
- Cardiovascular Medicine, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm 17176, Sweden.,Unit of Genomics of Complex Diseases, Institut d'Investigació Biomèdica Sant Pau (IIB-Sant Pau), Barcelona 08041, Spain
| | - Elnaz Naderi
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, The Netherlands
| | - Ilkka Seppälä
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
| | - Tina Shah
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
| | - Gaurav Singhal
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide 5005, Australia
| | - Marie Standl
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald 17475, Germany
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney 2052, Australia
| | - Elisabeth Thiering
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg 85764, Germany.,Division of Metabolic Diseases and Nutritional Medicine, Ludwig-Maximilians-University of Munich, Dr. von Hauner Children's Hospital, Munich 80337, Germany
| | - Stella Trompet
- Department of Cardiology, Leiden University Medical Center, Leiden 2300 RC, The Netherlands.,Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | | | - Emelia J Benjamin
- National Heart, Lung, and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA 01702, USA.,Section of Cardiovascular Medicine and Preventive Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Juan P Casas
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA 02130, USA
| | - Catherine Toben
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide 5005, Australia
| | - George Dedoussis
- 44Department of Nutrition-Dietetics, Harokopio University, Athens 17671, Greece
| | - Joris Deelen
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden 2300 RC, The Netherlands.,Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Peter Durda
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Jorgen Engmann
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110-1093, USA
| | - Harald Grallert
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg 85764, Germany.,German Center for Diabetes Research (DZD), Neuherberg 85764, Germany
| | - Ann Hammarstedt
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg SE-41345, Sweden
| | - Sarah E Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9JZ, UK.,Department of Psychology, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald 17475, Germany
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Behaviour and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081 BT, The Netherlands.,Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam 1105 AZ, The Netherlands
| | - Sirpa Jalkanen
- MediCity Research Laboratory, University of Turku, Turku 20520, Finland.,Department of Medical Microbiology and Immunology, University of Turku, Turku 20520, Finland
| | - Yalda Jamshidi
- Genetics Research Centre, Molecular and Clinical Sciences Institute, St George's University of London, London SW17 0RE, UK
| | - Magdalene C Jawahar
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide 5005, Australia
| | - Tine Jess
- 55Department of Epidemiology Research, Statens Serum Institute, Copenhagen DK2300, Denmark
| | - Mika Kivimaki
- Department of Epidemiology & Public Health, UCL Institute of Epidemiology & Health Care, University College London, London WC1E 7HB, UK
| | - Marcus E Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany
| | - Jari Lahti
- Turku Institute for Advanced Studies, University of Turku, Turku 20014, Finland.,Department of Psychology and Logopedics, University of Helsinki, Helsinki 00014, Finland
| | - Yongmei Liu
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Pedro Marques-Vidal
- Department of Internal Medicine, Lausanne University Hospital (CHUV), Lausanne 1011, Switzerland.,University of Lausanne, Lausanne 1011, Switzerland
| | - Dan Mellström
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, Centre for Bone and Arthritis Research (CBAR), University of Gothenburg, Gothenburg 41345, Sweden
| | - Simon P Mooijaart
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
| | - Martina Müller-Nurasyid
- IBE, Faculty of Medicine, Ludwig Maximilians University (LMU) Munich, Munich 81377, Germany.,Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johhanes Gutenberg University, Mainz 55101, Germany
| | - Brenda Penninx
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit, Amsterdam 1081 HJ, The Netherlands
| | - Joana A Revez
- QIMR Berghofer Medical Research Institute, Brisbane 4006, Australia
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte DK2820, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen DK2200, Denmark
| | - Katri Räikkönen
- Department of Psychology and Logopedics, University of Helsinki, Helsinki 00014, Finland
| | - Naveed Sattar
- BHF Glasgow Cardiovascular Research Centre, Faculty of Medicine, Glasgow G12 8TA, UK
| | - Hubert Scharnagl
- 66Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz 8036, Austria
| | - Bengt Sennblad
- Cardiovascular Medicine, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm 17176, Sweden.,Department of Cell and Molecular Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala 75124, Sweden
| | - Angela Silveira
- Cardiovascular Medicine, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm 17176, Sweden
| | - Beate St Pourcain
- MRC Integrative Epidemiology Unit, Population Health Sciences, University of Bristol, Bristol BS8 2BN, UK.,Max Planck Institute for Psycholinguistics, Nijmegen XD 6525, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen 6525 AJ, The Netherlands
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit, Population Health Sciences, University of Bristol, Bristol BS8 2BN, UK
| | - Julian Trollor
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney 2052, Australia.,Department of Developmental Disability Neuropsychiatry, School of Psychiatry, University of New South Wales, Sydney 2031, Australia
| | | | - Jenny van Dongen
- Department of Biological Psychology, Behaviour and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081 BT, The Netherlands.,Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam 1105 AZ, The Netherlands
| | | | | | - Peter Vollenweider
- Department of Internal Medicine, Lausanne University Hospital (CHUV), Lausanne 1011, Switzerland.,University of Lausanne, Lausanne 1011, Switzerland
| | - Uwe Völker
- MediCity Research Laboratory, University of Turku, Turku 20520, Finland
| | - Melanie Waldenberger
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Gonneke Willemsen
- Department of Biological Psychology, Behaviour and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081 BT, The Netherlands.,Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam 1105 AZ, The Netherlands
| | - Delilah Zabaneh
- Department of Genetics, Environment and Evolution, University College London Genetics Institute, London WC1E 6BT, UK
| | - Richard W Morris
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 1UD, UK
| | - Donna K Arnett
- Dean's Office, College of Public Health, University of Kentucky, Lexington, KY 40536, USA
| | - Bernhard T Baune
- Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Parkville 3000, Australia.,Department of Psychiatry and Psychotherapy, University of Muenster, Muenster 48149, Germany.,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville 3000, Australia
| | - Dorret I Boomsma
- Department of Biological Psychology, Behaviour and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081 BT, The Netherlands.,Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam 1105 AZ, The Netherlands
| | - Yen-Pei C Chang
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21202, USA
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9JZ, UK.,Department of Psychology, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Panos Deloukas
- William Harvey Research Institute, Barts & the London Medical School, Queen Mary University of London, London EC1M 6BQ, UK.,77Centre for Genomic Health, Queen Mary University of London, London EC1M 6BQ, UK
| | - Johan G Eriksson
- National Institute for Health and Welfare, University of Helsinki, Helsinki 00014, Finland.,Department of General Practice and Primary Health Care, University of Helsinki, Helsinki 00014, Finland
| | - David M Evans
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland 4102, Australia.,MRC Integrative Epidemiology Unit, Population Health Sciences, University of Bristol, Bristol BS8 2BN, UK
| | | | - Tom Gaunt
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol BS6 2BN, UK.,Population Health Science, Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kópavogur 201, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - Anders Hamsten
- Cardiovascular Medicine, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm 17176, Sweden
| | - Joachim Heinrich
