1
|
Wu C, Hu Z, Zhang P. Association of the platelet-to-high-density lipoprotein cholesterol ratio (PHR) with metabolic syndrome and metabolic overweight/obesity phenotypes: A study based on the Dryad database. PLoS One 2025; 20:e0321625. [PMID: 40327659 PMCID: PMC12054858 DOI: 10.1371/journal.pone.0321625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/10/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Overweight/obesity and metabolic syndrome (MetS) are major global public health challenges. The platelet-to-high-density lipoprotein cholesterol ratio (PHR) has emerged as a potential biomarker reflecting both inflammatory status and lipid metabolism; however, its association with MetS and metabolic overweight/obesity phenotypes is not well understood. The present study aims to investigate the association of the PHR with MetS and metabolic overweight/obesity phenotypes. METHODS We derived data from the Dryad data repository. This retrospective study included 1,592 physical examination participants in Wuhan Union Hospital from 2020 to 2021. Based on BMI categories and metabolic status, we defined and distinguished four metabolic overweight/obesity phenotypes: metabolically healthy with normal weight (MHNW), metabolically unhealthy with normal weight (MUNW), metabolically healthy with overweight/obesity (MHO), and metabolically unhealthy with overweight/obesity (MUO). PHR was calculated as the ratio of platelets to HDL cholesterol. Logistic regression analysis was used to test the independent association between PHR with MetS and metabolic overweight/obesity phenotypes. In addition, we performed smooth curve fitting and subgroup analyses. RESULTS Logistic regression analysis, after adjusting for covariates, indicated that each 10-unit increase in PHR was associated with elevated risks of MetS (OR = 1.29, 95% CI: 1.24-1.35). There was a significant positive association between PHR and the occurrence of both MHO (OR = 1.15, 95% CI: 1.11-1.19) and MUO (OR = 1.16, 95% CI: 1.12-1.20), while the association with MUNW remained unclear (P = 0.73). In addition, we found a nonlinear relationship between PHR and the incidence of MetS, MHO, and MUO. CONCLUSION PHR demonstrates strong associations with MetS, MHO, and MUO, indicating its potential utility as an early biomarker for metabolic dysfunction.
Collapse
Affiliation(s)
- Chuncheng Wu
- The Second Department of Digestion, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Ziyi Hu
- Classical Department of Traditional Chinese Medicine, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Ping Zhang
- The Second Department of Digestion, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| |
Collapse
|
2
|
Halme A, Salminen A, Suominen A, Havulinna A, Mäntylä P, Buhlin K, Paju S, Männistö S, Salomaa V, Sattler W, Sinisalo J, Pussinen P. Metabolic Profiling of Individuals with Missing Teeth and Tooth Loss. J Dent Res 2025; 104:389-397. [PMID: 39754308 PMCID: PMC11909773 DOI: 10.1177/00220345241298219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Missing teeth have been linked to incident cardiovascular disease, diabetes, and all-cause mortality. Our previous study revealed that signs of oral infections and inflammatory conditions (i.e., periodontal disease and dental caries) are associated with disadvantageous features of circulating metabolites. This study investigates whether missing teeth and tooth loss, the end points of these diseases, are associated with similar metabolic features. The 2 Finnish population-based studies Health-2000 (n = 6,197) and FINRISK-97 (n = 6,050) were included, as was Parogene (n = 465), a cohort of patients with an indication for coronary angiography. The number of teeth was recorded in clinical examinations. Serum concentrations of 157 metabolites were determined by a nuclear magnetic resonance spectroscopy-based method. Health-2000 participants (n = 3,371) provided follow-up serum samples, and 1,186 of them participated in a repeated oral examination 11 y after the baseline. Linear regression models adjusted for age, sex, smoking, body mass index, and diabetes were fitted to the number of teeth and metabolite measures. The results from the separate cohorts were combined in a fixed-effects meta-analysis. We also analyzed whether the number of teeth at baseline and tooth loss during follow-up were associated with changes in metabolite concentrations. Missing teeth were associated with increased very-low-density lipoprotein-related measures and triglyceride concentrations, as well as with decreased high-density lipoprotein parameters and small particle size. Missing teeth also had an association with low levels of unsaturated fatty acids (FAs), including omega-3 and omega-6 FAs, and elevated proportions of monounsaturated and saturated FAs. The number of teeth at baseline predicted changes in several concentrations, such as measures related to intermediate-density lipoprotein, low-density lipoprotein, and FAs, but no associations with tooth loss during the 11-y follow-up were observed. To conclude, missing teeth are associated with adverse metabolic features characterized by systemic inflammation and several risk factors for cardiometabolic diseases.
Collapse
Affiliation(s)
- A.M. Halme
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - A. Salminen
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - A.L. Suominen
- Institute of Dentistry, University of Eastern Finland, Kuopio, Finland
- Oral and Maxillofacial Teaching Unit, Kuopio University Hospital, Kuopio, Finland
- National Institute for Health and Welfare, Helsinki, Finland
| | - A. Havulinna
- National Institute for Health and Welfare, Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - P. Mäntylä
- Institute of Dentistry, University of Eastern Finland, Kuopio, Finland
- Oral and Maxillofacial Teaching Unit, Kuopio University Hospital, Kuopio, Finland
| | - K. Buhlin
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - S. Paju
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - S. Männistö
- National Institute for Health and Welfare, Helsinki, Finland
| | - V. Salomaa
- National Institute for Health and Welfare, Helsinki, Finland
| | - W. Sattler
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - J. Sinisalo
- HUCH Heart and Lung Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - P.J. Pussinen
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Institute of Dentistry, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
3
|
Orchard P, Blackwell TW, Kachuri L, Castaldi PJ, Cho MH, Christenson SA, Durda P, Gabriel S, Hersh CP, Huntsman S, Hwang S, Joehanes R, Johnson M, Li X, Lin H, Liu CT, Liu Y, Mak ACY, Manichaikul AW, Paik D, Saferali A, Smith JD, Taylor KD, Tracy RP, Wang J, Wang M, Weinstock JS, Weiss J, Wheeler HE, Zhou Y, Zoellner S, Wu JC, Mestroni L, Graw S, Taylor MRG, Ortega VE, Johnson CW, Gan W, Abecasis G, Nickerson DA, Gupta N, Ardlie K, Woodruff PG, Zheng Y, Bowler RP, Meyers DA, Reiner A, Kooperberg C, Ziv E, Ramachandran VS, Larson MG, Cupples LA, Burchard EG, Silverman EK, Rich SS, Heard-Costa N, Tang H, Rotter JI, Smith AV, Levy D, Aguet F, Scott L, Raffield LM, Parker SCJ. Cross-cohort analysis of expression and splicing quantitative trait loci in TOPMed. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.19.25322561. [PMID: 40034763 PMCID: PMC11875316 DOI: 10.1101/2025.02.19.25322561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Most genetic variants associated with complex traits and diseases occur in non-coding genomic regions and are hypothesized to regulate gene expression. To understand the genetics underlying gene expression variability, we characterize 14,324 ancestrally diverse RNA-sequencing samples from the NHLBI Trans-Omics for Precision Medicine (TOPMed) program and integrate whole genome sequencing data to perform cis and trans expression and splicing quantitative trait locus (cis-/trans-e/sQTL) analyses in six tissues and cell types, most notably whole blood (N=6,454) and lung (N=1,291). We show this dataset enables greater detection of secondary cis-e/sQTL signals than was achieved in previous studies, and that secondary cis-eQTL and primary trans-eQTL signal discovery is not saturated even though eGene discovery is. Most TOPMed trans-eQTL signals colocalize with cis-e/sQTL signals, suggesting many trans signals are mediated by cis signals. We fine-map European UK BioBank GWAS signals from 164 traits and colocalize the resulting 34,107 fine-mapped GWAS signals with TOPMed e/sQTL signals, finding that of 10,611 GWAS signals with a colocalization, 7,096 GWAS signals colocalize with at least one secondary e/sQTL signal. These results demonstrate that larger e/sQTL analyses will continue to uncover secondary e/sQTL signals, and that these new signals will benefit GWAS interpretation.
Collapse
Affiliation(s)
- Peter Orchard
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Thomas W Blackwell
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Linda Kachuri
- Department of Epidemiology and Population Health, School of Medicine, Stanford University, Stanford, CA, USA
| | - Peter J Castaldi
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephanie A Christenson
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Peter Durda
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | | | - Craig P Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Scott Huntsman
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Seungyong Hwang
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Roby Joehanes
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Framingham Heart Study, National Heart, Lung, and Blood Institute, National Institutes of Health and Boston University, Framingham, MA, USA
| | - Mari Johnson
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Xingnan Li
- Division of Data Driven and Digital Medicine, Department of Medicine, Icahn School of Medicine at Mount Sanai, New York, NY, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Icahn School of Medicine at Mount Sanai, New York, NY, USA
| | - Honghuang Lin
- Framingham Heart Study, National Heart, Lung, and Blood Institute, National Institutes of Health and Boston University, Framingham, MA, USA
- Department of Medicine, University of Massachusetts Chan Medical School, University of Massachusetts, Worcester, MA, USA
| | - Ching-Ti Liu
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Yongmei Liu
- Department of Medicine, School of Medicine, Duke University, Durham, NC, USA
| | - Angel C Y Mak
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Ani W Manichaikul
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - David Paik
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Aabida Saferali
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joshua D Smith
- Northwest Genomics Center, University of Washington, Seattle, WA, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Russell P Tracy
- Laboratory for Clinical Biochemistry Research, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Jiongming Wang
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Mingqiang Wang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Joshua S Weinstock
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jeffrey Weiss
- Northwest Genomics Center, University of Washington, Seattle, WA, USA
| | - Heather E Wheeler
- Program in Bioinformatics, Loyola University Chicago, Chicago, IL, USA
- Department of Biology, Loyola University Chicago, Chicago, IL, USA
| | - Ying Zhou
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Sebastian Zoellner
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Luisa Mestroni
- Department of Medicine, Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sharon Graw
- Department of Medicine, Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Matthew R G Taylor
- Department of Medicine, Adult Medical Genetics Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Victor E Ortega
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Phoenix, AZ, USA
| | - Craig W Johnson
- Collaborative Health Studies Coordinating Center, University of Washington, Seattle, WA, USA
| | - Weiniu Gan
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Goncalo Abecasis
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Deborah A Nickerson
- Northwest Genomics Center, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Namrata Gupta
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | | | - Prescott G Woodruff
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, School of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Yinan Zheng
- Department of Preventive Medicine, Northwestern University, Chicago, IL, USA
| | - Russell P Bowler
- Department of Genomic Sciences and Systems Biology, Cleveland Clinic, Cleveland, OH, USA
| | - Deborah A Meyers
- Department of Medicine, Division of Genetics, Genomics and Precision Medicine, University of Arizona, Tucson, AZ, USA
| | - Alex Reiner
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Elad Ziv
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Vasan S Ramachandran
- Framingham Heart Study, National Heart, Lung, and Blood Institute and Boston University, Framingham, MA, USA
- Department of Medicine, School of Medicine, Boston University, Boston, MA, USA
| | - Martin G Larson
- Framingham Heart Study, National Heart, Lung, and Blood Institute and Boston University, Framingham, MA, USA
| | - L Adrienne Cupples
- Framingham Heart Study, National Heart, Lung, and Blood Institute and Boston University, Framingham, MA, USA
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Esteban G Burchard
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephen S Rich
- Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Nancy Heard-Costa
- Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Hua Tang
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Albert V Smith
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Daniel Levy
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Framingham Heart Study, National Heart, Lung, and Blood Institute, National Institutes of Health and Boston University, Framingham, MA, USA
| | - François Aguet
- Illumina Artificial Intelligence Laboratory, Illumina, Foster City, CA, USA
| | - Laura Scott
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stephen C J Parker
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
4
|
Cheng S, Li Y, Sun X, Liu Z, Guo L, Wu J, Yang X, Wei S, Wu G, Xu S, Yang F, Wu J. The impact of glucose metabolism on inflammatory processes in sepsis-induced acute lung injury. Front Immunol 2024; 15:1508985. [PMID: 39712019 PMCID: PMC11659153 DOI: 10.3389/fimmu.2024.1508985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/18/2024] [Indexed: 12/24/2024] Open
Abstract
Acute lung injury (ALI) is a prevalent and critical complication of sepsis, marked by high incidence and mortality rates, with its pathogenesis still not being fully elucidated. Recent research has revealed a significant correlation between the metabolic reprogramming of glucose and sepsis-associated ALI (S-ALI). Throughout the course of S-ALI, immune cells, including macrophages and dendritic cells, undergo metabolic shifts to accommodate the intricate demands of immune function that emerge as sepsis advances. Indeed, glucose metabolic reprogramming in S-ALI serves as a double-edged sword, fueling inflammatory immune responses in the initial stages and subsequently initiating anti-inflammatory responses as the disease evolves. In this review, we delineate the current research progress concerning the pathogenic mechanisms linked to glucose metabolic reprogramming in S-ALI, with a focus on the pertinent immune cells implicated. We encapsulate the impact of glucose metabolic reprogramming on the onset, progression, and prognosis of S-ALI. Ultimately, by examining key regulatory factors within metabolic intermediates and enzymes, We have identified potential therapeutic targets linked to metabolic reprogramming, striving to tackle the inherent challenges in diagnosing and treating Severe Acute Lung Injury (S-ALI) with greater efficacy.
Collapse
Affiliation(s)
- Shilei Cheng
- School of Anesthesiology, Shandong Second Medical University, Weifang, China
| | - Yufei Li
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
- School of Pharmacy, Shandong University of Traditional Chinese Medicine (TCM), Jinan, China
| | - Xiaoliang Sun
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhirui Liu
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Liang Guo
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
| | - Jueheng Wu
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Xiaohan Yang
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Sisi Wei
- Department of Anesthesiology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, China
| | - Guanghan Wu
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
| | - Shilong Xu
- School of Anesthesiology, Shandong Second Medical University, Weifang, China
| | - Fan Yang
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
| | - Jianbo Wu
- Department of Anesthesiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Institute of Anesthesia and Respiratory Critical Medicine, Jinan, China
- Shandong Provincial Clinical Research Center for Anesthesiology, Jinan, China
| |
Collapse
|
5
|
Cote AC, Young HE, Huckins LM. Critical reasoning on the co-expression module QTL in the dorsolateral prefrontal cortex. HGG ADVANCES 2024; 5:100311. [PMID: 38773772 PMCID: PMC11214266 DOI: 10.1016/j.xhgg.2024.100311] [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: 10/23/2023] [Revised: 05/16/2024] [Accepted: 05/16/2024] [Indexed: 05/24/2024] Open
Abstract
Expression quantitative trait locus (eQTL) analysis is a popular method of gaining insight into the function of regulatory variation. While cis-eQTL resources have been instrumental in linking genome-wide association study variants to gene function, complex trait heritability may be additionally mediated by other forms of gene regulation. Toward this end, novel eQTL methods leverage gene co-expression (module-QTL) to investigate joint regulation of gene modules by single genetic variants. Here we broadly define a "module-QTL" as the association of a genetic variant with a summary measure of gene co-expression. This approach aims to reduce the multiple testing burden of a trans-eQTL search through the consolidation of gene-based testing and provide biological context to eQTLs shared between genes. In this article we provide an in-depth examination of the co-expression module eQTL (module-QTL) through literature review, theoretical investigation, and real-data application of the module-QTL to three large prefrontal cortex genotype-RNA sequencing datasets. We find module-QTLs in our study that are disease associated and reproducible are not additionally informative beyond cis- or trans-eQTLs for module genes. Through comparison to prior studies, we highlight promises and limitations of the module-QTL across study designs and provide recommendations for further investigation of the module-QTL framework.