- Institute of Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg 85764, Germany.,Institute and Clinic for Occupational, Social and Environmental Medicine, University Hospital, LMU Munich, Munich 81377, Germany.,Allergy and Lung Health Unit, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne 3010, Australia
| | - Aroon Hingorani
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
| | - Steve E Humphries
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
| | - J Wouter Jukema
- Section of Gerontology and Geriatrics, Department of Internal Medicine, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands.,Durrer Center for Cardiogenetic Research, Amsterdam 1105 AZ, The Netherlands
| | - Wolfgang Koenig
- Deutsches Herzzentrum München, Technische Universität München, Munich 80636, Germany.,88DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich 80336, Germany.,Institute of Epidemiology and Medical Biometry, University of Ulm, Ulm 89081, Germany
| | - Meena Kumari
- Department of Epidemiology & Public Health, UCL Institute of Epidemiology & Health Care, University College London, London WC1E 7HB, UK.,Institute for Social and Economic Research, University of Essex, Colchester CO4 3SQ, Germany
| | - Zoltan Kutalik
- SIB Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland.,University Center for Primary Care and Public Health, University of Lausanne, Lausanne 1010, Switzerland
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol BS6 2BN, UK.,Population Health Science, Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere 33520, Finland
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty Mannheim, University of Heidelberg, Mannheim 68167, Germany.,66Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz 8036, Austria.,SYNLAB Academy, SYNALB Holding Deutschland GmbH, Mannheim 68163, Germany
| | - Karen A Mather
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney 2052, Australia.,Neuroscience Research Australia, Sydney 2031, Australia
| | - Silvia Naitza
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato (CA) 09042, Italy
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald 17475, Germany.,DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald 17475, Germany
| | - Claes Ohlsson
- Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, Centre for Bone and Arthritis Research (CBAR), University of Gothenburg, Gothenburg 41345, Sweden
| | - Jackie F Price
- Usher Institute, University of Edinburgh, Edinburgh EH8 9AG, UK
| | - Olli Raitakari
- Centre for Population Health Research, University of Turku, Turku University Hospital, Turku 20520, Finland.,Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku 20520, Finland.,Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku 20014, Finland
| | - Ken Rice
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney 2052, Australia.,Neuropsychiatric Institute, Prince of Wales Hospital, Sydney 2031, Australia
| | - Eline Slagboom
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden 2300 RC, The Netherlands.,Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Thorkild I A Sørensen
- Novo Nordisk Foundation Center For Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK2200, Denmark.,Department of Public Health, Section on Epidemiology, University of Copenhagen, Copenhagen DK1014, Denmark
| | - Tim Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London SE1 7EH, UK
| | - David Stacey
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge CB1 8RN, UK
| | | | - Toshiko Tanaka
- Longitudinal Study Section, Translational Gerontology Branch, National Institute on Aging, Baltimore, MD 21224, USA
| | - S Goya Wannamethee
- Department of Primary Care & Population Health, UCL Institute of Epidemiology & Health Care, University College London, London NW3 2PF, UK
| | - Peter Whincup
- Population Health Research Institute, St George's, University of London, London SW17 0RE, UK
| | - Jerome I Rotter
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus MC, Rotterdam 3000 CA, The Netherlands
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.,Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 98101, USA.,Departments of Epidemiology and Health Services, University of Washington, Seattle, WA 98101, USA
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, The Netherlands
| | - Behrooz Z Alizadeh
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen 9700 RB, The Netherlands
| |
Collapse
|
29
|
Zhang X, Li R, Ritchie MD. Statistical Impact of Sample Size and Imbalance on Multivariate Analysis in silico and A Case Study in the UK Biobank. AMIA ... ANNUAL SYMPOSIUM PROCEEDINGS. AMIA SYMPOSIUM 2021; 2020:1383-1391. [PMID: 33936514 PMCID: PMC8075427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Large-scale biobank cohorts coupled with electronic health records offer unprecedented opportunities to study genotype-phenotype relationships. Genome-wide association studies uncovered disease-associated loci through univariate methods, with the focus on one trait at a time. With genetic variants being identifiedfor thousands of traits, researchers found that 90% of human genetic loci are associated with more than one trait, highlighting the ubiquity of pleiotropy. Recently, multivariate methods have been proposed to effectively identify pleiotropy. However, the statistical performance in natural biomedical data, which often have unbalanced case-control sample sizes, is largely known. In this work, we designed 21 scenarios of real-data informed simulations to thoroughly evaluate the statistical characteristics of univariate and multivariate methods. Our results can serve as a reference guide for the application of multivariate methods. We also investigated potential pleiotropy across type II diabetes, Alzheimer's disease, atherosclerosis of arteries, depression, and atherosclerotic heart disease in the UK Biobank.
Collapse
Affiliation(s)
- Xinyuan Zhang
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ruowang Li
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Marylyn D Ritchie
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
30
|
Dennis JK, Sealock JM, Straub P, Lee YH, Hucks D, Actkins K, Faucon A, Feng YCA, Ge T, Goleva SB, Niarchou M, Singh K, Morley T, Smoller JW, Ruderfer DM, Mosley JD, Chen G, Davis LK. Clinical laboratory test-wide association scan of polygenic scores identifies biomarkers of complex disease. Genome Med 2021; 13:6. [PMID: 33441150 PMCID: PMC7807864 DOI: 10.1186/s13073-020-00820-8] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 12/08/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Clinical laboratory (lab) tests are used in clinical practice to diagnose, treat, and monitor disease conditions. Test results are stored in electronic health records (EHRs), and a growing number of EHRs are linked to patient DNA, offering unprecedented opportunities to query relationships between genetic risk for complex disease and quantitative physiological measurements collected on large populations. METHODS A total of 3075 quantitative lab tests were extracted from Vanderbilt University Medical Center's (VUMC) EHR system and cleaned for population-level analysis according to our QualityLab protocol. Lab values extracted from BioVU were compared with previous population studies using heritability and genetic correlation analyses. We then tested the hypothesis that polygenic risk scores for biomarkers and complex disease are associated with biomarkers of disease extracted from the EHR. In a proof of concept analyses, we focused on lipids and coronary artery disease (CAD). We cleaned lab traits extracted from the EHR performed lab-wide association scans (LabWAS) of the lipids and CAD polygenic risk scores across 315 heritable lab tests then replicated the pipeline and analyses in the Massachusetts General Brigham Biobank. RESULTS Heritability estimates of lipid values (after cleaning with QualityLab) were comparable to previous reports and polygenic scores for lipids were strongly associated with their referent lipid in a LabWAS. LabWAS of the polygenic score for CAD recapitulated canonical heart disease biomarker profiles including decreased HDL, increased pre-medication LDL, triglycerides, blood glucose, and glycated hemoglobin (HgbA1C) in European and African descent populations. Notably, many of these associations remained even after adjusting for the presence of cardiovascular disease and were replicated in the MGBB. CONCLUSIONS Polygenic risk scores can be used to identify biomarkers of complex disease in large-scale EHR-based genomic analyses, providing new avenues for discovery of novel biomarkers and deeper understanding of disease trajectories in pre-symptomatic individuals. We present two methods and associated software, QualityLab and LabWAS, to clean and analyze EHR labs at scale and perform a Lab-Wide Association Scan.