Collapse
Affiliation(s)
- Alanna C Cote
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Hannah E Young
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Laura M Huckins
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA.
| |
Collapse
|
6
|
Salminen A, Määttä A, Mäntylä P, Leskelä J, Pietiäinen M, Buhlin K, Suominen A, Paju S, Sattler W, Sinisalo J, Pussinen P. Systemic Metabolic Signatures of Oral Diseases. J Dent Res 2024; 103:13-21. [PMID: 37968796 PMCID: PMC10734208 DOI: 10.1177/00220345231203562] [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] [Indexed: 11/17/2023] Open
Abstract
Systemic metabolic signatures of oral diseases have been rarely investigated, and prospective studies do not exist. We analyzed whether signs of current or past infectious/inflammatory oral diseases are associated with circulating metabolites. Two study populations were included: the population-based Health-2000 (n = 6,229) and Parogene (n = 452), a cohort of patients with an indication to coronary angiography. Health-2000 participants (n = 4,116) provided follow-up serum samples 11 y after the baseline. Serum concentrations of 157 metabolites were determined with a nuclear magnetic resonance spectroscopy-based method. The associations between oral parameters and metabolite concentrations were analyzed using linear regression models adjusted for age, sex, number of teeth, smoking, presence of diabetes, and education (in Health-2000 only). The number of decayed teeth presented positive associations with low-density lipoprotein diameter and the concentrations of pyruvate and citrate. Negative associations were found between caries and the unsaturation degree of fatty acids (FA) and relative proportions of docosahexaenoic and omega-3 FAs. The number of root canal fillings was positively associated with very low-density lipoprotein parameters, such as diameter, cholesterol, triglycerides, and number of particles. Deepened periodontal pockets were positively associated with concentrations of cholesterol, triglycerides, pyruvate, leucine, valine, phenylalanine, and glycoprotein acetyls and negatively associated with high-density lipoprotein (HDL) diameter, FA unsaturation degree, and relative proportions of omega-6 and polyunsaturated FAs. Bleeding on probing (BOP) was associated with increased concentrations of triglycerides and glycoprotein acetyls, as well as decreased proportions of omega-3 and omega-6 FAs. Caries at baseline predicted alterations in apolipoprotein B-containing lipoproteins and HDL-related metabolites in the follow-up, and both caries and BOP were associated with changes in HDL-related metabolites and omega-3 FAs in the follow-up. Signs of current or past infectious/inflammatory oral diseases, especially periodontitis, were associated with metabolic profiles typical for inflammation. Oral diseases may represent a modifiable risk factor for systemic chronic inflammation and thus cardiometabolic disorders.
Collapse
Affiliation(s)
- A. Salminen
- Oral and Maxillofacial diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - A.M. Määttä
- Oral and Maxillofacial diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - P. Mäntylä
- Institute of Dentistry, University of Eastern Finland, Kuopio, Finland
- Odontology Education, Kuopio University Hospital, Kuopio, Finland
| | - J. Leskelä
- Oral and Maxillofacial diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - M. Pietiäinen
- Oral and Maxillofacial diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - K. Buhlin
- Oral and Maxillofacial diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - A.L. Suominen
- Institute of Dentistry, University of Eastern Finland, Kuopio, Finland
- Odontology Education, Kuopio University Hospital, Kuopio, Finland
- Department of Public Health and Welfare, National Institute for Health and Welfare, Helsinki, Finland
| | - S. Paju
- Oral and Maxillofacial diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - W. Sattler
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - J. Sinisalo
- HUCH Heart and Lung Center, Helsinki University Hospital and University of Helsinki, Finland
| | - P.J. Pussinen
- Oral and Maxillofacial diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Institute of Dentistry, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
7
|
Jain PR, Yates M, de Celis CR, Drineas P, Jahanshad N, Thompson P, Paschou P. Multiomic approach and Mendelian randomization analysis identify causal associations between blood biomarkers and subcortical brain structure volumes. Neuroimage 2023; 284:120466. [PMID: 37995919 PMCID: PMC11647565 DOI: 10.1016/j.neuroimage.2023.120466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/17/2023] [Accepted: 11/20/2023] [Indexed: 11/25/2023] Open
Abstract
Alterations in subcortical brain structure volumes have been found to be associated with several neurodegenerative and psychiatric disorders. At the same time, genome-wide association studies (GWAS) have identified numerous common variants associated with brain structure. In this study, we integrate these findings, aiming to identify proteins, metabolites, or microbes that have a putative causal association with subcortical brain structure volumes via a two-sample Mendelian randomization approach. This method uses genetic variants as instrument variables to identify potentially causal associations between an exposure and an outcome. The exposure data that we analyzed comprised genetic associations for 2994 plasma proteins, 237 metabolites, and 103 microbial genera. The outcome data included GWAS data for seven subcortical brain structure volumes including accumbens, amygdala, caudate, hippocampus, pallidum, putamen, and thalamus. Eleven proteins and six metabolites were found to have a significant association with subcortical structure volumes, with nine proteins and five metabolites replicated using independent exposure data. We found causal associations between accumbens volume and plasma protease c1 inhibitor as well as strong association between putamen volume and Agouti signaling protein. Among metabolites, urate had the strongest association with thalamic volume. No significant associations were detected between the microbial genera and subcortical brain structure volumes. We also observed significant enrichment for biological processes such as proteolysis, regulation of the endoplasmic reticulum apoptotic signaling pathway, and negative regulation of DNA binding. Our findings provide insights to the mechanisms through which brain volumes may be affected in the pathogenesis of neurodevelopmental and psychiatric disorders and point to potential treatment targets for disorders that are associated with subcortical brain structure volumes.
Collapse
Affiliation(s)
- Pritesh R Jain
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, United States
| | - Madison Yates
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, United States
| | - Carlos Rubin de Celis
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, United States
| | - Petros Drineas
- Department of Computer Science, Purdue University, United States
| | - Neda Jahanshad
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of South California, United States
| | - Paul Thompson
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of South California, United States
| | - Peristera Paschou
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|
8
|
Mozhui K, Kim H, Villani F, Haghani A, Sen S, Horvath S. Pleiotropic influence of DNA methylation QTLs on physiological and ageing traits. Epigenetics 2023; 18:2252631. [PMID: 37691384 PMCID: PMC10496549 DOI: 10.1080/15592294.2023.2252631] [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: 05/01/2023] [Revised: 07/31/2023] [Accepted: 08/16/2023] [Indexed: 09/12/2023] Open
Abstract
DNA methylation is influenced by genetic and non-genetic factors. Here, we chart quantitative trait loci (QTLs) that modulate levels of methylation at highly conserved CpGs using liver methylome data from mouse strains belonging to the BXD family. A regulatory hotspot on chromosome 5 had the highest density of trans-acting methylation QTLs (trans-meQTLs) associated with multiple distant CpGs. We refer to this locus as meQTL.5a. Trans-modulated CpGs showed age-dependent changes and were enriched in developmental genes, including several members of the MODY pathway (maturity onset diabetes of the young). The joint modulation by genotype and ageing resulted in a more 'aged methylome' for BXD strains that inherited the DBA/2J parental allele at meQTL.5a. Further, several gene expression traits, body weight, and lipid levels mapped to meQTL.5a, and there was a modest linkage with lifespan. DNA binding motif and protein-protein interaction enrichment analyses identified the hepatic nuclear factor, Hnf1a (MODY3 gene in humans), as a strong candidate. The pleiotropic effects of meQTL.5a could contribute to variations in body size and metabolic traits, and influence CpG methylation and epigenetic ageing that could have an impact on lifespan.
Collapse
Affiliation(s)
- Khyobeni Mozhui
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Hyeonju Kim
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Flavia Villani
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Amin Haghani
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Altos Labs, San Diego, CA, USA
| | - Saunak Sen
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Altos Labs, San Diego, CA, USA
- Department of Biostatistics, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
9
|
Beuchel C, Dittrich J, Becker S, Kirsten H, Tönjes A, Kovacs P, Stumvoll M, Loeffler M, Teren A, Thiery J, Isermann B, Ceglarek U, Scholz M. An atlas of genome-wide gene expression and metabolite associations and possible mediation effects towards body mass index. J Mol Med (Berl) 2023; 101:1305-1321. [PMID: 37672078 PMCID: PMC10560167 DOI: 10.1007/s00109-023-02362-z] [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: 11/29/2022] [Revised: 08/07/2023] [Accepted: 08/15/2023] [Indexed: 09/07/2023]
Abstract
Investigating the cross talk of different omics layers is crucial to understand molecular pathomechanisms of metabolic diseases like obesity. Here, we present a large-scale association meta-analysis of genome-wide whole blood and peripheral blood mononuclear cell (PBMC) gene expressions profiled with Illumina HT12v4 microarrays and metabolite measurements from dried blood spots (DBS) characterized by targeted liquid chromatography tandem mass spectrometry (LC-MS/MS) in three large German cohort studies with up to 7706 samples. We found 37,295 associations comprising 72 amino acids (AA) and acylcarnitine (AC) metabolites (including ratios) and 8579 transcripts. We applied this catalogue of associations to investigate the impact of associating transcript-metabolite pairs on body mass index (BMI) as an example metabolic trait. This is achieved by conducting a comprehensive mediation analysis considering metabolites as mediators of gene expression effects and vice versa. We discovered large mediation networks comprising 27,023 potential mediation effects within 20,507 transcript-metabolite pairs. Resulting networks of highly connected (hub) transcripts and metabolites were leveraged to gain mechanistic insights into metabolic signaling pathways. In conclusion, here, we present the largest available multi-omics integration of genome-wide transcriptome data and metabolite data of amino acid and fatty acid metabolism and further leverage these findings to characterize potential mediation effects towards BMI proposing candidate mechanisms of obesity and related metabolic diseases. KEY MESSAGES: Thousands of associations of 72 amino acid and acylcarnitine metabolites and 8579 genes expand the knowledge of metabolome-transcriptome associations. A mediation analysis of effects on body mass index revealed large mediation networks of thousands of obesity-related gene-metabolite pairs. Highly connected, potentially mediating hub genes and metabolites enabled insight into obesity and related metabolic disease pathomechanisms.
Collapse
Affiliation(s)
- Carl Beuchel
- Institute for Medical Informatics, Statistics and Epidemiology, Leipzig University, Leipzig, Germany
| | - Julia Dittrich
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Leipzig, Germany
| | - Susen Becker
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Leipzig, Germany
- Department of Forensic Toxicology, Institute of Legal Medicine, University Leipzig, Leipzig, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology, Leipzig University, Leipzig, Germany
- LIFE - Leipzig Research Center for Civilization Diseases, Leipzig University, Leipzig, Germany
| | - Anke Tönjes
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University Hospital Leipzig, Leipzig, Germany
| | - Peter Kovacs
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University Hospital Leipzig, Leipzig, Germany
- Deutsches Zentrum für Diabetesforschung, Neuherberg, Germany
| | - Michael Stumvoll
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University Hospital Leipzig, Leipzig, Germany
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, Leipzig University, Leipzig, Germany
- LIFE - Leipzig Research Center for Civilization Diseases, Leipzig University, Leipzig, Germany
| | | | - Joachim Thiery
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Leipzig, Germany
- LIFE - Leipzig Research Center for Civilization Diseases, Leipzig University, Leipzig, Germany
| | - Berend Isermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Leipzig, Germany
- LIFE - Leipzig Research Center for Civilization Diseases, Leipzig University, Leipzig, Germany
| | - Uta Ceglarek
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Leipzig, Germany
- LIFE - Leipzig Research Center for Civilization Diseases, Leipzig University, Leipzig, Germany
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, Leipzig University, Leipzig, Germany.
- LIFE - Leipzig Research Center for Civilization Diseases, Leipzig University, Leipzig, Germany.
| |
Collapse
|
10
|
van der Spek A, Stewart ID, Kühnel B, Pietzner M, Alshehri T, Gauß F, Hysi PG, MahmoudianDehkordi S, Heinken A, Luik AI, Ladwig KH, Kastenmüller G, Menni C, Hertel J, Ikram MA, de Mutsert R, Suhre K, Gieger C, Strauch K, Völzke H, Meitinger T, Mangino M, Flaquer A, Waldenberger M, Peters A, Thiele I, Kaddurah-Daouk R, Dunlop BW, Rosendaal FR, Wareham NJ, Spector TD, Kunze S, Grabe HJ, Mook-Kanamori DO, Langenberg C, van Duijn CM, Amin N. Circulating metabolites modulated by diet are associated with depression. Mol Psychiatry 2023; 28:3874-3887. [PMID: 37495887 PMCID: PMC10730409 DOI: 10.1038/s41380-023-02180-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/03/2023] [Accepted: 07/03/2023] [Indexed: 07/28/2023]
Abstract
Metabolome reflects the interplay of genome and exposome at molecular level and thus can provide deep insights into the pathogenesis of a complex disease like major depression. To identify metabolites associated with depression we performed a metabolome-wide association analysis in 13,596 participants from five European population-based cohorts characterized for depression, and circulating metabolites using ultra high-performance liquid chromatography/tandem accurate mass spectrometry (UHPLC/MS/MS) based Metabolon platform. We tested 806 metabolites covering a wide range of biochemical processes including those involved in lipid, amino-acid, energy, carbohydrate, xenobiotic and vitamin metabolism for their association with depression. In a conservative model adjusting for life style factors and cardiovascular and antidepressant medication use we identified 8 metabolites, including 6 novel, significantly associated with depression. In individuals with depression, increased levels of retinol (vitamin A), 1-palmitoyl-2-palmitoleoyl-GPC (16:0/16:1) (lecithin) and mannitol/sorbitol and lower levels of hippurate, 4-hydroxycoumarin, 2-aminooctanoate (alpha-aminocaprylic acid), 10-undecenoate (11:1n1) (undecylenic acid), 1-linoleoyl-GPA (18:2) (lysophosphatidic acid; LPA 18:2) are observed. These metabolites are either directly food derived or are products of host and gut microbial metabolism of food-derived products. Our Mendelian randomization analysis suggests that low hippurate levels may be in the causal pathway leading towards depression. Our findings highlight putative actionable targets for depression prevention that are easily modifiable through diet interventions.
Collapse
Affiliation(s)
- Ashley van der Spek
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- SkylineDx B.V., Rotterdam, The Netherlands
| | | | - Brigitte Kühnel
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764, Neuherberg, Germany
| | - Maik Pietzner
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
- Computational Medicine, Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Tahani Alshehri
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Friederike Gauß
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Ferdinand-Sauerbruch-Str, 17475, Greifswald, Germany
| | - Pirro G Hysi
- Department of Twins Research and Genetic Epidemiology, Kings College London, London, UK
| | | | - Almut Heinken
- School of Medicine, University of Galway, University Road, Galway, Ireland
- Inserm UMRS 1256 NGERE - Nutrition, Genetics, and Environmental Risk Exposure, University of Lorraine, Nancy, France
| | - Annemarie I Luik
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Karl-Heinz Ladwig
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764, Neuherberg, Germany
- Department of Psychosomatic Medicine and Psychotherapy, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD e.V.), D-85764, Neuherberg, Germany
| | - Cristina Menni
- Department of Twins Research and Genetic Epidemiology, Kings College London, London, UK
| | - Johannes Hertel
- School of Medicine, University of Galway, University Road, Galway, Ireland
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Ellernholzstrasse 1-2, 17489, Greifswald, Germany
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, PO, 24144, Doha, Qatar
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD e.V.), D-85764, Neuherberg, Germany
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764, Neuherberg, Germany
- Chair of Genetic Epidemiology, IBE, Faculty of Medicine, LMU, Munich, Germany
| | - Henry Völzke
- Institute of Community Medicine, University Medicine Greifswald, Walter-Rathenau Str. 48, 17475, Greifswald, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764, Neuherberg, Germany
- Institute of Human Genetics, Technische Universität München, Munich, Germany
| | - Massimo Mangino
- Department of Twins Research and Genetic Epidemiology, Kings College London, London, UK
| | - Antonia Flaquer
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764, Neuherberg, Germany
- Chair of Genetic Epidemiology, IBE, Faculty of Medicine, LMU, Munich, Germany
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764, Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Annette Peters
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764, Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Ludwig-Maximilians-Universität München, IBE-Chair of Epidemiology, Munich, Germany
| | - Ines Thiele
- School of Medicine, University of Galway, University Road, Galway, Ireland
- Division of Microbiology, University of Galway, Galway, Ireland
- APC Microbiome, Ireland, Ireland
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
- Duke Institute of Brain Sciences, Duke University, Durham, NC, USA
- Department of Medicine, Duke University, Durham, NC, USA
| | - Boadie W Dunlop
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, US
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Tim D Spector
- Department of Twins Research and Genetic Epidemiology, Kings College London, London, UK
| | - Sonja Kunze
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, D-85764, Neuherberg, Germany
| | - Hans Jörgen Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Ellernholzstrasse 1-2, 17489, Greifswald, Germany
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, The Netherlands
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Nuffield Department of Population Health, University of Oxford, OX3 7LF, Oxford, UK
| | - Najaf Amin
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands.
- Nuffield Department of Population Health, University of Oxford, OX3 7LF, Oxford, UK.
| |
Collapse
|
11
|
Brown AA, Fernandez-Tajes JJ, Hong MG, Brorsson CA, Koivula RW, Davtian D, Dupuis T, Sartori A, Michalettou TD, Forgie IM, Adam J, Allin KH, Caiazzo R, Cederberg H, De Masi F, Elders PJM, Giordano GN, Haid M, Hansen T, Hansen TH, Hattersley AT, Heggie AJ, Howald C, Jones AG, Kokkola T, Laakso M, Mahajan A, Mari A, McDonald TJ, McEvoy D, Mourby M, Musholt PB, Nilsson B, Pattou F, Penet D, Raverdy V, Ridderstråle M, Romano L, Rutters F, Sharma S, Teare H, 't Hart L, Tsirigos KD, Vangipurapu J, Vestergaard H, Brunak S, Franks PW, Frost G, Grallert H, Jablonka B, McCarthy MI, Pavo I, Pedersen O, Ruetten H, Walker M, Adamski J, Schwenk JM, Pearson ER, Dermitzakis ET, Viñuela A. Genetic analysis of blood molecular phenotypes reveals common properties in the regulatory networks affecting complex traits. Nat Commun 2023; 14:5062. [PMID: 37604891 PMCID: PMC10442420 DOI: 10.1038/s41467-023-40569-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 08/02/2023] [Indexed: 08/23/2023] Open
Abstract
We evaluate the shared genetic regulation of mRNA molecules, proteins and metabolites derived from whole blood from 3029 human donors. We find abundant allelic heterogeneity, where multiple variants regulate a particular molecular phenotype, and pleiotropy, where a single variant associates with multiple molecular phenotypes over multiple genomic regions. The highest proportion of share genetic regulation is detected between gene expression and proteins (66.6%), with a further median shared genetic associations across 49 different tissues of 78.3% and 62.4% between plasma proteins and gene expression. We represent the genetic and molecular associations in networks including 2828 known GWAS variants, showing that GWAS variants are more often connected to gene expression in trans than other molecular phenotypes in the network. Our work provides a roadmap to understanding molecular networks and deriving the underlying mechanism of action of GWAS variants using different molecular phenotypes in an accessible tissue.
Collapse
Affiliation(s)
- Andrew A Brown
- Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, United Kingdom
| | - Juan J Fernandez-Tajes
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, United Kingdom
| | - Mun-Gwan Hong
- Science for Life Laboratory, School of Biotechnology, KTH - Royal Institute of Technology, Solna, SE-171 21, Sweden
| | - Caroline A Brorsson
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2100, Denmark
| | - Robert W Koivula
- Oxford Centre for Diabetes Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LJ, United Kingdom
| | - David Davtian
- Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, United Kingdom
| | - Théo Dupuis
- Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, United Kingdom
| | - Ambra Sartori
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, 1211, Switzerland
- Institute for Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, 1211, Switzerland
- Swiss Institute of Bioinformatics, Geneva, 1211, Switzerland
| | - Theodora-Dafni Michalettou
- Biosciences Institute, Faculty of Medical Sciences, University of Newcastle, Newcastle upon Tyne, NE1 4EP, United Kingdom
| | - Ian M Forgie
- Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, United Kingdom
| | - Jonathan Adam
- German Center for Diabetes Research (DZD), Neuherberg, 85764, Germany
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, 85764, Germany
| | - Kristine H Allin
- The Novo Nordisk Center for Basic Metabolic Research, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, DK-2100, Denmark
| | - Robert Caiazzo
- University of Lille, Inserm, Lille Pasteur Institute, Lille, France
| | - Henna Cederberg
- Internal Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Federico De Masi
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Petra J M Elders
- Department of General Practice, Amsterdam UMC- location Vumc, Amsterdam Public Health research institute, Amsterdam, The Netherlands
| | - Giuseppe N Giordano
- Department of Clinical Science, Genetic and Molecular Epidemiology, Lund University Diabetes Centre, Malmö, Sweden
| | - Mark Haid
- Metabolomics and Proteomics Core, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, 85764, Germany
| | - Torben Hansen
- The Novo Nordisk Center for Basic Metabolic Research, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, DK-2100, Denmark
| | - Tue H Hansen
- The Novo Nordisk Center for Basic Metabolic Research, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, DK-2100, Denmark
| | - Andrew T Hattersley
- Department of Clinical and Biomedical Sciences, University of Exeter College of Medicine & Health, Exeter, EX25DW, United Kingdom
| | - Alison J Heggie
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Cédric Howald
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, 1211, Switzerland
- Institute for Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, 1211, Switzerland
- Swiss Institute of Bioinformatics, Geneva, 1211, Switzerland
| | - Angus G Jones
- Department of Clinical and Biomedical Sciences, University of Exeter College of Medicine & Health, Exeter, EX25DW, United Kingdom
| | - Tarja Kokkola
- Internal Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Markku Laakso
- Internal Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, United Kingdom
| | - Andrea Mari
- Institute of Neuroscience, National Research Council, Padova, 35127, Italy
| | - Timothy J McDonald
- Blood Sciences, Royal Devon and Exeter NHS Foundation Trust, Exeter, EX2 5DW, United Kingdom
| | - Donna McEvoy
- Diabetes Research Network, Royal Victoria Infirmary, Newcastle upon Tyne, United Kingdom
| | - Miranda Mourby
- Nuffield Department of Population Health, Centre for Health, Law and Emerging Technologies (HeLEX), University of Oxford, Oxford, OX2 7DD, United Kingdom
| | - Petra B Musholt
- Global Development, Sanofi-Aventis Deutschland GmbH, Hoechst Industrial Park, Frankfurt am Main, 65926, Germany
| | - Birgitte Nilsson
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Francois Pattou
- University of Lille, Inserm, Lille Pasteur Institute, Lille, France
| | - Deborah Penet
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, 1211, Switzerland
- Institute for Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, 1211, Switzerland
- Swiss Institute of Bioinformatics, Geneva, 1211, Switzerland
| | - Violeta Raverdy
- University of Lille, Inserm, Lille Pasteur Institute, Lille, France
| | | | - Luciana Romano
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, 1211, Switzerland
- Institute for Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, 1211, Switzerland
- Swiss Institute of Bioinformatics, Geneva, 1211, Switzerland
| | - Femke Rutters
- Epidemiology and Data Science, VUMC, Amsterdam, The Netherlands
| | - Sapna Sharma
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, 85764, Germany
- Food Chemistry and Molecular and Sensory Science, Technical University of Munich, München, Germany
| | - Harriet Teare
- Centre for Health Law and Emerging Technologies, Department of Population Health, University of Oxford, Old Road Campus, Oxford, OX3 7DQ, United Kingdom
| | - Leen 't Hart
- Epidemiology and Data Science, VUMC, Amsterdam, The Netherlands
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Biomedical Data Sciences, Molecular Epidemiology section, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Jagadish Vangipurapu
- Internal Medicine, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland
| | - Henrik Vestergaard
- The Novo Nordisk Center for Basic Metabolic Research, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, DK-2100, Denmark
- Steno Diabetes Center Copenhagen, Copenhagen, Denmark
| | - Søren Brunak
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2100, Denmark
| | - Paul W Franks
- Department of Clinical Science, Genetic and Molecular Epidemiology, Lund University Diabetes Centre, Malmö, Sweden
| | - Gary Frost
- Nutrition and Dietetics Research Group, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Harald Grallert
- German Center for Diabetes Research (DZD), Neuherberg, 85764, Germany
- Research Unit of Molecular Epidemiology, Institute of Epidemiology, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, 85764, Germany
| | - Bernd Jablonka
- Sanofi Partnering, Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, 65926, Germany
| | - Mark I McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, United Kingdom
- GENENTECH, 1 DNA Way, San Francisco, CA, 94080, USA
| | - Imre Pavo
- Eli Lilly Regional Operations Ges.m.b.H, Vienna, 1030, Austria
| | - Oluf Pedersen
- Center for Clinical Metabolic Research, Herlev and Gentofte University Hospital, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2100, Denmark
| | - Hartmut Ruetten
- Sanofi Partnering, Sanofi-Aventis Deutschland GmbH, Frankfurt am Main, 65926, Germany
| | - Mark Walker
- Translational and Clinical Research Institute, Faculty of Medical Sciences, University of Newcastle, Newcastle upon Tyne, United Kingdom
| | - Jerzy Adamski
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Institute of Experimental Genetics, German Research Center for Environmental Health, Helmholtz Zentrum München, Neuherberg, 85764, Germany
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Jochen M Schwenk
- Science for Life Laboratory, School of Biotechnology, KTH - Royal Institute of Technology, Solna, SE-171 21, Sweden
| | - Ewan R Pearson
- Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, United Kingdom
| | - Emmanouil T Dermitzakis
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, 1211, Switzerland.
- Institute for Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, 1211, Switzerland.
- Swiss Institute of Bioinformatics, Geneva, 1211, Switzerland.
| | - Ana Viñuela
- Biosciences Institute, Faculty of Medical Sciences, University of Newcastle, Newcastle upon Tyne, NE1 4EP, United Kingdom.
| |
Collapse
|
12
|
Abstract
Over the last decade, immunometabolism has emerged as a novel interdisciplinary field of research and yielded significant fundamental insights into the regulation of immune responses. Multiple classical approaches to interrogate immunometabolism, including bulk metabolic profiling and analysis of metabolic regulator expression, paved the way to appreciating the physiological complexity of immunometabolic regulation in vivo. Studying immunometabolism at the systems level raised the need to transition towards the next-generation technology for metabolic profiling and analysis. Spatially resolved metabolic imaging and computational algorithms for multi-modal data integration are new approaches to connecting metabolism and immunity. In this review, we discuss recent studies that highlight the complex physiological interplay between immune responses and metabolism and give an overview of technological developments that bear the promise of capturing this complexity most directly and comprehensively.
Collapse
Affiliation(s)
- Denis A Mogilenko
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA; ,
- Current affiliation: Department of Medicine, Department of Pathology, Microbiology, and Immunology, and Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee, USA;
| | - Alexey Sergushichev
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA; ,
- Computer Technologies Laboratory, ITMO University, Saint Petersburg, Russia
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA; ,
| |
Collapse
|
13
|
Jain P, Yates M, de Celis CR, Drineas P, Jahanshad N, Thompson P, Paschou P. Multiomic approach and Mendelian randomization analysis identify causal associations between blood biomarkers and subcortical brain structure volumes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.30.23287968. [PMID: 37066330 PMCID: PMC10104218 DOI: 10.1101/2023.03.30.23287968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Alterations in subcortical brain structure volumes have been found to be associated with several neurodegenerative and psychiatric disorders. At the same time, genome-wide association studies (GWAS) have identified numerous common variants associated with brain structure. In this study, we integrate these findings, aiming to identify proteins, metabolites, or microbes that have a putative causal association with subcortical brain structure volumes via a two-sample Mendelian randomization approach. This method uses genetic variants as instrument variables to identify potentially causal associations between an exposure and an outcome. The exposure data that we analyzed comprised genetic associations for 2,994 plasma proteins, 237 metabolites, and 103 microbial genera. The outcome data included GWAS data for seven subcortical brain structure volumes including accumbens, amygdala, caudate, hippocampus, pallidum, putamen, and thalamus. Eleven proteins and six metabolites were found to have a significant association with subcortical structure volumes. We found causal associations between amygdala volume and granzyme A as well as association between accumbens volume and plasma protease c1 inhibitor. Among metabolites, urate had the strongest association with thalamic volume. No significant associations were detected between the microbial genera and subcortical brain structure volumes. We also observed significant enrichment for biological processes such as proteolysis, regulation of the endoplasmic reticulum apoptotic signaling pathway, and negative regulation of DNA binding. Our findings provide insights to the mechanisms through which brain volumes may be affected in the pathogenesis of neurodevelopmental and psychiatric disorders and point to potential treatment targets for disorders that are associated with subcortical brain structure volumes.
Collapse
Affiliation(s)
- Pritesh Jain
- Department of Biological Sciences, Purdue University
| | - Madison Yates
- Department of Biological Sciences, Purdue University
| | | | | | - Neda Jahanshad
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of South California
| | - Paul Thompson
- Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of South California
| | | |
Collapse
|
14
|
Fu J, Zhu F, Xu CJ, Li Y. Metabolomics meets systems immunology. EMBO Rep 2023; 24:e55747. [PMID: 36916532 PMCID: PMC10074123 DOI: 10.15252/embr.202255747] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 12/24/2022] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
Metabolic processes play a critical role in immune regulation. Metabolomics is the systematic analysis of small molecules (metabolites) in organisms or biological samples, providing an opportunity to comprehensively study interactions between metabolism and immunity in physiology and disease. Integrating metabolomics into systems immunology allows the exploration of the interactions of multilayered features in the biological system and the molecular regulatory mechanism of these features. Here, we provide an overview on recent technological developments of metabolomic applications in immunological research. To begin, two widely used metabolomics approaches are compared: targeted and untargeted metabolomics. Then, we provide a comprehensive overview of the analysis workflow and the computational tools available, including sample preparation, raw spectra data preprocessing, data processing, statistical analysis, and interpretation. Third, we describe how to integrate metabolomics with other omics approaches in immunological studies using available tools. Finally, we discuss new developments in metabolomics and its prospects for immunology research. This review provides guidance to researchers using metabolomics and multiomics in immunity research, thus facilitating the application of systems immunology to disease research.
Collapse
Affiliation(s)
- Jianbo Fu
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.,TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany.,College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Cheng-Jian Xu
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.,TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany.,Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Yang Li
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany.,TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School (MHH), Hannover, Germany.,Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
15
|
Player JK, Riordan SM, Duncan RS, Koulen P. Analysis of Glaucoma Associated Genes in Response to Inflammation, an Examination of a Public Data Set Derived from Peripheral Blood from Patients with Hepatitis C. Clin Ophthalmol 2022; 16:2093-2103. [PMID: 35770250 PMCID: PMC9236525 DOI: 10.2147/opth.s364739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/02/2022] [Indexed: 12/16/2022] Open
Abstract
Introduction Glaucoma is the second leading cause of blindness worldwide and despite its prevalence, there are still many unanswered questions related to its pathogenesis. There is evidence that oxidative stress and inflammation play a major role in disease progression. Glaucoma patients from several studies showed altered gene expression in leukocytes, revealing the possibility of using peripheral biomarkers to diagnose or stage glaucoma. The fact that glaucoma is associated with gene expression changes in tissues distant from the retina underscores the possible involvement of systemic oxidative stress and inflammation as potential contributing or compounding factors in glaucoma. Methods We assembled a list of oxidative stress and inflammatory markers related to glaucoma based on a review of the literature. In addition, we utilized publicly available data sets of gene expression values collected from peripheral blood mononuclear cells and macrophages from two patient groups: those chronically infected by the hepatitis C virus and those who have cleared it. Activation of the innate immune response can render cells or tissues more responsive to a second delayed proinflammatory stimulus. Additional gene expression data from these cells after subsequent polyinosinic:polycytidylic acid treatment, used to elicit an acute inflammatory response, allowed for the investigation of the acute inflammatory response in these groups. We used fold-change comparison values between the two patient groups to identify genes of interest. Results A comparison analysis identified 17 glaucoma biomarkers that were differentially expressed in response to HCV-mediated inflammation. Of these 17, six had significant p-values in the baseline vs treated values. Expression data of these genes were compared between patients who had cleared the Hepatitis C virus versus those who had not and identified three genes of interest for further study. Discussion These results support our hypothesis that inflammation secondary to Hepatitis C virus infection affects the expression of glaucoma biomarker genes related to the antioxidant response and inflammation. In addition, they provide several potential targets for further research into understanding the relationship between innate responses to viral infection and inflammatory aspects of glaucoma and for potential use as a predictive biomarker or pharmacological intervention in glaucoma.
Collapse
Affiliation(s)
- Jacob K Player
- Vision Research Center, Department of Ophthalmology, School of Medicine, University of Missouri – Kansas City, Kansas City, MO, 64108, USA
| | - Sean M Riordan
- Vision Research Center, Department of Ophthalmology, School of Medicine, University of Missouri – Kansas City, Kansas City, MO, 64108, USA
| | - R Scott Duncan
- Vision Research Center, Department of Ophthalmology, School of Medicine, University of Missouri – Kansas City, Kansas City, MO, 64108, USA
| | - Peter Koulen
- Vision Research Center, Department of Ophthalmology, School of Medicine, University of Missouri – Kansas City, Kansas City, MO, 64108, USA
- Department of Biomedical Sciences, School of Medicine, University of Missouri – Kansas City, Kansas City, MO, 64108, USA
- Correspondence: Peter Koulen, Vision Research Center, Department of Ophthalmology, School of Medicine, University of Missouri – Kansas City, 2411 Holmes Street, Kansas City, MO, 64108, USA, Tel +1 816-235-6773, Email
| |
Collapse
|
16
|
Chu X, Jaeger M, Beumer J, Bakker OB, Aguirre-Gamboa R, Oosting M, Smeekens SP, Moorlag S, Mourits VP, Koeken VACM, de Bree C, Jansen T, Mathews IT, Dao K, Najhawan M, Watrous JD, Joosten I, Sharma S, Koenen HJPM, Withoff S, Jonkers IH, Netea-Maier RT, Xavier RJ, Franke L, Xu CJ, Joosten LAB, Sanna S, Jain M, Kumar V, Clevers H, Wijmenga C, Netea MG, Li Y. Integration of metabolomics, genomics, and immune phenotypes reveals the causal roles of metabolites in disease. Genome Biol 2021; 22:198. [PMID: 34229738 PMCID: PMC8259168 DOI: 10.1186/s13059-021-02413-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 06/21/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Recent studies highlight the role of metabolites in immune diseases, but it remains unknown how much of this effect is driven by genetic and non-genetic host factors. RESULT We systematically investigate circulating metabolites in a cohort of 500 healthy subjects (500FG) in whom immune function and activity are deeply measured and whose genetics are profiled. Our data reveal that several major metabolic pathways, including the alanine/glutamate pathway and the arachidonic acid pathway, have a strong impact on cytokine production in response to ex vivo stimulation. We also examine the genetic regulation of metabolites associated with immune phenotypes through genome-wide association analysis and identify 29 significant loci, including eight novel independent loci. Of these, one locus (rs174584-FADS2) associated with arachidonic acid metabolism is causally associated with Crohn's disease, suggesting it is a potential therapeutic target. CONCLUSION This study provides a comprehensive map of the integration between the blood metabolome and immune phenotypes, reveals novel genetic factors that regulate blood metabolite concentrations, and proposes an integrative approach for identifying new disease treatment targets.