Collapse
Affiliation(s)
- Jessica K Dennis
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Julia M Sealock
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Peter Straub
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Younga H Lee
- Psychiatric & Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Donald Hucks
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Ky'Era Actkins
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, TN, 37232, USA
| | - Annika Faucon
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Yen-Chen Anne Feng
- Psychiatric & Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Analytic and Translational Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Tian Ge
- Psychiatric & Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Slavina B Goleva
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Maria Niarchou
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Kritika Singh
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Theodore Morley
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Jordan W Smoller
- Psychiatric & Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Douglas M Ruderfer
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Departments of Medicine and Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Jonathan D Mosley
- Departments of Medicine and Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Guanhua Chen
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Lea K Davis
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Departments of Medicine and Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University, 511-A Light Hall, 2215 Garland Ave, Nashville, TN, 37232, USA.
| |
Collapse
|
31
|
Dueñas HR, Seah C, Johnson JS, Huckins LM. Implicit bias of encoded variables: frameworks for addressing structured bias in EHR-GWAS data. Hum Mol Genet 2020; 29:R33-R41. [PMID: 32879975 PMCID: PMC7530523 DOI: 10.1093/hmg/ddaa192] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/20/2022] Open
Abstract
The 'discovery' stage of genome-wide association studies required amassing large, homogeneous cohorts. In order to attain clinically useful insights, we must now consider the presentation of disease within our clinics and, by extension, within our medical records. Large-scale use of electronic health record (EHR) data can help to understand phenotypes in a scalable manner, incorporating lifelong and whole-phenome context. However, extending analyses to incorporate EHR and biobank-based analyses will require careful consideration of phenotype definition. Judgements and clinical decisions that occur 'outside' the system inevitably contain some degree of bias and become encoded in EHR data. Any algorithmic approach to phenotypic characterization that assumes non-biased variables will generate compounded biased conclusions. Here, we discuss and illustrate potential biases inherent within EHR analyses, how these may be compounded across time and suggest frameworks for large-scale phenotypic analysis to minimize and uncover encoded bias.
Collapse
Affiliation(s)
- Hillary R Dueñas
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carina Seah
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jessica S Johnson
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Laura M Huckins
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mental Illness Research, Education and Clinical Centers, James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY 10468, USA
| |
Collapse
|
32
|
Leung C, Rescan M, Grulois D, Chevin LM. Reduced phenotypic plasticity evolves in less predictable environments. Ecol Lett 2020; 23:1664-1672. [PMID: 32869431 PMCID: PMC7754491 DOI: 10.1111/ele.13598] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/22/2020] [Accepted: 08/06/2020] [Indexed: 01/16/2023]
Abstract
Phenotypic plasticity is a prominent mechanism for coping with variable environments, and a key determinant of extinction risk. Evolutionary theory predicts that phenotypic plasticity should evolve to lower levels in environments that fluctuate less predictably, because they induce mismatches between plastic responses and selective pressures. However, this prediction is difficult to test in nature, where environmental predictability is not controlled. Here, we exposed 32 lines of the halotolerant microalga Dunaliella salina to ecologically realistic, randomly fluctuating salinity, with varying levels of predictability, for 500 generations. We found that morphological plasticity evolved to lower degrees in lines that experienced less predictable environments. Evolution of plasticity mostly concerned phases with slow population growth, rather than the exponential phase where microbes are typically phenotyped. This study underlines that long‐term experiments with complex patterns of environmental change are needed to test theories about population responses to altered environmental predictability, as currently observed under climate change.
Collapse
Affiliation(s)
- Christelle Leung
- CEFE, Université de Montpellier, CNRS, EPHE, IRD, Université Paul Valéry Montpellier 3, Montpellier, France
| | - Marie Rescan
- CEFE, Université de Montpellier, CNRS, EPHE, IRD, Université Paul Valéry Montpellier 3, Montpellier, France
| | - Daphné Grulois
- CEFE, Université de Montpellier, CNRS, EPHE, IRD, Université Paul Valéry Montpellier 3, Montpellier, France
| | - Luis-Miguel Chevin
- CEFE, Université de Montpellier, CNRS, EPHE, IRD, Université Paul Valéry Montpellier 3, Montpellier, France
| |
Collapse
|
33
|
Zong N, Sharma DK, Yu Y, Egan JB, Davila JI, Wang C, Jiang G. Developing a FHIR-based Framework for Phenome Wide Association Studies: A Case Study with A Pan-Cancer Cohort. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE PROCEEDINGS. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE 2020; 2020:750-759. [PMID: 32477698 PMCID: PMC7233075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Phenome Wide Association Studies (PheWAS) enables phenome-wide scans to discover novel associations between genotype and clinical phenotypes via linking available genomic reports and large-scale Electronic Health Record (EHR). Data heterogeneity from different EHR systems and genetic reports has been a critical challenge that hinders meaningful validation. To address this, we propose an FHIR-based framework to model the PheWAS study in a standard manner. We developed an FHIR-based data model profile to enable the standard representation of data elements from genetic reports and EHR data that are used in the PheWAS study. As a proof-of-concept, we implemented the proposed method using a cohort of 1,595 pan-cancer patients with genetic reports from Foundation Medicine as well as the corresponding lab tests and diagnosis from Mayo EHRs. A PheWAS study is conducted and 81 significant genotype-phenotype associations are identified, in which 36 significant associations for cancers are validated based on a literature review.