Collapse
Affiliation(s)
- Xiaojing Chu
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700, RB, Groningen, the Netherlands
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine, CiiM, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Martin Jaeger
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525, HP, Nijmegen, the Netherlands
| | - Joep Beumer
- Oncode Institute, Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, 3584, CT, Utrecht, the Netherlands
| | - Olivier B Bakker
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700, RB, Groningen, the Netherlands
| | - Raul Aguirre-Gamboa
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700, RB, Groningen, the Netherlands
| | - Marije Oosting
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525, HP, Nijmegen, the Netherlands
| | - Sanne P Smeekens
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525, HP, Nijmegen, the Netherlands
| | - Simone Moorlag
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525, HP, Nijmegen, the Netherlands
| | - Vera P Mourits
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525, HP, Nijmegen, the Netherlands
| | - Valerie A C M Koeken
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine, CiiM, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525, HP, Nijmegen, the Netherlands
| | - Charlotte de Bree
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525, HP, Nijmegen, the Netherlands
| | - Trees Jansen
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525, HP, Nijmegen, the Netherlands
| | - Ian T Mathews
- Departments of Medicine and Pharmacology, University of California, San Diego, CA, USA
- La Jolla Institute, La Jolla, CA, USA
| | - Khoi Dao
- Departments of Medicine and Pharmacology, University of California, San Diego, CA, USA
| | - Mahan Najhawan
- Departments of Medicine and Pharmacology, University of California, San Diego, CA, USA
| | - Jeramie D Watrous
- Departments of Medicine and Pharmacology, University of California, San Diego, CA, USA
| | - Irma Joosten
- Department of Laboratory Medicine, Laboratory for Medical Immunology, Radboud University Medical Center, 6525, GA, Nijmegen, the Netherlands
| | | | - Hans J P M Koenen
- Department of Laboratory Medicine, Laboratory for Medical Immunology, Radboud University Medical Center, 6525, GA, Nijmegen, the Netherlands
| | - Sebo Withoff
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700, RB, Groningen, the Netherlands
| | - Iris H Jonkers
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700, RB, Groningen, the Netherlands
| | - Romana T Netea-Maier
- Department of Internal Medicine, Division of Endocrinology, Radboud University Medical Center, 6525, HP, Nijmegen, the Netherlands
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard University, Cambridge, MA, 02142, USA
- Center for Computational and Integrative Biology and Gastrointestinal Unit, Massachusetts General Hospital, Harvard School of Medicine, Boston, MA, 02114, USA
| | - Lude Franke
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700, RB, Groningen, the Netherlands
| | - Cheng-Jian Xu
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine, CiiM, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525, HP, Nijmegen, the Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525, HP, Nijmegen, the Netherlands
| | - Serena Sanna
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700, RB, Groningen, the Netherlands
| | - Mohit Jain
- Departments of Medicine and Pharmacology, University of California, San Diego, CA, USA
| | - Vinod Kumar
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700, RB, Groningen, the Netherlands
| | - Hans Clevers
- Oncode Institute, Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, 3584, CT, Utrecht, the Netherlands
- Oncode Institute, Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584, CS, Utrecht, the Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700, RB, Groningen, the Netherlands.
- Department of Immunology, University of Oslo, Oslo University Hospital, Rikshospitalet, 0372, Oslo, Norway.
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525, HP, Nijmegen, the Netherlands.
- Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115, Bonn, Germany.
| | - Yang Li
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9700, RB, Groningen, the Netherlands.
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine, CiiM, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany.
- TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany.
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525, HP, Nijmegen, the Netherlands.
| |
Collapse
|
17
|
Jhun MA, Mendelson M, Wilson R, Gondalia R, Joehanes R, Salfati E, Zhao X, Braun KVE, Do AN, Hedman ÅK, Zhang T, Carnero-Montoro E, Shen J, Bartz TM, Brody JA, Montasser ME, O’Connell JR, Yao C, Xia R, Boerwinkle E, Grove M, Guan W, Liliane P, Singmann P, Müller-Nurasyid M, Meitinger T, Gieger C, Peters A, Zhao W, Ware EB, Smith JA, Dhana K, van Meurs J, Uitterlinden A, Ikram MA, Ghanbari M, Zhi D, Gustafsson S, Lind L, Li S, Sun D, Spector TD, Chen YDI, Damcott C, Shuldiner AR, Absher DM, Horvath S, Tsao PS, Kardia S, Psaty BM, Sotoodehnia N, Bell JT, Ingelsson E, Chen W, Dehghan A, Arnett DK, Waldenberger M, Hou L, Whitsel EA, Baccarelli A, Levy D, Fornage M, Irvin MR, Assimes TL. A multi-ethnic epigenome-wide association study of leukocyte DNA methylation and blood lipids. Nat Commun 2021; 12:3987. [PMID: 34183656 PMCID: PMC8238961 DOI: 10.1038/s41467-021-23899-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
Here we examine the association between DNA methylation in circulating leukocytes and blood lipids in a multi-ethnic sample of 16,265 subjects. We identify 148, 35, and 4 novel associations among Europeans, African Americans, and Hispanics, respectively, and an additional 186 novel associations through a trans-ethnic meta-analysis. We observe a high concordance in the direction of effects across racial/ethnic groups, a high correlation of effect sizes between high-density lipoprotein and triglycerides, a modest overlap of associations with epigenome-wide association studies of other cardio-metabolic traits, and a largely non-overlap with lipid loci identified to date through genome-wide association studies. Thirty CpGs reached significance in at least 2 racial/ethnic groups including 7 that showed association with the expression of an annotated gene. CpGs annotated to CPT1A showed evidence of being influenced by triglycerides levels. DNA methylation levels of circulating leukocytes show robust and consistent association with blood lipid levels across multiple racial/ethnic groups.
Collapse
Affiliation(s)
- Min-A Jhun
- grid.214458.e0000000086837370Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI USA ,grid.168010.e0000000419368956Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Michael Mendelson
- grid.94365.3d0000 0001 2297 5165Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD USA ,grid.2515.30000 0004 0378 8438Department of Cardiology, Boston Children’s Hospital, Boston, MA USA
| | - Rory Wilson
- grid.4567.00000 0004 0483 2525Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany ,grid.4567.00000 0004 0483 2525Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Rahul Gondalia
- grid.410711.20000 0001 1034 1720Department of Epidemiology, University of North Carolina, Chapel Hill, NC USA
| | - Roby Joehanes
- grid.38142.3c000000041936754XHebrew SeniorLife, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA USA
| | - Elias Salfati
- grid.168010.e0000000419368956Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Xiaoping Zhao
- grid.267308.80000 0000 9206 2401The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX USA
| | - Kim Valeska Emilie Braun
- grid.5645.2000000040459992XDepartment of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Anh Nguyet Do
- grid.265892.20000000106344187Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL USA
| | - Åsa K. Hedman
- grid.8993.b0000 0004 1936 9457Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Tao Zhang
- grid.265219.b0000 0001 2217 8588Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA USA
| | - Elena Carnero-Montoro
- grid.13097.3c0000 0001 2322 6764Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King’s College London, London, UK ,grid.470860.d0000 0004 4677 7069GENYO, Center for Genomics and Oncological Research Pfizer/University of Granada/Andalusian Regional Government, Granada, Spain
| | - Jincheng Shen
- grid.223827.e0000 0001 2193 0096Department of Population Health Sciences, University of Utah, Salt Lake City, UT USA
| | - Traci M. Bartz
- grid.34477.330000000122986657Cardiovascular Health Research Unit, Departments of Medicine and Biostatistics, University of Washington, Seattle, WA USA
| | - Jennifer A. Brody
- grid.34477.330000000122986657Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA USA
| | - May E. Montasser
- grid.411024.20000 0001 2175 4264Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD USA ,grid.411024.20000 0001 2175 4264Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD USA
| | - Jeff R. O’Connell
- grid.411024.20000 0001 2175 4264Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD USA ,grid.411024.20000 0001 2175 4264Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD USA
| | - Chen Yao
- grid.94365.3d0000 0001 2297 5165Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD USA
| | - Rui Xia
- grid.267308.80000 0000 9206 2401The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX USA
| | - Eric Boerwinkle
- grid.267308.80000 0000 9206 2401School of Public Health, University of Texas Health Science Center at Houston, Huston, TX USA
| | - Megan Grove
- grid.267308.80000 0000 9206 2401School of Public Health, University of Texas Health Science Center at Houston, Huston, TX USA
| | - Weihua Guan
- grid.17635.360000000419368657Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN USA
| | - Pfeiffer Liliane
- grid.4567.00000 0004 0483 2525Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany ,grid.4567.00000 0004 0483 2525Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Paula Singmann
- grid.4567.00000 0004 0483 2525Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany ,grid.4567.00000 0004 0483 2525Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Martina Müller-Nurasyid
- grid.4567.00000 0004 0483 2525Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany ,grid.5252.00000 0004 1936 973XIBE, Faculty of Medicine, LMU Munich, Munich, Germany ,grid.410607.4Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Thomas Meitinger
- grid.410607.4Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany ,grid.4567.00000 0004 0483 2525Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany ,grid.6936.a0000000123222966Institute of Human Genetics, Technical University Munich, Munich, Germany
| | - Christian Gieger
- grid.4567.00000 0004 0483 2525Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany ,grid.4567.00000 0004 0483 2525Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Annette Peters
- grid.4567.00000 0004 0483 2525Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany ,grid.410607.4Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Wei Zhao
- grid.214458.e0000000086837370Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI USA
| | - Erin B. Ware
- grid.214458.e0000000086837370Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI USA ,Survey Research Center, Institute for Social Research, Ann Arbor, MI USA
| | - Jennifer A. Smith
- grid.214458.e0000000086837370Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI USA ,Survey Research Center, Institute for Social Research, Ann Arbor, MI USA
| | - Klodian Dhana
- grid.240684.c0000 0001 0705 3621Department of Internal Medicine, Rush University Medical Center, Chicago, IL USA
| | - Joyce van Meurs
- grid.5645.2000000040459992XDepartment of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Andre Uitterlinden
- grid.5645.2000000040459992XDepartment of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Mohammad Arfan Ikram
- grid.5645.2000000040459992XDepartment of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Mohsen Ghanbari
- grid.5645.2000000040459992XDepartment of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Deugi Zhi
- grid.267308.80000 0000 9206 2401School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX USA
| | - Stefan Gustafsson
- grid.8993.b0000 0004 1936 9457Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Lars Lind
- grid.8993.b0000 0004 1936 9457Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Shengxu Li
- grid.265219.b0000 0001 2217 8588Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA USA
| | - Dianjianyi Sun
- grid.265219.b0000 0001 2217 8588Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA USA ,grid.11135.370000 0001 2256 9319Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Tim D. Spector
- grid.13097.3c0000 0001 2322 6764Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King’s College London, London, UK
| | - Yii-der Ida Chen
- grid.239844.00000 0001 0157 6501Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute, and Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA USA
| | - Coleen Damcott
- grid.411024.20000 0001 2175 4264Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD USA ,grid.411024.20000 0001 2175 4264Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD USA
| | - Alan R. Shuldiner
- grid.411024.20000 0001 2175 4264Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD USA ,grid.411024.20000 0001 2175 4264Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD USA
| | - Devin M. Absher
- grid.417691.c0000 0004 0408 3720HudsonAlpha Institute for Biotechnology, Huntsville, AL USA
| | - Steve Horvath
- grid.19006.3e0000 0000 9632 6718Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA USA ,grid.19006.3e0000 0000 9632 6718Department of Biostatistics, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA USA
| | - Philip S. Tsao
- grid.168010.e0000000419368956Department of Medicine, Stanford University School of Medicine, Stanford, CA USA ,grid.280747.e0000 0004 0419 2556VA Palo Alto Healthcare System, Palo Alto, CA USA ,grid.168010.e0000000419368956Stanford Cardiovascular Institute, Stanford University, Stanford, CA USA
| | - Sharon Kardia
- grid.214458.e0000000086837370Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI USA
| | - Bruce M. Psaty
- grid.34477.330000000122986657Cardiovascular Health Research Unit, Departments of Epidemiology, Medicine, and Health Services, University of Washington, Seattle, WA USA ,grid.488833.c0000 0004 0615 7519Kaiser Permanente Washington Health Research Institute, Seattle, WA USA
| | - Nona Sotoodehnia
- grid.34477.330000000122986657Cardiovascular Health Research Unit, Division of Cardiology, University of Washington, Seattle, WA USA
| | - Jordana T. Bell
- grid.13097.3c0000 0001 2322 6764Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King’s College London, London, UK
| | - Erik Ingelsson
- grid.168010.e0000000419368956Department of Medicine, Stanford University School of Medicine, Stanford, CA USA ,grid.8993.b0000 0004 1936 9457Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden ,grid.168010.e0000000419368956Stanford Cardiovascular Institute, Stanford University, Stanford, CA USA ,grid.168010.e0000000419368956Stanford Diabetes Research Center, Stanford University, Stanford, CA USA
| | - Wei Chen
- grid.265219.b0000 0001 2217 8588Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA USA
| | - Abbas Dehghan
- grid.5645.2000000040459992XDepartment of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands ,grid.7445.20000 0001 2113 8111Department of Biostatistics and Epidemiology, MRC Centre for Environment and Health, School of Public Health, Imperial College, London, UK
| | - Donna K. Arnett
- grid.265892.20000000106344187Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL USA
| | - Melanie Waldenberger
- grid.4567.00000 0004 0483 2525Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany ,grid.4567.00000 0004 0483 2525Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany
| | - Lifang Hou
- grid.16753.360000 0001 2299 3507Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL USA
| | - Eric A. Whitsel
- grid.410711.20000 0001 1034 1720Department of Epidemiology, University of North Carolina, Chapel Hill, NC USA ,grid.410711.20000 0001 1034 1720Department of Medicine, University of North Carolina, Chapel Hill, NC USA
| | - Andrea Baccarelli
- grid.38142.3c000000041936754XDepartment of Environmental Health Sciences, Harvard T.H. Chan School of Public Health, Boston, MA USA ,grid.21729.3f0000000419368729Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY USA
| | - Daniel Levy
- grid.94365.3d0000 0001 2297 5165Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD USA ,grid.510954.c0000 0004 0444 3861Framingham Heart Study, Framingham, MA USA
| | - Myriam Fornage
- grid.267308.80000 0000 9206 2401The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX USA
| | - Marguerite R. Irvin
- grid.265892.20000000106344187Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL USA
| | - Themistocles L. Assimes
- grid.168010.e0000000419368956Department of Medicine, Stanford University School of Medicine, Stanford, CA USA ,grid.280747.e0000 0004 0419 2556VA Palo Alto Healthcare System, Palo Alto, CA USA ,grid.168010.e0000000419368956Stanford Cardiovascular Institute, Stanford University, Stanford, CA USA
| |
Collapse
|
18
|
Chu X, Zhang B, Koeken VACM, Gupta MK, Li Y. Multi-Omics Approaches in Immunological Research. Front Immunol 2021; 12:668045. [PMID: 34177908 PMCID: PMC8226116 DOI: 10.3389/fimmu.2021.668045] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/28/2021] [Indexed: 12/14/2022] Open
Abstract
The immune system plays a vital role in health and disease, and is regulated through a complex interactive network of many different immune cells and mediators. To understand the complexity of the immune system, we propose to apply a multi-omics approach in immunological research. This review provides a complete overview of available methodological approaches for the different omics data layers relevant for immunological research, including genetics, epigenetics, transcriptomics, proteomics, metabolomics, and cellomics. Thereafter, we describe the various methods for data analysis as well as how to integrate different layers of omics data. Finally, we discuss the possible applications of multi-omics studies and opportunities they provide for understanding the complex regulatory networks as well as immune variation in various immune-related diseases.
Collapse
Affiliation(s)
- Xiaojing Chu
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Bowen Zhang
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Valerie A. C. M. Koeken
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Manoj Kumar Gupta
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Yang Li
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
- Department of Computational Biology for Individualised Medicine, Centre for Individualised Infection Medicine (CiiM), a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
19
|
Lu H, Zhang J, Chen YE, Garcia-Barrio MT. Integration of Transformative Platforms for the Discovery of Causative Genes in Cardiovascular Diseases. Cardiovasc Drugs Ther 2021; 35:637-654. [PMID: 33856594 PMCID: PMC8216854 DOI: 10.1007/s10557-021-07175-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/18/2021] [Indexed: 12/11/2022]
Abstract
Cardiovascular diseases are the leading cause of morbidity and mortality worldwide. Genome-wide association studies (GWAS) are powerful epidemiological tools to find genes and variants associated with cardiovascular diseases while follow-up biological studies allow to better understand the etiology and mechanisms of disease and assign causality. Improved methodologies and reduced costs have allowed wider use of bulk and single-cell RNA sequencing, human-induced pluripotent stem cells, organoids, metabolomics, epigenomics, and novel animal models in conjunction with GWAS. In this review, we feature recent advancements relevant to cardiovascular diseases arising from the integration of genetic findings with multiple enabling technologies within multidisciplinary teams to highlight the solidifying transformative potential of this approach. Well-designed workflows integrating different platforms are greatly improving and accelerating the unraveling and understanding of complex disease processes while promoting an effective way to find better drug targets, improve drug design and repurposing, and provide insight towards a more personalized clinical practice.
Collapse
Affiliation(s)
- Haocheng Lu
- Department of Internal Medicine, University of Michigan Medical Center, 2800 Plymouth Rd, Ann Arbor, MI, 48109-2800, USA
| | - Jifeng Zhang
- Department of Internal Medicine, University of Michigan Medical Center, 2800 Plymouth Rd, Ann Arbor, MI, 48109-2800, USA
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Y Eugene Chen
- Department of Internal Medicine, University of Michigan Medical Center, 2800 Plymouth Rd, Ann Arbor, MI, 48109-2800, USA.
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA.
| | - Minerva T Garcia-Barrio
- Department of Internal Medicine, University of Michigan Medical Center, 2800 Plymouth Rd, Ann Arbor, MI, 48109-2800, USA.
| |
Collapse
|
20
|
Abstract
Mast cells and eosinophils are the key effector cells of allergy [1]. In general, allergic reactions are composed of two phases, namely an early phase and a late phase, and after that resolution occurs. If the allergic reactions fail to resolve after the late phase, allergic inflammation (AI) can evolve into a chronic phase mainly involving mast cells and eosinophils that abundantly coexist in the inflamed tissue in the late and chronic phases and cross-talk in a bidirectional manner. We defined these bidirectional interactions between MCs and Eos, as the "allergic effector unit." This cross talk is mediated by both physical cell-cell contacts through cell surface receptors such as CD48, 2B4, and respective ligands and through released mediators such as various specific granular mediators, arachidonic acid metabolites, cytokines, and chemokines [2, 3]. The allergic effector unit can be studied in vitro in a customized co-culture system using mast cells and eosinophils derived from either mouse or human sources.
Collapse
|
21
|
Burton-Pimentel KJ, Pimentel G, Hughes M, Michielsen CC, Fatima A, Vionnet N, Afman LA, Roche HM, Brennan L, Ibberson M, Vergères G. Discriminating Dietary Responses by Combining Transcriptomics and Metabolomics Data in Nutrition Intervention Studies. Mol Nutr Food Res 2020; 65:e2000647. [PMID: 33325641 PMCID: PMC8221028 DOI: 10.1002/mnfr.202000647] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 11/03/2020] [Indexed: 12/17/2022]
Abstract
Scope Combining different “omics” data types in a single, integrated analysis may better characterize the effects of diet on human health. Methods and results The performance of two data integration tools, similarity network fusion tool (SNFtool) and Data Integration Analysis for Biomarker discovery using Latent variable approaches for “Omics” (DIABLO; MixOmics), in discriminating responses to diet and metabolic phenotypes is investigated by combining transcriptomics and metabolomics datasets from three human intervention studies: a postprandial crossover study testing dairy foods (n = 7; study 1), a postprandial challenge study comparing obese and non‐obese subjects (n = 13; study 2); and an 8‐week parallel intervention study that assessed three diets with variable lipid content on fasting parameters (n = 39; study 3). In study 1, combining datasets using SNF or DIABLO significantly improve sample classification. For studies 2 and 3, the value of SNF integration depends on the dietary groups being compared, while DIABLO discriminates samples well but does not perform better than transcriptomic data alone. Conclusion The integration of associated “omics” datasets can help clarify the subtle signals observed in nutritional interventions. The performance of each integration tool is differently influenced by study design, size of the datasets, and sample size.
Collapse
Affiliation(s)
- Kathryn J Burton-Pimentel
- Federal Department of Economic Affairs, Education and Research EAER, Agroscope, Schwarzenburgstrasse 161, Bern, 3003, Switzerland
| | - Grégory Pimentel
- Federal Department of Economic Affairs, Education and Research EAER, Agroscope, Schwarzenburgstrasse 161, Bern, 3003, Switzerland
| | - Maria Hughes
- UCD Institute of Food and Health, School of Public Health, Physiotherapy, and Sports Science, University College Dublin, Belfield, Dublin 4, D04 C7X2, Ireland.,Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, Belfield, Dublin 4, Ireland.,Nutrigenomics Research Group, UCD Conway Institute and UCD Institute of Food and Health, School of Public Health, Physiotherapy and Sports Science, Belfield, Dublin 4, D04 V1W8, Ireland
| | - Charlotte Cjr Michielsen
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University and Research, P.O. Box 17, Wageningen, 6700 AA, The Netherlands
| | - Attia Fatima
- UCD Institute of Food and Health, School of Public Health, Physiotherapy, and Sports Science, University College Dublin, Belfield, Dublin 4, D04 C7X2, Ireland.,Nutrigenomics Research Group, UCD Conway Institute and UCD Institute of Food and Health, School of Public Health, Physiotherapy and Sports Science, Belfield, Dublin 4, D04 V1W8, Ireland
| | - Nathalie Vionnet
- Service of Endocrinology, Diabetes and Metabolism, Lausanne University Hospital, Lausanne, 1011, Switzerland
| | - Lydia A Afman
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University and Research, P.O. Box 17, Wageningen, 6700 AA, The Netherlands
| | - Helen M Roche
- UCD Institute of Food and Health, School of Public Health, Physiotherapy, and Sports Science, University College Dublin, Belfield, Dublin 4, D04 C7X2, Ireland.,Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, Belfield, Dublin 4, Ireland.,Nutrigenomics Research Group, UCD Conway Institute and UCD Institute of Food and Health, School of Public Health, Physiotherapy and Sports Science, Belfield, Dublin 4, D04 V1W8, Ireland.,Institute for Global Food Security, Queens University Belfast, Belfast, BT7 1NN, United Kingdom
| | - Lorraine Brennan
- UCD Institute of Food & Health, School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, D04 V1W8, Ireland
| | - Mark Ibberson
- Vital IT, Quartier UNIL-Sorge, Lausanne, 1015, Switzerland.,Swiss Institute of Bioinformatics, Quartier UNIL-Sorge, Lausanne, 1015, Switzerland
| | - Guy Vergères
- Federal Department of Economic Affairs, Education and Research EAER, Agroscope, Schwarzenburgstrasse 161, Bern, 3003, Switzerland
| |
Collapse
|
22
|
Whole blood co-expression modules associate with metabolic traits and type 2 diabetes: an IMI-DIRECT study. Genome Med 2020; 12:109. [PMID: 33261667 PMCID: PMC7708171 DOI: 10.1186/s13073-020-00806-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/11/2020] [Indexed: 01/04/2023] Open
Abstract
Background The rising prevalence of type 2 diabetes (T2D) poses a major global challenge. It remains unresolved to what extent transcriptomic signatures of metabolic dysregulation and T2D can be observed in easily accessible tissues such as blood. Additionally, large-scale human studies are required to further our understanding of the putative inflammatory component of insulin resistance and T2D. Here we used transcriptomics data from individuals with (n = 789) and without (n = 2127) T2D from the IMI-DIRECT cohorts to describe the co-expression structure of whole blood that mainly reflects processes and cell types of the immune system, and how it relates to metabolically relevant clinical traits and T2D. Methods Clusters of co-expressed genes were identified in the non-diabetic IMI-DIRECT cohort and evaluated with regard to stability, as well as preservation and rewiring in the cohort of individuals with T2D. We performed functional and immune cell signature enrichment analyses, and a genome-wide association study to describe the genetic regulation of the modules. Phenotypic and trans-omics associations of the transcriptomic modules were investigated across both IMI-DIRECT cohorts. Results We identified 55 whole blood co-expression modules, some of which clustered in larger super-modules. We identified a large number of associations between these transcriptomic modules and measures of insulin action and glucose tolerance. Some of the metabolically linked modules reflect neutrophil-lymphocyte ratio in blood while others are independent of white blood cell estimates, including a module of genes encoding neutrophil granule proteins with antibacterial properties for which the strongest associations with clinical traits and T2D status were observed. Through the integration of genetic and multi-omics data, we provide a holistic view of the regulation and molecular context of whole blood transcriptomic modules. We furthermore identified an overlap between genetic signals for T2D and co-expression modules involved in type II interferon signaling. Conclusions Our results offer a large-scale map of whole blood transcriptomic modules in the context of metabolic disease and point to novel biological candidates for future studies related to T2D.
Collapse
|
23
|
Kolberg L, Kerimov N, Peterson H, Alasoo K. Co-expression analysis reveals interpretable gene modules controlled by trans-acting genetic variants. eLife 2020; 9:e58705. [PMID: 32880574 PMCID: PMC7470823 DOI: 10.7554/elife.58705] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 08/20/2020] [Indexed: 12/16/2022] Open
Abstract
Understanding the causal processes that contribute to disease onset and progression is essential for developing novel therapies. Although trans-acting expression quantitative trait loci (trans-eQTLs) can directly reveal cellular processes modulated by disease variants, detecting trans-eQTLs remains challenging due to their small effect sizes. Here, we analysed gene expression and genotype data from six blood cell types from 226 to 710 individuals. We used co-expression modules inferred from gene expression data with five methods as traits in trans-eQTL analysis to limit multiple testing and improve interpretability. In addition to replicating three established associations, we discovered a novel trans-eQTL near SLC39A8 regulating a module of metallothionein genes in LPS-stimulated monocytes. Interestingly, this effect was mediated by a transient cis-eQTL present only in early LPS response and lost before the trans effect appeared. Our analyses highlight how co-expression combined with functional enrichment analysis improves the identification and prioritisation of trans-eQTLs when applied to emerging cell-type-specific datasets.
Collapse
Affiliation(s)
- Liis Kolberg
- Institute of Computer Science, University of TartuTartuEstonia
| | - Nurlan Kerimov
- Institute of Computer Science, University of TartuTartuEstonia
| | - Hedi Peterson
- Institute of Computer Science, University of TartuTartuEstonia
| | - Kaur Alasoo
- Institute of Computer Science, University of TartuTartuEstonia
| |
Collapse
|
24
|
Vuckovic D, Bao EL, Akbari P, Lareau CA, Mousas A, Jiang T, Chen MH, Raffield LM, Tardaguila M, Huffman JE, Ritchie SC, Megy K, Ponstingl H, Penkett CJ, Albers PK, Wigdor EM, Sakaue S, Moscati A, Manansala R, Lo KS, Qian H, Akiyama M, Bartz TM, Ben-Shlomo Y, Beswick A, Bork-Jensen J, Bottinger EP, Brody JA, van Rooij FJA, Chitrala KN, Wilson PWF, Choquet H, Danesh J, Di Angelantonio E, Dimou N, Ding J, Elliott P, Esko T, Evans MK, Felix SB, Floyd JS, Broer L, Grarup N, Guo MH, Guo Q, Greinacher A, Haessler J, Hansen T, Howson JMM, Huang W, Jorgenson E, Kacprowski T, Kähönen M, Kamatani Y, Kanai M, Karthikeyan S, Koskeridis F, Lange LA, Lehtimäki T, Linneberg A, Liu Y, Lyytikäinen LP, Manichaikul A, Matsuda K, Mohlke KL, Mononen N, Murakami Y, Nadkarni GN, Nikus K, Pankratz N, Pedersen O, Preuss M, Psaty BM, Raitakari OT, Rich SS, Rodriguez BAT, Rosen JD, Rotter JI, Schubert P, Spracklen CN, Surendran P, Tang H, Tardif JC, Ghanbari M, Völker U, Völzke H, Watkins NA, Weiss S, Cai N, Kundu K, Watt SB, Walter K, Zonderman AB, Cho K, Li Y, Loos RJF, Knight JC, Georges M, Stegle O, Evangelou E, et alVuckovic D, Bao EL, Akbari P, Lareau CA, Mousas A, Jiang T, Chen MH, Raffield LM, Tardaguila M, Huffman JE, Ritchie SC, Megy K, Ponstingl H, Penkett CJ, Albers PK, Wigdor EM, Sakaue S, Moscati A, Manansala R, Lo KS, Qian H, Akiyama M, Bartz TM, Ben-Shlomo Y, Beswick A, Bork-Jensen J, Bottinger EP, Brody JA, van Rooij FJA, Chitrala KN, Wilson PWF, Choquet H, Danesh J, Di Angelantonio E, Dimou N, Ding J, Elliott P, Esko T, Evans MK, Felix SB, Floyd JS, Broer L, Grarup N, Guo MH, Guo Q, Greinacher A, Haessler J, Hansen T, Howson JMM, Huang W, Jorgenson E, Kacprowski T, Kähönen M, Kamatani Y, Kanai M, Karthikeyan S, Koskeridis F, Lange LA, Lehtimäki T, Linneberg A, Liu Y, Lyytikäinen LP, Manichaikul A, Matsuda K, Mohlke KL, Mononen N, Murakami Y, Nadkarni GN, Nikus K, Pankratz N, Pedersen O, Preuss M, Psaty BM, Raitakari OT, Rich SS, Rodriguez BAT, Rosen JD, Rotter JI, Schubert P, Spracklen CN, Surendran P, Tang H, Tardif JC, Ghanbari M, Völker U, Völzke H, Watkins NA, Weiss S, Cai N, Kundu K, Watt SB, Walter K, Zonderman AB, Cho K, Li Y, Loos RJF, Knight JC, Georges M, Stegle O, Evangelou E, Okada Y, Roberts DJ, Inouye M, Johnson AD, Auer PL, Astle WJ, Reiner AP, Butterworth AS, Ouwehand WH, Lettre G, Sankaran VG, Soranzo N. The Polygenic and Monogenic Basis of Blood Traits and Diseases. Cell 2020; 182:1214-1231.e11. [PMID: 32888494 PMCID: PMC7482360 DOI: 10.1016/j.cell.2020.08.008] [Show More Authors] [Citation(s) in RCA: 419] [Impact Index Per Article: 83.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 06/29/2020] [Accepted: 08/03/2020] [Indexed: 12/13/2022]
Abstract
Blood cells play essential roles in human health, underpinning physiological processes such as immunity, oxygen transport, and clotting, which when perturbed cause a significant global health burden. Here we integrate data from UK Biobank and a large-scale international collaborative effort, including data for 563,085 European ancestry participants, and discover 5,106 new genetic variants independently associated with 29 blood cell phenotypes covering a range of variation impacting hematopoiesis. We holistically characterize the genetic architecture of hematopoiesis, assess the relevance of the omnigenic model to blood cell phenotypes, delineate relevant hematopoietic cell states influenced by regulatory genetic variants and gene networks, identify novel splice-altering variants mediating the associations, and assess the polygenic prediction potential for blood traits and clinical disorders at the interface of complex and Mendelian genetics. These results show the power of large-scale blood cell trait GWAS to interrogate clinically meaningful variants across a wide allelic spectrum of human variation.