Collapse
Affiliation(s)
- Nansu Zong
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Deepak K Sharma
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Yue Yu
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Jan B Egan
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Jaime I Davila
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Chen Wang
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Guoqian Jiang
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
34
|
Tang HHF, Sly PD, Holt PG, Holt KE, Inouye M. Systems biology and big data in asthma and allergy: recent discoveries and emerging challenges. Eur Respir J 2020; 55:13993003.00844-2019. [PMID: 31619470 DOI: 10.1183/13993003.00844-2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 09/12/2019] [Indexed: 12/15/2022]
Abstract
Asthma is a common condition caused by immune and respiratory dysfunction, and it is often linked to allergy. A systems perspective may prove helpful in unravelling the complexity of asthma and allergy. Our aim is to give an overview of systems biology approaches used in allergy and asthma research. Specifically, we describe recent "omic"-level findings, and examine how these findings have been systematically integrated to generate further insight.Current research suggests that allergy is driven by genetic and epigenetic factors, in concert with environmental factors such as microbiome and diet, leading to early-life disturbance in immunological development and disruption of balance within key immuno-inflammatory pathways. Variation in inherited susceptibility and exposures causes heterogeneity in manifestations of asthma and other allergic diseases. Machine learning approaches are being used to explore this heterogeneity, and to probe the pathophysiological patterns or "endotypes" that correlate with subphenotypes of asthma and allergy. Mathematical models are being built based on genomic, transcriptomic and proteomic data to predict or discriminate disease phenotypes, and to describe the biomolecular networks behind asthma.The use of systems biology in allergy and asthma research is rapidly growing, and has so far yielded fruitful results. However, the scale and multidisciplinary nature of this research means that it is accompanied by new challenges. Ultimately, it is hoped that systems medicine, with its integration of omics data into clinical practice, can pave the way to more precise, personalised and effective management of asthma.
Collapse
Affiliation(s)
- Howard H F Tang
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Australia .,Cambridge Baker Systems Genomics Initiative, Dept of Public Health and Primary Care, University of Cambridge, Cambridge, UK.,School of BioSciences, The University of Melbourne, Parkville, Australia
| | - Peter D Sly
- Queensland Children's Medical Research Institute, The University of Queensland, Brisbane, Australia.,Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Patrick G Holt
- Queensland Children's Medical Research Institute, The University of Queensland, Brisbane, Australia.,Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Kathryn E Holt
- Dept of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Australia.,London School of Hygiene and Tropical Medicine, London, UK
| | - Michael Inouye
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Australia.,Cambridge Baker Systems Genomics Initiative, Dept of Public Health and Primary Care, University of Cambridge, Cambridge, UK.,School of BioSciences, The University of Melbourne, Parkville, Australia.,The Alan Turing Institute, London, UK
| |
Collapse
|
35
|
Cai T, Cai TT, Liao K, Liu W. Large-Scale Simultaneous Testing of Cross-Covariance Matrices with Applications to PheWAS. Stat Sin 2020; 29:983-1005. [PMID: 31889766 DOI: 10.5705/ss.202017.0189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Motivated by applications in phenome-wide association studies (PheWAS), we consider in this paper simultaneous testing of columns of high-dimensional cross-covariance matrices and develop a multiple testing procedure with theoretical guarantees. It is shown that the proposed testing procedure maintains a desired false discovery rate (FDR) and false discovery proportion (FDP) under mild regularity conditions. We also provide results on the magnitudes of the signals that can be detected with high power. Simulation studies demonstrate that the proposed procedure can be substantially more powerful than existing FDR controlling procedures in the presence of correlation of unknown structure. The proposed multiple testing procedure is applied to a PheWAS of two auto-immune genetic markers using a rheumatoid arthritis patient cohort constructed from the electronic medical records of Partners Healthcare System.
Collapse
Affiliation(s)
- Tianxi Cai
- Department of Biostatistics, Harvard T.H Chan School of Public Health
| | - T Tony Cai
- Department of Statistics, The Wharton School, University of Pennsylvania
| | | | - Weidong Liu
- Department of Mathematics, Institute of Natural Sciences and MOE-LSC, Shanghai Jiao Tong University
| |
Collapse
|
36
|
Chhetri HB, Furches A, Macaya-Sanz D, Walker AR, Kainer D, Jones P, Harman-Ware AE, Tschaplinski TJ, Jacobson D, Tuskan GA, DiFazio SP. Genome-Wide Association Study of Wood Anatomical and Morphological Traits in Populus trichocarpa. FRONTIERS IN PLANT SCIENCE 2020; 11:545748. [PMID: 33013968 PMCID: PMC7509168 DOI: 10.3389/fpls.2020.545748] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 08/21/2020] [Indexed: 05/04/2023]
Abstract
To understand the genetic mechanisms underlying wood anatomical and morphological traits in Populus trichocarpa, we used 869 unrelated genotypes from a common garden in Clatskanie, Oregon that were previously collected from across the distribution range in western North America. Using GEMMA mixed model analysis, we tested for the association of 25 phenotypic traits and nine multitrait combinations with 6.741 million SNPs covering the entire genome. Broad-sense trait heritabilities ranged from 0.117 to 0.477. Most traits were significantly correlated with geoclimatic variables suggesting a role of climate and geography in shaping the variation of this species. Fifty-seven SNPs from single trait GWAS and 11 SNPs from multitrait GWAS passed an FDR threshold of 0.05, leading to the identification of eight and seven nearby candidate genes, respectively. The percentage of phenotypic variance explained (PVE) by the significant SNPs for both single and multitrait GWAS ranged from 0.01% to 6.18%. To further evaluate the potential roles of candidate genes, we used a multi-omic network containing five additional data sets, including leaf and wood metabolite GWAS layers and coexpression and comethylation networks. We also performed a functional enrichment analysis on coexpression nearest neighbors for each gene model identified by the wood anatomical and morphological trait GWAS analyses. Genes affecting cell wall composition and transport related genes were enriched in wood anatomy and stomatal density trait networks. Signaling and metabolism related genes were also common in networks for stomatal density. For leaf morphology traits (leaf dry and wet weight) the networks were significantly enriched for GO terms related to photosynthetic processes as well as cellular homeostasis. The identified genes provide further insights into the genetic control of these traits, which are important determinants of the suitability and sustainability of improved genotypes for lignocellulosic biofuel production.