Collapse
Affiliation(s)
- Dragana Vuckovic
- Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK; National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Erik L Bao
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA; Harvard-MIT Health Sciences and Technology, Harvard Medical School, Boston, MA, 02142, USA
| | - Parsa Akbari
- Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK; National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, University of Cambridge, Cambridge, CB1 8RN, UK; MRC Biostatistics Unit, University of Cambridge, Cambridge, CB2 0SR, UK; Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | - Caleb A Lareau
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Abdou Mousas
- Montreal Heart Institute, Montreal, Quebec, H1T 1C8, Canada
| | - Tao Jiang
- Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK; National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, CB2 0QQ, UK
| | - Ming-Huei Chen
- The Framingham Heart Study, National Heart, Lung and Blood Institute, Framingham, MA, 01702, USA; Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, Framingham, MA, 01702, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | | | - Jennifer E Huffman
- Center for Population Genomics, Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, 02130, USA
| | - Scott C Ritchie
- Department of Public Health and Primary Care, Cambridge Baker Systems Genomics Initiative, University of Cambridge, Cambridge, CB1 8RN, UK; Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, VIC 3004, Australia; Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK; British Heart Foundation Centre of Excellence, Division of Cardiovascular Medicine, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK; National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, CB2 0QQ, UK
| | - Karyn Megy
- Department of Haematology, University of Cambridge, Cambridge, CB2 0PT, UK; National Institute for Health Research (NIHR) BioResource, Cambridge University Hospitals, Cambridge, CB2 0PT, UK; National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Cambridge, CB2 0PT, UK
| | - Hannes Ponstingl
- Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | - Christopher J Penkett
- National Institute for Health Research (NIHR) BioResource, Cambridge University Hospitals, Cambridge, CB2 0PT, UK; Department of Haematology, University of Cambridge, Cambridge, CB2 0PT, UK
| | - Patrick K Albers
- Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | - Emilie M Wigdor
- Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | - Saori Sakaue
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan; Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan
| | - Arden Moscati
- Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY, 10029, USA
| | - Regina Manansala
- Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI, 53201, USA
| | - Ken Sin Lo
- Montreal Heart Institute, Montreal, Quebec, H1T 1C8, Canada
| | - Huijun Qian
- Department of Statistics and Operation Research, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Masato Akiyama
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan; Department of Ocular Pathology and Imaging Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8581, Japan
| | - Traci M Bartz
- Department of Biostatistics, University of Washington, Seattle, WA, 98101, USA
| | - Yoav Ben-Shlomo
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 1QU, UK
| | - Andrew Beswick
- Translational Health Sciences, Musculoskeletal Research Unit, Bristol Medical School, University of Bristol, Bristol, BS10 5NB, UK
| | - Jette Bork-Jensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Erwin P Bottinger
- Hasso-Plattner-Institut, Universität Potsdam, Potsdam, 14469, Germany; Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY, 10029, USA
| | - Jennifer A Brody
- Department of Medicine, University of Washington, Seattle, WA, 98101, USA
| | - Frank J A van Rooij
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, Rotterdam, 3015 GE, the Netherlands
| | - Kumaraswamy N Chitrala
- Laboratory of Epidemiology and Population Science, National Institute on Aging/NIH, Baltimore, MD, 21224, USA
| | | | - Hélène Choquet
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, 94612, USA
| | - John Danesh
- Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK; Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, CB10 1SA, UK; National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, University of Cambridge, Cambridge, CB1 8RN, UK; Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK; National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, CB2 0QQ, UK; British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Emanuele Di Angelantonio
- Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK; Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, CB10 1SA, UK; National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, University of Cambridge, Cambridge, CB1 8RN, UK; Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK; National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, CB2 0QQ, UK; British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Niki Dimou
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, 69008, France; Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, 45110, Greece
| | - Jingzhong Ding
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, Imperial College London, London, W2 1PG, UK; Imperial Biomedical Research Centre, Imperial College London and Imperial College NHS Healthcare Trust, London, W2 1NY, UK; Medical Research Council Centre for Environment and Health, Imperial College London, London, W2 1PG, UK; UK Dementia Research Institute, Imperial College London, London, WC1E 6BT, UK; Health Data Research UK London, London, W2 1PG, UK
| | - Tõnu Esko
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Science, National Institute on Aging/NIH, Baltimore, MD, 21224, USA
| | - Stephan B Felix
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, 17475, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, 17475, Germany
| | - James S Floyd
- Department of Medicine, University of Washington, Seattle, WA, 98101, USA; Department of Epidemiology, University of Washington, Seattle, WA, 98101, USA
| | - Linda Broer
- Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, 3015 GE, the Netherlands
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Michael H Guo
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA; Department of Neurology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Qi Guo
- Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Andreas Greinacher
- Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, 17475, Germany
| | - Jeff Haessler
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, 98101, USA
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Joanna M M Howson
- Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK; National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, CB2 0QQ, UK; Novo Nordisk Research Centre Oxford, Oxford, OX3 7FZ, UK
| | - Wei Huang
- Department of Genetics, Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center and Shanghai Industrial Technology Institute (SITI), Shanghai, 201203, China
| | - Eric Jorgenson
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, 94612, USA
| | - Tim Kacprowski
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, 17475, Germany; Chair of Experimental Bioinformatics, Research Group Computational Systems Medicine, Technical University of Munich, Freising-Weihenstephan, 85354, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, 17475, Germany
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere, 33521, Finland; Department of Clinical Physiology, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, 33014, Finland
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan; Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Masahiro Kanai
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Savita Karthikeyan
- Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Fotios Koskeridis
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, 45110, Greece
| | - Leslie A Lange
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, 33520, Finland; Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, 33014, Finland
| | - Allan Linneberg
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Frederiksberg, 2000, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Yongmei Liu
- Department of Medicine, Division of Cardiology, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, 27701, USA
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, 33520, Finland; Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, 33014, Finland
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, 22903, USA
| | - Koichi Matsuda
- Department of Computational Biology and Medical Sciences, Graduate school of Frontier Sciences, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Nina Mononen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, 33520, Finland; Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, 33014, Finland
| | - Yoshinori Murakami
- Division of Molecular Pathology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Girish N Nadkarni
- Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY, 10029, USA
| | - Kjell Nikus
- Department of Cardiology, Heart Center, Tampere University Hospital, Tampere, 33521, Finland; Department of Cardiology, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, 33014, Finland
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Michael Preuss
- Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY, 10029, USA
| | - Bruce M Psaty
- Departments of Epidemiology, University of Washington, Seattle, WA, 98101, USA; Department of Medicine, University of Washington, Seattle, WA, 98101, USA; Department of Health Services, University of Washington, Seattle, WA, 98101, USA; Kaiser Permanente Washington Health Research Institute, Seattle, WA, 98101, USA
| | - Olli T Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, 20521, Finland; Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, 20521, Finland; Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, 20521, Finland
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, 22903, USA
| | - Benjamin A T Rodriguez
- The Framingham Heart Study, National Heart, Lung and Blood Institute, Framingham, MA, 01702, USA; Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, Framingham, MA, 01702, USA
| | - Jonathan D Rosen
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Jerome I Rotter
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation (formerly Los Angeles Biomedical Research Institute) at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Petra Schubert
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, 02130, USA
| | - Cassandra N Spracklen
- Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA; Department of Biostatistics and Epidemiology, University of Massachusetts-Amherst, Amherst, MA, 01002, USA
| | - Praveen Surendran
- Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK; British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, CB1 8RN, UK; Health Data Research UK Cambridge, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK; Department of Public Health and Primary Care, Rutherford Fund Fellow, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Hua Tang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jean-Claude Tardif
- Montreal Heart Institute, Montreal, Quebec, H1T 1C8, Canada; Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, H3T 1J4, Canada
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, Rotterdam, 3015 GE, the Netherlands; Department of Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, 9177948564, Iran
| | - Uwe Völker
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, 17475, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, 17475, Germany
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, 17475, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, 17475, Germany
| | - Nicholas A Watkins
- National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Cambridge, CB2 0PT, UK
| | - Stefan Weiss
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, 17475, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, 17475, Germany
| | - Na Cai
- Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | - Kousik Kundu
- Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK; Department of Haematology, University of Cambridge, Cambridge, CB2 0PT, UK
| | - Stephen B Watt
- Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | - Klaudia Walter
- Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Science, National Institute on Aging/NIH, Baltimore, MD, 21224, USA
| | - Kelly Cho
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, 02130, USA; Department of Medicine, Division on Aging, Brigham and Women's Hospital, Boston, MA, 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Yun Li
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, 27599, USA; Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA; Department of Computer Science, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Ruth J F Loos
- Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY, 10029, USA
| | - Julian C Knight
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Michel Georges
- Unit of Animal Genomics, GIGA-R & Faculty of Veterinary Medicine, University of Liège, Liège, B-4000, Belgium
| | - Oliver Stegle
- European Bioinformatics Institute, European Molecular Biology Laboratory, Hinxton, CB10 1SA, UK
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, Imperial College London, London, W2 1PG, UK; Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, 45110, Greece
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan; Laboratory of Statistical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - David J Roberts
- BRC Haematology Theme and Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK; NHSBT Blood and Transplant - Oxford Center, John Radcliffe Hospital, Oxford, OX3 9BQ, UK
| | - Michael Inouye
- Department of Public Health and Primary Care, Cambridge Baker Systems Genomics Initiative, University of Cambridge, Cambridge, CB1 8RN, UK; Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, VIC 3004, Australia; Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK; British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, CB1 8RN, UK; National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, CB2 0QQ, UK; Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, CB10 1SA, UK; The Alan Turing Institute, London, NW1 2DB, UK
| | - Andrew D Johnson
- The Framingham Heart Study, National Heart, Lung and Blood Institute, Framingham, MA, 01702, USA; Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, Framingham, MA, 01702, USA
| | - Paul L Auer
- Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI, 53201, USA
| | - William J Astle
- MRC Biostatistics Unit, University of Cambridge, Cambridge, CB2 0SR, UK; National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, University of Cambridge, Cambridge, CB1 8RN, UK; National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Cambridge, CB2 0PT, UK
| | - Alexander P Reiner
- Department of Epidemiology, University of Washington, Seattle, WA, 98109, USA
| | - Adam S Butterworth
- Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK; Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, CB10 1SA, UK; National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, University of Cambridge, Cambridge, CB1 8RN, UK; Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK; National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, CB2 0QQ, UK; British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Willem H Ouwehand
- Department of Haematology, University of Cambridge, Cambridge, CB2 0PT, UK; National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Cambridge, CB2 0PT, UK; Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK; National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, University of Cambridge, Cambridge, CB1 8RN, UK; British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Guillaume Lettre
- Montreal Heart Institute, Montreal, Quebec, H1T 1C8, Canada; Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, H3T 1J4, Canada
| | - Vijay G Sankaran
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| | - Nicole Soranzo
- Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK; National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, University of Cambridge, Cambridge, CB1 8RN, UK; British Heart Foundation Centre of Excellence, Division of Cardiovascular Medicine, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK; Department of Haematology, University of Cambridge, Cambridge, CB2 0PT, UK.
| |
Collapse
|
25
|
Vuckovic D, Bao EL, Akbari P, Lareau CA, Mousas A, Jiang T, Chen MH, Raffield LM, Tardaguila M, Huffman JE, Ritchie SC, Megy K, Ponstingl H, Penkett CJ, Albers PK, Wigdor EM, Sakaue S, Moscati A, Manansala R, Lo KS, Qian H, Akiyama M, Bartz TM, Ben-Shlomo Y, Beswick A, Bork-Jensen J, Bottinger EP, Brody JA, van Rooij FJA, Chitrala KN, Wilson PWF, Choquet H, Danesh J, Di Angelantonio E, Dimou N, Ding J, Elliott P, Esko T, Evans MK, Felix SB, Floyd JS, Broer L, Grarup N, Guo MH, Guo Q, Greinacher A, Haessler J, Hansen T, Howson JMM, Huang W, Jorgenson E, Kacprowski T, Kähönen M, Kamatani Y, Kanai M, Karthikeyan S, Koskeridis F, Lange LA, Lehtimäki T, Linneberg A, Liu Y, Lyytikäinen LP, Manichaikul A, Matsuda K, Mohlke KL, Mononen N, Murakami Y, Nadkarni GN, Nikus K, Pankratz N, Pedersen O, Preuss M, Psaty BM, Raitakari OT, Rich SS, Rodriguez BAT, Rosen JD, Rotter JI, Schubert P, Spracklen CN, Surendran P, Tang H, Tardif JC, Ghanbari M, Völker U, Völzke H, Watkins NA, Weiss S, Cai N, Kundu K, Watt SB, Walter K, Zonderman AB, Cho K, Li Y, Loos RJF, Knight JC, Georges M, Stegle O, Evangelou E, et alVuckovic D, Bao EL, Akbari P, Lareau CA, Mousas A, Jiang T, Chen MH, Raffield LM, Tardaguila M, Huffman JE, Ritchie SC, Megy K, Ponstingl H, Penkett CJ, Albers PK, Wigdor EM, Sakaue S, Moscati A, Manansala R, Lo KS, Qian H, Akiyama M, Bartz TM, Ben-Shlomo Y, Beswick A, Bork-Jensen J, Bottinger EP, Brody JA, van Rooij FJA, Chitrala KN, Wilson PWF, Choquet H, Danesh J, Di Angelantonio E, Dimou N, Ding J, Elliott P, Esko T, Evans MK, Felix SB, Floyd JS, Broer L, Grarup N, Guo MH, Guo Q, Greinacher A, Haessler J, Hansen T, Howson JMM, Huang W, Jorgenson E, Kacprowski T, Kähönen M, Kamatani Y, Kanai M, Karthikeyan S, Koskeridis F, Lange LA, Lehtimäki T, Linneberg A, Liu Y, Lyytikäinen LP, Manichaikul A, Matsuda K, Mohlke KL, Mononen N, Murakami Y, Nadkarni GN, Nikus K, Pankratz N, Pedersen O, Preuss M, Psaty BM, Raitakari OT, Rich SS, Rodriguez BAT, Rosen JD, Rotter JI, Schubert P, Spracklen CN, Surendran P, Tang H, Tardif JC, Ghanbari M, Völker U, Völzke H, Watkins NA, Weiss S, Cai N, Kundu K, Watt SB, Walter K, Zonderman AB, Cho K, Li Y, Loos RJF, Knight JC, Georges M, Stegle O, Evangelou E, Okada Y, Roberts DJ, Inouye M, Johnson AD, Auer PL, Astle WJ, Reiner AP, Butterworth AS, Ouwehand WH, Lettre G, Sankaran VG, Soranzo N. The Polygenic and Monogenic Basis of Blood Traits and Diseases. Cell 2020. [PMID: 32888494 DOI: 10.1016/j.cell.2020.08.008ll] [Show More Authors] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
Blood cells play essential roles in human health, underpinning physiological processes such as immunity, oxygen transport, and clotting, which when perturbed cause a significant global health burden. Here we integrate data from UK Biobank and a large-scale international collaborative effort, including data for 563,085 European ancestry participants, and discover 5,106 new genetic variants independently associated with 29 blood cell phenotypes covering a range of variation impacting hematopoiesis. We holistically characterize the genetic architecture of hematopoiesis, assess the relevance of the omnigenic model to blood cell phenotypes, delineate relevant hematopoietic cell states influenced by regulatory genetic variants and gene networks, identify novel splice-altering variants mediating the associations, and assess the polygenic prediction potential for blood traits and clinical disorders at the interface of complex and Mendelian genetics. These results show the power of large-scale blood cell trait GWAS to interrogate clinically meaningful variants across a wide allelic spectrum of human variation.