Collapse
Affiliation(s)
- Hari B. Chhetri
- Department of Biology, West Virginia University, Morgantown, WV, United States
| | - Anna Furches
- Biosciences Division, and The Center for Bioenergy Innovation, Oak Ridge National Laboratory, Oak Ridge, TN, United States
- The Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee, Knoxville, TN, United States
| | - David Macaya-Sanz
- Department of Biology, West Virginia University, Morgantown, WV, United States
| | - Alejandro R. Walker
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, United States
| | - David Kainer
- Biosciences Division, and The Center for Bioenergy Innovation, Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Piet Jones
- Biosciences Division, and The Center for Bioenergy Innovation, Oak Ridge National Laboratory, Oak Ridge, TN, United States
- The Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee, Knoxville, TN, United States
| | - Anne E. Harman-Ware
- Biosciences Center, and National Bioenergy Center, National Renewable Energy Laboratory, Golden, CO, United States
| | - Timothy J. Tschaplinski
- Biosciences Division, and The Center for Bioenergy Innovation, Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Daniel Jacobson
- Biosciences Division, and The Center for Bioenergy Innovation, Oak Ridge National Laboratory, Oak Ridge, TN, United States
- The Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee, Knoxville, TN, United States
| | - Gerald A. Tuskan
- Biosciences Division, and The Center for Bioenergy Innovation, Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Stephen P. DiFazio
- Department of Biology, West Virginia University, Morgantown, WV, United States
- *Correspondence: Stephen P. DiFazio,
| |
Collapse
|
37
|
Systematic analysis of genes and diseases using PheWAS-Associated networks. Comput Biol Med 2019; 109:311-321. [PMID: 31128465 DOI: 10.1016/j.compbiomed.2019.04.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/28/2019] [Accepted: 04/28/2019] [Indexed: 02/08/2023]
|
38
|
Weighill D, Jones P, Bleker C, Ranjan P, Shah M, Zhao N, Martin M, DiFazio S, Macaya-Sanz D, Schmutz J, Sreedasyam A, Tschaplinski T, Tuskan G, Jacobson D. Multi-Phenotype Association Decomposition: Unraveling Complex Gene-Phenotype Relationships. Front Genet 2019; 10:417. [PMID: 31134130 PMCID: PMC6522845 DOI: 10.3389/fgene.2019.00417] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 04/16/2019] [Indexed: 01/18/2023] Open
Abstract
Various patterns of multi-phenotype associations (MPAs) exist in the results of Genome Wide Association Studies (GWAS) involving different topologies of single nucleotide polymorphism (SNP)-phenotype associations. These can provide interesting information about the different impacts of a gene on closely related phenotypes or disparate phenotypes (pleiotropy). In this work we present MPA Decomposition, a new network-based approach which decomposes the results of a multi-phenotype GWAS study into three bipartite networks, which, when used together, unravel the multi-phenotype signatures of genes on a genome-wide scale. The decomposition involves the construction of a phenotype powerset space, and subsequent mapping of genes into this new space. Clustering of genes in this powerset space groups genes based on their detailed MPA signatures. We show that this method allows us to find multiple different MPA and pleiotropic signatures within individual genes and to classify and cluster genes based on these SNP-phenotype association topologies. We demonstrate the use of this approach on a GWAS analysis of a large population of 882 Populus trichocarpa genotypes using untargeted metabolomics phenotypes. This method should prove invaluable in the interpretation of large GWAS datasets and aid in future synthetic biology efforts designed to optimize phenotypes of interest.
Collapse
Affiliation(s)
- Deborah Weighill
- The Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee, Knoxville, TN, United States.,Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Piet Jones
- The Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee, Knoxville, TN, United States.,Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Carissa Bleker
- The Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee, Knoxville, TN, United States.,Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Priya Ranjan
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, United States.,Department of Plant Sciences, The University of Tennessee Institute of Agriculture, University of Tennessee, Knoxville, TN, United States
| | - Manesh Shah
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Nan Zhao
- Department of Plant Sciences, The University of Tennessee Institute of Agriculture, University of Tennessee, Knoxville, TN, United States
| | - Madhavi Martin
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Stephen DiFazio
- Department of Biology, West Virginia University, Morgantown, WV, United States
| | - David Macaya-Sanz
- Department of Biology, West Virginia University, Morgantown, WV, United States
| | - Jeremy Schmutz
- Department of Energy Joint Genome Institute, Walnut Creek, CA, United States.,HudsonAlpha Institute for Biotechnology, Huntsville, AL, United States
| | | | - Timothy Tschaplinski
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Gerald Tuskan
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Daniel Jacobson
- The Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee, Knoxville, TN, United States.,Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, United States
| |
Collapse
|
39
|
Mir RR, Reynolds M, Pinto F, Khan MA, Bhat MA. High-throughput phenotyping for crop improvement in the genomics era. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2019; 282:60-72. [PMID: 31003612 DOI: 10.1016/j.plantsci.2019.01.007] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 12/10/2018] [Accepted: 01/09/2019] [Indexed: 05/24/2023]
Abstract
Tremendous progress has been made with continually expanding genomics technologies to unravel and understand crop genomes. However, the impact of genomics data on crop improvement is still far from satisfactory, in large part due to a lack of effective phenotypic data; our capacity to collect useful high quality phenotypic data lags behind the current capacity to generate high-throughput genomics data. Thus, the research bottleneck in plant sciences is shifting from genotyping to phenotyping. This article review the current status of efforts made in the last decade to systematically collect phenotypic data to alleviate this 'phenomics bottlenecks' by recording trait data through sophisticated non-invasive imaging, spectroscopy, image analysis, robotics, high-performance computing facilities and phenomics databases. These modern phenomics platforms and tools aim to record data on traits like plant development, architecture, plant photosynthesis, growth or biomass productivity, on hundreds to thousands of plants in a single day, as a phenomics revolution. It is believed that this revolution will provide plant scientists with the knowledge and tools necessary for unlocking information coded in plant genomes. Efforts have been also made to present the advances made in the last 10 years in phenomics platforms and their use in generating phenotypic data on different traits in several major crops including rice, wheat, barley, and maize. The article also highlights the need for phenomics databases and phenotypic data sharing for crop improvement. The phenomics data generated has been used to identify genes/QTL through QTL mapping, association mapping and genome-wide association studies (GWAS) for genomics-assisted breeding (GAB) for crop improvement.
Collapse
Affiliation(s)
- Reyazul Rouf Mir
- Division of Genetics & Plant Breeding, Faculty of Agriculture (FoA), Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Wadura Campus, Sopore-193201, Kashmir, India.