Collapse
Affiliation(s)
- Dragana Vuckovic
- Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK; National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Erik L Bao
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA; Harvard-MIT Health Sciences and Technology, Harvard Medical School, Boston, MA, 02142, USA
| | - Parsa Akbari
- Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK; National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, University of Cambridge, Cambridge, CB1 8RN, UK; MRC Biostatistics Unit, University of Cambridge, Cambridge, CB2 0SR, UK; Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | - Caleb A Lareau
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Abdou Mousas
- Montreal Heart Institute, Montreal, Quebec, H1T 1C8, Canada
| | - Tao Jiang
- Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK; National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, CB2 0QQ, UK
| | - Ming-Huei Chen
- The Framingham Heart Study, National Heart, Lung and Blood Institute, Framingham, MA, 01702, USA; Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, Framingham, MA, 01702, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | | | - Jennifer E Huffman
- Center for Population Genomics, Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, 02130, USA
| | - Scott C Ritchie
- Department of Public Health and Primary Care, Cambridge Baker Systems Genomics Initiative, University of Cambridge, Cambridge, CB1 8RN, UK; Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, VIC 3004, Australia; Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK; British Heart Foundation Centre of Excellence, Division of Cardiovascular Medicine, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK; National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, CB2 0QQ, UK
| | - Karyn Megy
- Department of Haematology, University of Cambridge, Cambridge, CB2 0PT, UK; National Institute for Health Research (NIHR) BioResource, Cambridge University Hospitals, Cambridge, CB2 0PT, UK; National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Cambridge, CB2 0PT, UK
| | - Hannes Ponstingl
- Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | - Christopher J Penkett
- National Institute for Health Research (NIHR) BioResource, Cambridge University Hospitals, Cambridge, CB2 0PT, UK; Department of Haematology, University of Cambridge, Cambridge, CB2 0PT, UK
| | - Patrick K Albers
- Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | - Emilie M Wigdor
- Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | - Saori Sakaue
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan; Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan
| | - Arden Moscati
- Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY, 10029, USA
| | - Regina Manansala
- Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI, 53201, USA
| | - Ken Sin Lo
- Montreal Heart Institute, Montreal, Quebec, H1T 1C8, Canada
| | - Huijun Qian
- Department of Statistics and Operation Research, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Masato Akiyama
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan; Department of Ocular Pathology and Imaging Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8581, Japan
| | - Traci M Bartz
- Department of Biostatistics, University of Washington, Seattle, WA, 98101, USA
| | - Yoav Ben-Shlomo
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 1QU, UK
| | - Andrew Beswick
- Translational Health Sciences, Musculoskeletal Research Unit, Bristol Medical School, University of Bristol, Bristol, BS10 5NB, UK
| | - Jette Bork-Jensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Erwin P Bottinger
- Hasso-Plattner-Institut, Universität Potsdam, Potsdam, 14469, Germany; Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY, 10029, USA
| | - Jennifer A Brody
- Department of Medicine, University of Washington, Seattle, WA, 98101, USA
| | - Frank J A van Rooij
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, Rotterdam, 3015 GE, the Netherlands
| | - Kumaraswamy N Chitrala
- Laboratory of Epidemiology and Population Science, National Institute on Aging/NIH, Baltimore, MD, 21224, USA
| | | | - Hélène Choquet
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, 94612, USA
| | - John Danesh
- Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK; Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, CB10 1SA, UK; National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, University of Cambridge, Cambridge, CB1 8RN, UK; Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK; National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, CB2 0QQ, UK; British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Emanuele Di Angelantonio
- Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK; Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, CB10 1SA, UK; National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, University of Cambridge, Cambridge, CB1 8RN, UK; Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK; National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, CB2 0QQ, UK; British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Niki Dimou
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, 69008, France; Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, 45110, Greece
| | - Jingzhong Ding
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27101, USA
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, Imperial College London, London, W2 1PG, UK; Imperial Biomedical Research Centre, Imperial College London and Imperial College NHS Healthcare Trust, London, W2 1NY, UK; Medical Research Council Centre for Environment and Health, Imperial College London, London, W2 1PG, UK; UK Dementia Research Institute, Imperial College London, London, WC1E 6BT, UK; Health Data Research UK London, London, W2 1PG, UK
| | - Tõnu Esko
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Science, National Institute on Aging/NIH, Baltimore, MD, 21224, USA
| | - Stephan B Felix
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, 17475, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, 17475, Germany
| | - James S Floyd
- Department of Medicine, University of Washington, Seattle, WA, 98101, USA; Department of Epidemiology, University of Washington, Seattle, WA, 98101, USA
| | - Linda Broer
- Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, 3015 GE, the Netherlands
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Michael H Guo
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA; Department of Neurology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Qi Guo
- Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Andreas Greinacher
- Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, 17475, Germany
| | - Jeff Haessler
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, 98101, USA
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Joanna M M Howson
- Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK; National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, CB2 0QQ, UK; Novo Nordisk Research Centre Oxford, Oxford, OX3 7FZ, UK
| | - Wei Huang
- Department of Genetics, Shanghai-MOST Key Laboratory of Health and Disease Genomics, Chinese National Human Genome Center and Shanghai Industrial Technology Institute (SITI), Shanghai, 201203, China
| | - Eric Jorgenson
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, 94612, USA
| | - Tim Kacprowski
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, 17475, Germany; Chair of Experimental Bioinformatics, Research Group Computational Systems Medicine, Technical University of Munich, Freising-Weihenstephan, 85354, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, 17475, Germany
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere, 33521, Finland; Department of Clinical Physiology, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, 33014, Finland
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan; Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Masahiro Kanai
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, 230-0045, Japan; Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Savita Karthikeyan
- Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Fotios Koskeridis
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, 45110, Greece
| | - Leslie A Lange
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, 33520, Finland; Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, 33014, Finland
| | - Allan Linneberg
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Frederiksberg, 2000, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Yongmei Liu
- Department of Medicine, Division of Cardiology, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, 27701, USA
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, 33520, Finland; Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, 33014, Finland
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, 22903, USA
| | - Koichi Matsuda
- Department of Computational Biology and Medical Sciences, Graduate school of Frontier Sciences, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Nina Mononen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, 33520, Finland; Department of Clinical Chemistry, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, 33014, Finland
| | - Yoshinori Murakami
- Division of Molecular Pathology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Girish N Nadkarni
- Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY, 10029, USA
| | - Kjell Nikus
- Department of Cardiology, Heart Center, Tampere University Hospital, Tampere, 33521, Finland; Department of Cardiology, Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, 33014, Finland
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Michael Preuss
- Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY, 10029, USA
| | - Bruce M Psaty
- Departments of Epidemiology, University of Washington, Seattle, WA, 98101, USA; Department of Medicine, University of Washington, Seattle, WA, 98101, USA; Department of Health Services, University of Washington, Seattle, WA, 98101, USA; Kaiser Permanente Washington Health Research Institute, Seattle, WA, 98101, USA
| | - Olli T Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, 20521, Finland; Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, 20521, Finland; Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, 20521, Finland
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, 22903, USA
| | - Benjamin A T Rodriguez
- The Framingham Heart Study, National Heart, Lung and Blood Institute, Framingham, MA, 01702, USA; Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, Framingham, MA, 01702, USA
| | - Jonathan D Rosen
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Jerome I Rotter
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation (formerly Los Angeles Biomedical Research Institute) at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Petra Schubert
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, 02130, USA
| | - Cassandra N Spracklen
- Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA; Department of Biostatistics and Epidemiology, University of Massachusetts-Amherst, Amherst, MA, 01002, USA
| | - Praveen Surendran
- Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK; British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, CB1 8RN, UK; Health Data Research UK Cambridge, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK; Department of Public Health and Primary Care, Rutherford Fund Fellow, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Hua Tang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jean-Claude Tardif
- Montreal Heart Institute, Montreal, Quebec, H1T 1C8, Canada; Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, H3T 1J4, Canada
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, Rotterdam, 3015 GE, the Netherlands; Department of Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, 9177948564, Iran
| | - Uwe Völker
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, 17475, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, 17475, Germany
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, 17475, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, 17475, Germany
| | - Nicholas A Watkins
- National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Cambridge, CB2 0PT, UK
| | - Stefan Weiss
- Interfaculty Institute of Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, 17475, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, 17475, Germany
| | - Na Cai
- Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | - Kousik Kundu
- Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK; Department of Haematology, University of Cambridge, Cambridge, CB2 0PT, UK
| | - Stephen B Watt
- Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | - Klaudia Walter
- Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Science, National Institute on Aging/NIH, Baltimore, MD, 21224, USA
| | - Kelly Cho
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA, 02130, USA; Department of Medicine, Division on Aging, Brigham and Women's Hospital, Boston, MA, 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Yun Li
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, 27599, USA; Department of Genetics, University of North Carolina, Chapel Hill, NC, 27599, USA; Department of Computer Science, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Ruth J F Loos
- Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY, 10029, USA
| | - Julian C Knight
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Michel Georges
- Unit of Animal Genomics, GIGA-R & Faculty of Veterinary Medicine, University of Liège, Liège, B-4000, Belgium
| | - Oliver Stegle
- European Bioinformatics Institute, European Molecular Biology Laboratory, Hinxton, CB10 1SA, UK
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, Imperial College London, London, W2 1PG, UK; Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, 45110, Greece
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan; Laboratory of Statistical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - David J Roberts
- BRC Haematology Theme and Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK; NHSBT Blood and Transplant - Oxford Center, John Radcliffe Hospital, Oxford, OX3 9BQ, UK
| | - Michael Inouye
- Department of Public Health and Primary Care, Cambridge Baker Systems Genomics Initiative, University of Cambridge, Cambridge, CB1 8RN, UK; Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Victoria, VIC 3004, Australia; Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK; British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, CB1 8RN, UK; National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, CB2 0QQ, UK; Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, CB10 1SA, UK; The Alan Turing Institute, London, NW1 2DB, UK
| | - Andrew D Johnson
- The Framingham Heart Study, National Heart, Lung and Blood Institute, Framingham, MA, 01702, USA; Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, Framingham, MA, 01702, USA
| | - Paul L Auer
- Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI, 53201, USA
| | - William J Astle
- MRC Biostatistics Unit, University of Cambridge, Cambridge, CB2 0SR, UK; National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, University of Cambridge, Cambridge, CB1 8RN, UK; National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Cambridge, CB2 0PT, UK
| | - Alexander P Reiner
- Department of Epidemiology, University of Washington, Seattle, WA, 98109, USA
| | - Adam S Butterworth
- Department of Public Health and Primary Care, British Heart Foundation Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, CB1 8RN, UK; Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, CB10 1SA, UK; National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, University of Cambridge, Cambridge, CB1 8RN, UK; Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK; National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, CB2 0QQ, UK; British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Willem H Ouwehand
- Department of Haematology, University of Cambridge, Cambridge, CB2 0PT, UK; National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Cambridge, CB2 0PT, UK; Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK; National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, University of Cambridge, Cambridge, CB1 8RN, UK; British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Guillaume Lettre
- Montreal Heart Institute, Montreal, Quebec, H1T 1C8, Canada; Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, H3T 1J4, Canada
| | - Vijay G Sankaran
- Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| | - Nicole Soranzo
- Human Genetics, Wellcome Sanger Institute, Hinxton, CB10 1SA, UK; National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Donor Health and Genomics, University of Cambridge, Cambridge, CB1 8RN, UK; British Heart Foundation Centre of Excellence, Division of Cardiovascular Medicine, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK; Department of Haematology, University of Cambridge, Cambridge, CB2 0PT, UK.
| |
Collapse
|
26
|
Waddington KE, Papadaki A, Coelewij L, Adriani M, Nytrova P, Kubala Havrdova E, Fogdell-Hahn A, Farrell R, Dönnes P, Pineda-Torra I, Jury EC. Using Serum Metabolomics to Predict Development of Anti-drug Antibodies in Multiple Sclerosis Patients Treated With IFNβ. Front Immunol 2020; 11:1527. [PMID: 32765529 PMCID: PMC7380268 DOI: 10.3389/fimmu.2020.01527] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022] Open
Abstract
Background: Neutralizing anti-drug antibodies (ADA) can greatly reduce the efficacy of biopharmaceuticals used to treat patients with multiple sclerosis (MS). However, the biological factors pre-disposing an individual to develop ADA are poorly characterized. Thus, there is an unmet clinical need for biomarkers to predict the development of immunogenicity, and subsequent treatment failure. Up to 35% of MS patients treated with beta interferons (IFNβ) develop ADA. Here we use machine learning to predict immunogenicity against IFNβ utilizing serum metabolomics data. Methods: Serum samples were collected from 89 MS patients as part of the ABIRISK consortium-a multi-center prospective study of ADA development. Metabolites and ADA were quantified prior to and after IFNβ treatment. Thirty patients became ADA positive during the first year of treatment (ADA+). We tested the efficacy of six binary classification models using 10-fold cross validation; k-nearest neighbors, decision tree, random forest, support vector machine and lasso (Least Absolute Shrinkage and Selection Operator) logistic regression with and without interactions. Results: We were able to predict future immunogenicity from baseline metabolomics data. Lasso logistic regression with/without interactions and support vector machines were the most successful at identifying ADA+ or ADA- cases, respectively. Furthermore, patients who become ADA+ had a distinct metabolic response to IFNβ in the first 3 months, with 29 differentially regulated metabolites. Machine learning algorithms could also predict ADA status based on metabolite concentrations at 3 months. Lasso logistic regressions had the greatest proportion of correct classifications [F1 score (accuracy measure) = 0.808, specificity = 0.913]. Finally, we hypothesized that serum lipids could contribute to ADA development by altering immune-cell lipid rafts. This was supported by experimental evidence demonstrating that, prior to IFNβ exposure, lipid raft-associated lipids were differentially expressed between MS patients who became ADA+ or remained ADA-. Conclusion: Serum metabolites are a promising biomarker for prediction of ADA development in MS patients treated with IFNβ, and could provide novel insight into mechanisms of immunogenicity.
Collapse
Affiliation(s)
- Kirsty E. Waddington
- Centre for Rheumatology, University College London, London, United Kingdom
- Centre for Cardiometabolic and Vascular Medicine, University College London, London, United Kingdom
| | - Artemis Papadaki
- Centre for Rheumatology, University College London, London, United Kingdom
| | - Leda Coelewij
- Centre for Rheumatology, University College London, London, United Kingdom
- Centre for Cardiometabolic and Vascular Medicine, University College London, London, United Kingdom
| | - Marsilio Adriani
- Centre for Rheumatology, University College London, London, United Kingdom
| | - Petra Nytrova
- Department of Neurology and Centre of Clinical Neuroscience, General University Hospital and First Faculty of Medicine, Charles University in Prague, Prague, Czechia
| | - Eva Kubala Havrdova
- Department of Neurology and Centre of Clinical Neuroscience, General University Hospital and First Faculty of Medicine, Charles University in Prague, Prague, Czechia
| | - Anna Fogdell-Hahn
- Department of Clinical Neuroscience, Center for Molecular Medicine (CMM), Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | - Rachel Farrell
- Department of Neuroinflammation, University College London, Institute of Neurology and National Hospital of Neurology and Neurosurgery, London, United Kingdom
| | - Pierre Dönnes
- Centre for Rheumatology, University College London, London, United Kingdom
- Scicross AB, Skövde, Sweden
| | - Inés Pineda-Torra
- Centre for Cardiometabolic and Vascular Medicine, University College London, London, United Kingdom
| | - Elizabeth C. Jury
- Centre for Rheumatology, University College London, London, United Kingdom
| |
Collapse
|
27
|
Reustle A, Di Marco M, Meyerhoff C, Nelde A, Walz JS, Winter S, Kandabarau S, Büttner F, Haag M, Backert L, Kowalewski DJ, Rausch S, Hennenlotter J, Stühler V, Scharpf M, Fend F, Stenzl A, Rammensee HG, Bedke J, Stevanović S, Schwab M, Schaeffeler E. Integrative -omics and HLA-ligandomics analysis to identify novel drug targets for ccRCC immunotherapy. Genome Med 2020; 12:32. [PMID: 32228647 PMCID: PMC7106651 DOI: 10.1186/s13073-020-00731-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 03/12/2020] [Indexed: 12/24/2022] Open
Abstract
Background Clear cell renal cell carcinoma (ccRCC) is the dominant subtype of renal cancer. With currently available therapies, cure of advanced and metastatic ccRCC is achieved only in rare cases. Here, we developed a workflow integrating different -omics technologies to identify ccRCC-specific HLA-presented peptides as potential drug targets for ccRCC immunotherapy. Methods We analyzed HLA-presented peptides by MS-based ligandomics of 55 ccRCC tumors (cohort 1), paired non-tumor renal tissues, and 158 benign tissues from other organs. Pathways enriched in ccRCC compared to its cell type of origin were identified by transcriptome and gene set enrichment analyses in 51 tumor tissues of the same cohort. To retrieve a list of candidate targets with involvement in ccRCC pathogenesis, ccRCC-specific pathway genes were intersected with the source genes of tumor-exclusive peptides. The candidates were validated in an independent cohort from The Cancer Genome Atlas (TCGA KIRC, n = 452). DNA methylation (TCGA KIRC, n = 273), somatic mutations (TCGA KIRC, n = 392), and gene ontology (GO) and correlations with tumor metabolites (cohort 1, n = 30) and immune-oncological markers (cohort 1, n = 37) were analyzed to characterize regulatory and functional involvements. CD8+ T cell priming assays were used to identify immunogenic peptides. The candidate gene EGLN3 was functionally investigated in cell culture. Results A total of 34,226 HLA class I- and 19,325 class II-presented peptides were identified in ccRCC tissue, of which 443 class I and 203 class II peptides were ccRCC-specific and presented in ≥ 3 tumors. One hundred eighty-five of the 499 corresponding source genes were involved in pathways activated by ccRCC tumors. After validation in the independent cohort from TCGA, 113 final candidate genes remained. Candidates were involved in extracellular matrix organization, hypoxic signaling, immune processes, and others. Nine of the 12 peptides assessed by immunogenicity analysis were able to activate naïve CD8+ T cells, including peptides derived from EGLN3. Functional analysis of EGLN3 revealed possible tumor-promoting functions. Conclusions Integration of HLA ligandomics, transcriptomics, genetic, and epigenetic data leads to the identification of novel functionally relevant therapeutic targets for ccRCC immunotherapy. Validation of the identified targets is recommended to expand the treatment landscape of ccRCC.