| | - Mathew Reynolds
- Global Wheat Program, International Maize and Wheat Improvement Centre (CIMMYT), Mexico, D.F., Mexico
| | - Francisco Pinto
- Global Wheat Program, International Maize and Wheat Improvement Centre (CIMMYT), Mexico, D.F., Mexico
| | - Mohd Anwar Khan
- Division of Genetics & Plant Breeding, Faculty of Agriculture (FoA), Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Wadura Campus, Sopore-193201, Kashmir, India
| | - Mohd Ashraf Bhat
- Division of Genetics & Plant Breeding, Faculty of Agriculture (FoA), Sher-e-Kashmir University of Agricultural Sciences and Technology of Kashmir (SKUAST-K), Wadura Campus, Sopore-193201, Kashmir, India
| |
Collapse
|
40
|
James G, Reisberg S, Lepik K, Galwey N, Avillach P, Kolberg L, Mägi R, Esko T, Alexander M, Waterworth D, Loomis AK, Vilo J. An exploratory phenome wide association study linking asthma and liver disease genetic variants to electronic health records from the Estonian Biobank. PLoS One 2019; 14:e0215026. [PMID: 30978214 PMCID: PMC6461350 DOI: 10.1371/journal.pone.0215026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/25/2019] [Indexed: 12/22/2022] Open
Abstract
The Estonian Biobank, governed by the Institute of Genomics at the University of Tartu (Biobank), has stored genetic material/DNA and continuously collected data since 2002 on a total of 52,274 individuals representing ~5% of the Estonian adult population and is increasing. To explore the utility of data available in the Biobank, we conducted a phenome-wide association study (PheWAS) in two areas of interest to healthcare researchers; asthma and liver disease. We used 11 asthma and 13 liver disease-associated single nucleotide polymorphisms (SNPs), identified from published genome-wide association studies, to test our ability to detect established associations. We confirmed 2 asthma and 5 liver disease associated variants at nominal significance and directionally consistent with published results. We found 2 associations that were opposite to what was published before (rs4374383:AA increases risk of NASH/NAFLD, rs11597086 increases ALT level). Three SNP-diagnosis pairs passed the phenome-wide significance threshold: rs9273349 and E06 (thyroiditis, p = 5.50x10-8); rs9273349 and E10 (type-1 diabetes, p = 2.60x10-7); and rs2281135 and K76 (non-alcoholic liver diseases, including NAFLD, p = 4.10x10-7). We have validated our approach and confirmed the quality of the data for these conditions. Importantly, we demonstrate that the extensive amount of genetic and medical information from the Estonian Biobank can be successfully utilized for scientific research.
Collapse
Affiliation(s)
- Glen James
- AstraZeneca, Global Medical Affairs, Cambridge, United Kingdom
| | - Sulev Reisberg
- Institute of Computer Science, University of Tartu, Tartu, Estonia
- STACC, Tartu, Estonia
- Quretec, Tartu, Estonia
| | - Kaido Lepik
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Nicholas Galwey
- GlaxoSmithKline, Research and Development, Stevenage, United Kingdom
| | - Paul Avillach
- Department of Biomedical Informatics, Harvard Medical School, Boston, United States of America
- Department of Medical Informatics, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Liis Kolberg
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Reedik Mägi
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Tõnu Esko
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Myriam Alexander
- GlaxoSmithKline, Research and Development, Stevenage, United Kingdom
| | - Dawn Waterworth
- GlaxoSmithKline, Genetics, Collegeville, PA, United States of America
| | - A. Katrina Loomis
- Pfizer Worldwide Research and Development, Groton, CT, United States of America
| | - Jaak Vilo
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| |
Collapse
|
41
|
Unlu G, Gamazon ER, Qi X, Levic DS, Bastarache L, Denny JC, Roden DM, Mayzus I, Breyer M, Zhong X, Konkashbaev AI, Rzhetsky A, Knapik EW, Cox NJ. GRIK5 Genetically Regulated Expression Associated with Eye and Vascular Phenomes: Discovery through Iteration among Biobanks, Electronic Health Records, and Zebrafish. Am J Hum Genet 2019; 104:503-519. [PMID: 30827500 PMCID: PMC6407495 DOI: 10.1016/j.ajhg.2019.01.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/29/2019] [Indexed: 12/15/2022] Open
Abstract
Although the use of model systems for studying the mechanism of mutations that have a large effect is common, we highlight here the ways that zebrafish-model-system studies of a gene, GRIK5, that contributes to the polygenic liability to develop eye diseases have helped to illuminate a mechanism that implicates vascular biology in eye disease. A gene-expression prediction derived from a reference transcriptome panel applied to BioVU, a large electronic health record (EHR)-linked biobank at Vanderbilt University Medical Center, implicated reduced GRIK5 expression in diverse eye diseases. We tested the function of GRIK5 by depletion of its ortholog in zebrafish, and we observed reduced blood vessel numbers and integrity in the eye and increased vascular permeability. Analyses of EHRs in >2.6 million Vanderbilt subjects revealed significant comorbidity of eye and vascular diseases (relative risks 2-15); this comorbidity was confirmed in 150 million individuals from a large insurance claims dataset. Subsequent studies in >60,000 genotyped BioVU participants confirmed the association of reduced genetically predicted expression of GRIK5 with comorbid vascular and eye diseases. Our studies pioneer an approach that allows a rapid iteration of the discovery of gene-phenotype relationships to the primary genetic mechanism contributing to the pathophysiology of human disease. Our findings also add dimension to the understanding of the biology driven by glutamate receptors such as GRIK5 (also referred to as GLUK5 in protein form) and to mechanisms contributing to human eye diseases.
Collapse
Affiliation(s)
- Gokhan Unlu
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Eric R Gamazon
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Data Science Institute, Vanderbilt University, Nashville, TN 37232, USA; Clare Hall, University of Cambridge, Cambridge CB3 9AL, UK
| | - Xinzi Qi
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Daniel S Levic
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Lisa Bastarache
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Departments of Medicine and Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Joshua C Denny
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Departments of Medicine and Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Dan M Roden
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Departments of Medicine and Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Ilya Mayzus
- Departments of Medicine and Human Genetics, the University of Chicago, Chicago, IL 60637, USA
| | - Max Breyer
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Xue Zhong
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Anuar I Konkashbaev
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Andrey Rzhetsky
- Departments of Medicine and Human Genetics, the University of Chicago, Chicago, IL 60637, USA
| | - Ela W Knapik
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Nancy J Cox
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Data Science Institute, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
42
|
Safarova MS, Satterfield BA, Fan X, Austin EE, Ye Z, Bastarache L, Zheng N, Ritchie MD, Borthwick KM, Williams MS, Larson EB, Scrol A, Jarvik GP, Crosslin DR, Leppig K, Rasmussen-Torvik LJ, Pendergrass SA, Sturm AC, Namjou B, Shah AS, Carroll RJ, Chung WK, Wei WQ, Feng Q, Stein CM, Roden DM, Manolio TA, Schaid DJ, Denny JC, Hebbring SJ, de Andrade M, Kullo IJ. A phenome-wide association study to discover pleiotropic effects of PCSK9, APOB, and LDLR. NPJ Genom Med 2019; 4:3. [PMID: 30774981 PMCID: PMC6370860 DOI: 10.1038/s41525-019-0078-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 01/16/2019] [Indexed: 01/09/2023] Open
Abstract
We conducted an electronic health record (EHR)-based phenome-wide association study (PheWAS) to discover pleiotropic effects of variants in three lipoprotein metabolism genes PCSK9, APOB, and LDLR. Using high-density genotype data, we tested the associations of variants in the three genes with 1232 EHR-derived binary phecodes in 51,700 European-ancestry (EA) individuals and 585 phecodes in 10,276 African-ancestry (AA) individuals; 457 PCSK9, 730 APOB, and 720 LDLR variants were filtered by imputation quality (r 2 > 0.4), minor allele frequency (>1%), linkage disequilibrium (r 2 < 0.3), and association with LDL-C levels, yielding a set of two PCSK9, three APOB, and five LDLR variants in EA but no variants in AA. Cases and controls were defined for each phecode using the PheWAS package in R. Logistic regression assuming an additive genetic model was used with adjustment for age, sex, and the first two principal components. Significant associations were tested in additional cohorts from Vanderbilt University (n = 29,713), the Marshfield Clinic Personalized Medicine Research Project (n = 9562), and UK Biobank (n = 408,455). We identified one PCSK9, two APOB, and two LDLR variants significantly associated with an examined phecode. Only one of the variants was associated with a non-lipid disease phecode, ("myopia") but this association was not significant in the replication cohorts. In this large-scale PheWAS we did not find LDL-C-related variants in PCSK9, APOB, and LDLR to be associated with non-lipid-related phenotypes including diabetes, neurocognitive disorders, or cataracts.
Collapse
Affiliation(s)
- Maya S. Safarova
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905 USA
| | | | - Xiao Fan
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905 USA
| | - Erin E. Austin
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905 USA
| | - Zhan Ye
- Biomedical Informatics Research Center, Marshfield Clinic Research Foundation, Marshfield, WI 54449 USA
| | - Lisa Bastarache
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN 37235 USA
| | - Neil Zheng
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN 37235 USA
| | - Marylyn D. Ritchie
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19111 USA
| | - Kenneth M. Borthwick
- Department of Biomedical and Translational Informatics, Geisinger, Danville, PA 17821 USA
| | | | | | - Aaron Scrol
- Group Health Research Institute, Seattle, WA 98101 USA
| | - Gail P. Jarvik
- Department of Medicine (Medical Genetics), University of Washington Medical Center, Seattle, WA 98195 USA
| | - David R. Crosslin
- Department of Medicine (Medical Genetics), University of Washington Medical Center, Seattle, WA 98195 USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195 USA
| | - Kathleen Leppig
- Genetic Services, Kaiser Permanente of Washington, Seattle, WA 98122 USA
| | - Laura J. Rasmussen-Torvik
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Sarah A. Pendergrass
- Department of Biomedical and Translational Informatics, Geisinger, Danville, PA 17821 USA
| | - Amy C. Sturm
- Genomic Medicine Institute, Geisinger, Danville, PA 17822 USA
| | - Bahram Namjou
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, and Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH 45229 USA
| | - Amy Sanghavi Shah
- Division of Endocrinology, Cincinnati Children’s Hospital Medical Center and University of Cincinnati, Cincinnati, OH 45229 USA
| | - Robert J. Carroll
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN 37235 USA
| | - Wendy K. Chung
- Department of Pediatrics, Columbia University, New York, NY 10032 USA
- Department of Medicine, Columbia University, New York, NY 10032 USA
| | - Wei-Qi Wei
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN 37235 USA
| | - QiPing Feng
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, TN 37232 USA
| | - C. Michael Stein
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, TN 37232 USA
| | - Dan M. Roden
- Department of Medicine, Vanderbilt University, Nashville, TN 37232 USA
| | - Teri A. Manolio
- Division of Genomic Medicine, National Human Genome Research Institute, Bethesda, MD 20892 USA
| | - Daniel J. Schaid
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905 USA
| | - Joshua C. Denny
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN 37235 USA
| | - Scott J. Hebbring
- Center for Human Genetics, Marshfield Clinic Research Foundation, Marshfield, WI 54449 USA
| | - Mariza de Andrade
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905 USA
| | - Iftikhar J. Kullo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905 USA
| |
Collapse
|
43
|
Pei G, Sun H, Dai Y, Liu X, Zhao Z, Jia P. Investigation of multi-trait associations using pathway-based analysis of GWAS summary statistics. BMC Genomics 2019; 20:79. [PMID: 30712509 PMCID: PMC6360716 DOI: 10.1186/s12864-018-5373-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background Genome-wide association studies (GWAS) have been successful in identifying disease-associated genetic variants. Recently, an increasing number of GWAS summary statistics have been made available to the research community, providing extensive repositories for studies of human complex diseases. In particular, cross-trait associations at the genetic level can be beneficial from large-scale GWAS summary statistics by using genetic variants that are associated with multiple traits. However, direct assessment of cross-trait associations using susceptibility loci has been challenging due to the complex genetic architectures in most diseases, calling for advantageous methods that could integrate functional interpretation and imply biological mechanisms. Results We developed an analytical framework for systematic integration of cross-trait associations. It incorporates two different approaches to detect enriched pathways and requires only summary statistics. We demonstrated the framework using 25 traits belonging to four phenotype groups. Our results revealed an average of 54 significantly associated pathways (ranged between 18 and 175) per trait. We further proved that pathway-based analysis provided increased power to estimate cross-trait associations compared to gene-level analysis. Based on Fisher’s Exact Test (FET), we identified a total of 24 (53) pairs of trait-trait association at adjusted pFET < 1 × 10− 3 (pFET < 0.01) among the 25 traits. Our trait-trait association network revealed not only many relationships among the traits within the same group but also novel relationships among traits from different groups, which warrants further investigation in future. Conclusions Our study revealed that risk variants for 25 different traits aggregated in particular biological pathways and that these pathways were frequently shared among traits. Our results confirmed known mechanisms and also suggested several novel insights into the etiology of multi-traits. Electronic supplementary material The online version of this article (10.1186/s12864-018-5373-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Guangsheng Pei
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin St. Suite 820, Houston, TX, 77030, USA
| | - Hua Sun
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin St. Suite 820, Houston, TX, 77030, USA
| | - Yulin Dai
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin St. Suite 820, Houston, TX, 77030, USA
| | - Xiaoming Liu
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin St. Suite 820, Houston, TX, 77030, USA. .,Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA. .,Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, 37203, USA.
| | - Peilin Jia
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin St. Suite 820, Houston, TX, 77030, USA.
| |
Collapse
|
44
|
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
|
45
|
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
|
46
|
Mochida K, Koda S, Inoue K, Hirayama T, Tanaka S, Nishii R, Melgani F. Computer vision-based phenotyping for improvement of plant productivity: a machine learning perspective. Gigascience 2019; 8:5232233. [PMID: 30520975 PMCID: PMC6312910 DOI: 10.1093/gigascience/giy153] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 09/06/2018] [Accepted: 11/24/2018] [Indexed: 11/29/2022] Open
Abstract
Employing computer vision to extract useful information from images and videos is becoming a key technique for identifying phenotypic changes in plants. Here, we review the emerging aspects of computer vision for automated plant phenotyping. Recent advances in image analysis empowered by machine learning-based techniques, including convolutional neural network-based modeling, have expanded their application to assist high-throughput plant phenotyping. Combinatorial use of multiple sensors to acquire various spectra has allowed us to noninvasively obtain a series of datasets, including those related to the development and physiological responses of plants throughout their life. Automated phenotyping platforms accelerate the elucidation of gene functions associated with traits in model plants under controlled conditions. Remote sensing techniques with image collection platforms, such as unmanned vehicles and tractors, are also emerging for large-scale field phenotyping for crop breeding and precision agriculture. Computer vision-based phenotyping will play significant roles in both the nowcasting and forecasting of plant traits through modeling of genotype/phenotype relationships.
Collapse
Affiliation(s)
- Keiichi Mochida
- Bioproductivity Informatics Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
- Microalgae Production Control Technology Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science, Technology and Innovation Hub, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
- Institute of Plant Science and Resources, Okayama University, 2-20-1 Chuo, Kurashiki, Okayama 710-0046, Japan
- Kihara Institute for Biological Research, Yokohama City University, 641-12 Maioka-cho, Totsuka-ku, Yokohama, Kanagawa 244–0813, Japan
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan
| | - Satoru Koda
- Graduate School of Mathematics, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Komaki Inoue
- Bioproductivity Informatics Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Takashi Hirayama
- Institute of Plant Science and Resources, Okayama University, 2-20-1 Chuo, Kurashiki, Okayama 710-0046, Japan
| | - Shojiro Tanaka
- Hiroshima University of Economics, 5-37-1, Gion, Asaminami, Hiroshima-shi Hiroshima 731-0138, Japan
| | - Ryuei Nishii
- Institute of Mathematics for Industry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Farid Melgani
- Department of Information Engineering and Computer Science, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| |
Collapse
|
47
|
Mochida K, Koda S, Inoue K, Hirayama T, Tanaka S, Nishii R, Melgani F. Computer vision-based phenotyping for improvement of plant productivity: a machine learning perspective. Gigascience 2019. [PMID: 30520975 DOI: 10.1093/gigascience/giy153/5232233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2023] Open
Abstract
Employing computer vision to extract useful information from images and videos is becoming a key technique for identifying phenotypic changes in plants. Here, we review the emerging aspects of computer vision for automated plant phenotyping. Recent advances in image analysis empowered by machine learning-based techniques, including convolutional neural network-based modeling, have expanded their application to assist high-throughput plant phenotyping. Combinatorial use of multiple sensors to acquire various spectra has allowed us to noninvasively obtain a series of datasets, including those related to the development and physiological responses of plants throughout their life. Automated phenotyping platforms accelerate the elucidation of gene functions associated with traits in model plants under controlled conditions. Remote sensing techniques with image collection platforms, such as unmanned vehicles and tractors, are also emerging for large-scale field phenotyping for crop breeding and precision agriculture. Computer vision-based phenotyping will play significant roles in both the nowcasting and forecasting of plant traits through modeling of genotype/phenotype relationships.
Collapse
Affiliation(s)
- Keiichi Mochida
- Bioproductivity Informatics Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
- Microalgae Production Control Technology Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science, Technology and Innovation Hub, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
- Institute of Plant Science and Resources, Okayama University, 2-20-1 Chuo, Kurashiki, Okayama 710-0046, Japan
- Kihara Institute for Biological Research, Yokohama City University, 641-12 Maioka-cho, Totsuka-ku, Yokohama, Kanagawa 244-0813, Japan
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama, Kanagawa 236-0027, Japan
| | - Satoru Koda
- Graduate School of Mathematics, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Komaki Inoue
- Bioproductivity Informatics Research Team, RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Takashi Hirayama
- Institute of Plant Science and Resources, Okayama University, 2-20-1 Chuo, Kurashiki, Okayama 710-0046, Japan
| | - Shojiro Tanaka
- Hiroshima University of Economics, 5-37-1, Gion, Asaminami, Hiroshima-shi Hiroshima 731-0138, Japan
| | - Ryuei Nishii
- Institute of Mathematics for Industry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Farid Melgani
- Department of Information Engineering and Computer Science, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| |
Collapse
|
48
|
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
|
49
|
Zhang X, Veturi Y, Verma S, Bone W, Verma A, Lucas A, Hebbring S, Denny JC, Stanaway IB, Jarvik GP, Crosslin D, Larson EB, Rasmussen-Torvik L, Pendergrass SA, Smoller JW, Hakonarson H, Sleiman P, Weng C, Fasel D, Wei WQ, Kullo I, Schaid D, Chung WK, Ritchie MD. Detecting potential pleiotropy across cardiovascular and neurological diseases using univariate, bivariate, and multivariate methods on 43,870 individuals from the eMERGE network. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2019; 24:272-283. [PMID: 30864329 PMCID: PMC6457436] [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: 11/05/2022]
Abstract
The link between cardiovascular diseases and neurological disorders has been widely observed in the aging population. Disease prevention and treatment rely on understanding the potential genetic nexus of multiple diseases in these categories. In this study, we were interested in detecting pleiotropy, or the phenomenon in which a genetic variant influences more than one phenotype. Marker-phenotype association approaches can be grouped into univariate, bivariate, and multivariate categories based on the number of phenotypes considered at one time. Here we applied one statistical method per category followed by an eQTL colocalization analysis to identify potential pleiotropic variants that contribute to the link between cardiovascular and neurological diseases. We performed our analyses on ~530,000 common SNPs coupled with 65 electronic health record (EHR)-based phenotypes in 43,870 unrelated European adults from the Electronic Medical Records and Genomics (eMERGE) network. There were 31 variants identified by all three methods that showed significant associations across late onset cardiac- and neurologic- diseases. We further investigated functional implications of gene expression on the detected "lead SNPs" via colocalization analysis, providing a deeper understanding of the discovered associations. In summary, we present the framework and landscape for detecting potential pleiotropy using univariate, bivariate, multivariate, and colocalization methods. Further exploration of these potentially pleiotropic genetic variants will work toward understanding disease causing mechanisms across cardiovascular and neurological diseases and may assist in considering disease prevention as well as drug repositioning in future research.
Collapse
Affiliation(s)
- Xinyuan Zhang
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA*Authors contributed equally to this work
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
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
|