Collapse
Affiliation(s)
- Anna Reustle
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Moreno Di Marco
- Department of Immunology, Institute for Cell Biology, University of Tuebingen, Tuebingen, Germany
| | - Carolin Meyerhoff
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Annika Nelde
- Department of Immunology, Institute for Cell Biology, University of Tuebingen, Tuebingen, Germany.,Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), University Hospital Tuebingen, Tuebingen, Germany.,German Cancer Consortium (DKTK), Partner Site Tuebingen, Tuebingen, Germany
| | - Juliane S Walz
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), University Hospital Tuebingen, Tuebingen, Germany.,German Cancer Consortium (DKTK), Partner Site Tuebingen, Tuebingen, Germany.,iFIT Cluster of Excellence (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany
| | - Stefan Winter
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Siahei Kandabarau
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Florian Büttner
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Mathias Haag
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Linus Backert
- Department of Immunology, Institute for Cell Biology, University of Tuebingen, Tuebingen, Germany
| | - Daniel J Kowalewski
- Department of Immunology, Institute for Cell Biology, University of Tuebingen, Tuebingen, Germany
| | - Steffen Rausch
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany
| | - Jörg Hennenlotter
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany
| | - Viktoria Stühler
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany
| | - Marcus Scharpf
- Institute of Pathology and Neuropathology, University Hospital Tuebingen, Tuebingen, Germany
| | - Falko Fend
- Institute of Pathology and Neuropathology, University Hospital Tuebingen, Tuebingen, Germany
| | - Arnulf Stenzl
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany
| | - Hans-Georg Rammensee
- Department of Immunology, Institute for Cell Biology, University of Tuebingen, Tuebingen, Germany.,German Cancer Consortium (DKTK), Partner Site Tuebingen, Tuebingen, Germany.,iFIT Cluster of Excellence (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany
| | - Jens Bedke
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany
| | - Stefan Stevanović
- Department of Immunology, Institute for Cell Biology, University of Tuebingen, Tuebingen, Germany.,German Cancer Consortium (DKTK), Partner Site Tuebingen, Tuebingen, Germany.,iFIT Cluster of Excellence (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany. .,University of Tuebingen, Tuebingen, Germany. .,German Cancer Consortium (DKTK), Partner Site Tuebingen, Tuebingen, Germany. .,iFIT Cluster of Excellence (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany. .,Departments of Clinical Pharmacology, Pharmacy and Biochemistry, University of Tuebingen, Tuebingen, Germany.
| | - Elke Schaeffeler
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany.,iFIT Cluster of Excellence (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
28
|
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
|
29
|
Elevated serum alpha-1 antitrypsin is a major component of GlycA-associated risk for future morbidity and mortality. PLoS One 2019; 14:e0223692. [PMID: 31644575 PMCID: PMC6808431 DOI: 10.1371/journal.pone.0223692] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 09/25/2019] [Indexed: 12/14/2022] Open
Abstract
Background GlycA is a nuclear magnetic resonance (NMR) spectroscopy biomarker that predicts risk of disease from myriad causes. It is heterogeneous; arising from five circulating glycoproteins with dynamic concentrations: alpha-1 antitrypsin (AAT), alpha-1-acid glycoprotein (AGP), haptoglobin (HP), transferrin (TF), and alpha-1-antichymotrypsin (AACT). The contributions of each glycoprotein to the disease and mortality risks predicted by GlycA remain unknown. Methods We trained imputation models for AAT, AGP, HP, and TF from NMR metabolite measurements in 626 adults from a population cohort with matched NMR and immunoassay data. Levels of AAT, AGP, and HP were estimated in 11,861 adults from two population cohorts with eight years of follow-up, then each biomarker was tested for association with all common endpoints. Whole blood gene expression data was used to identify cellular processes associated with elevated AAT. Results Accurate imputation models were obtained for AAT, AGP, and HP but not for TF. While AGP had the strongest correlation with GlycA, our analysis revealed variation in imputed AAT levels was the most predictive of morbidity and mortality for the widest range of diseases over the eight year follow-up period, including heart failure (meta-analysis hazard ratio = 1.60 per standard deviation increase of AAT, P-value = 1×10−10), influenza and pneumonia (HR = 1.37, P = 6×10−10), and liver diseases (HR = 1.81, P = 1×10−6). Transcriptional analyses revealed association of elevated AAT with diverse inflammatory immune pathways. Conclusions This study clarifies the molecular underpinnings of the GlycA biomarker’s associated disease risk, and indicates a previously unrecognised association between elevated AAT and severe disease onset and mortality.
Collapse
|
30
|
Gallois A, Mefford J, Ko A, Vaysse A, Julienne H, Ala-Korpela M, Laakso M, Zaitlen N, Pajukanta P, Aschard H. A comprehensive study of metabolite genetics reveals strong pleiotropy and heterogeneity across time and context. Nat Commun 2019; 10:4788. [PMID: 31636271 PMCID: PMC6803661 DOI: 10.1038/s41467-019-12703-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 09/11/2019] [Indexed: 12/20/2022] Open
Abstract
Genetic studies of metabolites have identified thousands of variants, many of which are associated with downstream metabolic and obesogenic disorders. However, these studies have relied on univariate analyses, reducing power and limiting context-specific understanding. Here we aim to provide an integrated perspective of the genetic basis of metabolites by leveraging the Finnish Metabolic Syndrome In Men (METSIM) cohort, a unique genetic resource which contains metabolic measurements, mostly lipids, across distinct time points as well as information on statin usage. We increase effective sample size by an average of two-fold by applying the Covariates for Multi-phenotype Studies (CMS) approach, identifying 588 significant SNP-metabolite associations, including 228 new associations. Our analysis pinpoints a small number of master metabolic regulator genes, balancing the relative proportion of dozens of metabolite levels. We further identify associations to changes in metabolic levels across time as well as genetic interactions with statin at both the master metabolic regulator and genome-wide level.
Collapse
Affiliation(s)
- Apolline Gallois
- Department of Computational Biology - USR 3756 CNRS, Institut Pasteur, Paris, France
| | - Joel Mefford
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Arthur Ko
- Department of Human Genetics, University of California, Los Angeles, CA, USA
| | - Amaury Vaysse
- Department of Computational Biology - USR 3756 CNRS, Institut Pasteur, Paris, France
| | - Hanna Julienne
- Department of Computational Biology - USR 3756 CNRS, Institut Pasteur, Paris, France
| | - Mika Ala-Korpela
- Systems Epidemiology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, VIC, Australia
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Noah Zaitlen
- Department of Medicine, University of California, San Francisco, CA, USA.
| | - Päivi Pajukanta
- Department of Human Genetics, University of California, Los Angeles, CA, USA.
| | - Hugues Aschard
- Department of Computational Biology - USR 3756 CNRS, Institut Pasteur, Paris, France.
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
31
|
Vandiedonck C. Genetic association of molecular traits: A help to identify causative variants in complex diseases. Clin Genet 2019; 93:520-532. [PMID: 29194587 DOI: 10.1111/cge.13187] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/24/2017] [Accepted: 11/27/2017] [Indexed: 12/14/2022]
Abstract
In the past 15 years, major progresses have been made in the understanding of the genetic basis of regulation of gene expression. These new insights have revolutionized our approach to resolve the genetic variation underlying complex diseases. Gene transcript levels were the first expression phenotypes that were studied. They are heritable and therefore amenable to genome-wide association studies. The genetic variants that modulate them are called expression quantitative trait loci. Their study has been extended to other molecular quantitative trait loci (molQTLs) that regulate gene expression at the various levels, from chromatin state to cellular responses. Altogether, these studies have generated a wealth of basic information on the genome-wide patterns of gene expression and their inter-individual variation. Most importantly, molQTLs have become an invaluable asset in the genetic study of complex diseases. Although the identification of the disease-causing variants on the basis of their overlap with molQTLs requires caution, molQTLs can help to prioritize the relevant candidate gene(s) in the disease-associated regions and bring a functional interpretation of the associated variants, therefore, bridging the gap between genotypes and clinical phenotypes.
Collapse
Affiliation(s)
- C Vandiedonck
- Univ Paris Diderot, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
32
|
Beuchel C, Becker S, Dittrich J, Kirsten H, Toenjes A, Stumvoll M, Loeffler M, Thiele H, Beutner F, Thiery J, Ceglarek U, Scholz M. Clinical and lifestyle related factors influencing whole blood metabolite levels - A comparative analysis of three large cohorts. Mol Metab 2019; 29:76-85. [PMID: 31668394 PMCID: PMC6734104 DOI: 10.1016/j.molmet.2019.08.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/13/2019] [Accepted: 08/13/2019] [Indexed: 12/31/2022] Open
Abstract
Objective Human blood metabolites are influenced by a number of lifestyle and environmental factors. Identification of these factors and the proper quantification of their relevance provides insights into human biological and metabolic disease processes, is key for standardized translation of metabolite biomarkers into clinical applications, and is a prerequisite for comparability of data between studies. However, so far only limited data exist from large and well-phenotyped human cohorts and current methods for analysis do not fully account for the characteristics of these data. The primary aim of this study was to identify, quantify and compare the impact of a comprehensive set of clinical and lifestyle related factors on metabolite levels in three large human cohorts. To achieve this goal, we improve current methodology by developing a principled analysis approach, which could be translated to other cohorts and metabolite panels. Methods 63 Metabolites (amino acids, acylcarnitines) were quantified by liquid chromatography tandem mass spectrometry in three cohorts (total N = 16,222). Supported by a simulation study evaluating various analytical approaches, we developed an analysis pipeline including preprocessing, identification, and quantification of factors affecting metabolite levels. We comprehensively identified uni- and multivariable metabolite associations considering 29 environmental and clinical factors and performed metabolic pathway enrichment and network analyses. Results Inverse normal transformation of batch corrected and outlier removed metabolite levels accompanied by linear regression analysis proved to be the best suited method to deal with the metabolite data. Association analyses revealed numerous uni- and multivariable significant associations. 15 of the analyzed 29 factors explained >1% of variance for at least one of the metabolites. Strongest factors are application of steroid hormones, reticulocytes, waist-to-hip ratio, sex, haematocrit, and age. Effect sizes of factors are comparable across studies. Conclusions We introduced a principled approach for the analysis of MS data allowing identification, and quantification of effects of clinical and lifestyle factors with metabolite levels. We detected a number of known and novel associations broadening our understanding of the regulation of the human metabolome. The large heterogeneity observed between cohorts could almost completely be explained by differences in the distribution of influencing factors emphasizing the necessity of a proper confounder analysis when interpreting metabolite associations. Amino-acids and acylcarnitines analyzed in three studies with >16,000 individuals. Develop a generic and adaptable bioinformatics workflow. Analysis of the impact of 29 clinical and life-style factors on blood metabolites. Analysis of network between factors and metabolites. Comparison of results between studies.
Collapse
Affiliation(s)
- Carl Beuchel
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Susen Becker
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Leipzig, Germany; Department of Pediatric Surgery, University of Leipzig, Leipzig, Germany
| | - Julia Dittrich
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Leipzig, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany; LIFE - Leipzig Research Center for Civilization Diseases, Leipzig University, Leipzig, Germany
| | - Anke Toenjes
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University Hospital Leipzig, Leipzig, Germany
| | - Michael Stumvoll
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University Hospital Leipzig, Leipzig, Germany
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany; LIFE - Leipzig Research Center for Civilization Diseases, Leipzig University, Leipzig, Germany
| | | | | | - Joachim Thiery
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Leipzig, Germany; LIFE - Leipzig Research Center for Civilization Diseases, Leipzig University, Leipzig, Germany
| | - Uta Ceglarek
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Leipzig University, Leipzig, Germany; LIFE - Leipzig Research Center for Civilization Diseases, Leipzig University, Leipzig, Germany
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany; LIFE - Leipzig Research Center for Civilization Diseases, Leipzig University, Leipzig, Germany; IFB Adiposity Diseases, University Hospital Leipzig, Leipzig, Germany.
| |
Collapse
|
33
|
Ellul S, Wake M, Clifford SA, Lange K, Würtz P, Juonala M, Dwyer T, Carlin JB, Burgner DP, Saffery R. Metabolomics: population epidemiology and concordance in Australian children aged 11-12 years and their parents. BMJ Open 2019; 9:106-117. [PMID: 31273021 PMCID: PMC6624050 DOI: 10.1136/bmjopen-2017-020900] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES Nuclear magnetic resonance (NMR) metabolomics is high throughput and cost-effective, with the potential to improve the understanding of disease and risk. We examine the circulating metabolic profile by quantitative NMR metabolomics of a sample of Australian 11-12 year olds children and their parents, describe differences by age and sex, and explore the correlation of metabolites in parent-child dyads. DESIGN The population-based cross-sectional Child Health CheckPoint study nested within the Longitudinal Study of Australian Children. SETTING Blood samples collected from CheckPoint participants at assessment centres in seven Australian cities and eight regional towns; February 2015-March 2016. PARTICIPANTS 1180 children and 1325 parents provided a blood sample and had metabolomics data available. This included 1133 parent-child dyads (518 mother-daughter, 469 mother-son, 68 father-daughter and 78 father-son). OUTCOME MEASURES 228 metabolic measures were obtained for each participant. We focused on 74 biomarkers including amino acid species, lipoprotein subclass measures, lipids, fatty acids, measures related to fatty acid saturation, and composite markers of inflammation and energy homeostasis. RESULTS We identified differences in the concentration of specific metabolites between childhood and adulthood and in metabolic profiles in children and adults by sex. In general, metabolite concentrations were higher in adults than children and sex differences were larger in adults than in children. Positive correlations were observed for the majority of metabolites including isoleucine (CC 0.33, 95% CI 0.27 to 0.38), total cholesterol (CC 0.30, 95% CI 0.24 to 0.35) and omega 6 fatty acids (CC 0.28, 95% CI 0.23 to 0.34) in parent-child comparisons. CONCLUSIONS We describe the serum metabolite profiles from mid-childhood and adulthood in a population-based sample, together with a parent-child concordance. Differences in profiles by age and sex were observed. These data will be informative for investigation of the childhood origins of adult non-communicable diseases and for comparative studies in other populations.
Collapse
Affiliation(s)
- Susan Ellul
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
| | - Melissa Wake
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics and The Liggins Institute, University of Auckland, Auckland, New Zealand
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Susan A Clifford
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Katherine Lange
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Peter Würtz
- Research Programs Unit, Diabetes and Obesity, University of Helsinki, Helsinki, Finland
- Nightingale Health Ltd., Helsinki, Finland
| | - Markus Juonala
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
- Department of Medicine, University of Turku, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Terence Dwyer
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- The George Institute for Global Health, Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford, United Kingdom
- Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - John B Carlin
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - David P Burgner
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Department of Paediatrics, Monash University, Clayton, Victoria, Australia
| | - Richard Saffery
- Murdoch Children’s Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
34
|
Everson TM, Marsit CJ. Integrating -Omics Approaches into Human Population-Based Studies of Prenatal and Early-Life Exposures. Curr Environ Health Rep 2019; 5:328-337. [PMID: 30054820 DOI: 10.1007/s40572-018-0204-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW We present the study design and methodological suggestions for population-based studies that integrate molecular -omics data and highlight recent studies that have used such data to examine the potential impacts of prenatal environmental exposures on fetal health. RECENT FINDINGS Epidemiologic studies have observed numerous relationships between prenatal exposures (smoking, toxic metals, endocrine disruptors) and fetal and early-life molecular profiles, though such investigations have so far been dominated by epigenomic association studies. However, recent transcriptomic, proteomic, and metabolomic studies have demonstrated their promise for the identification of exposure and response biomarkers. Molecular -omics have opened new avenues of research in environmental health that can improve our understanding of disease etiology and contribute to the development of exposure and response biomarkers. Studies that incorporate multiple -omics data from different molecular domains in longitudinally collected samples hold particular promise.
Collapse
Affiliation(s)
- Todd M Everson
- Departments of Environmental Health, Rollins School of Public Health, Emory University, 1518 Clifton Road, Claudia Nance Rollins Room 2021, Atlanta, GA, 30322, USA
| | - Carmen J Marsit
- Departments of Environmental Health, Rollins School of Public Health, Emory University, 1518 Clifton Road, Claudia Nance Rollins Room 2021, Atlanta, GA, 30322, USA. .,Departments of Environmental Health and Epidemiology, Rollins School of Public Health, Emory University, 1518 Clifton Road, Claudia Nance Rollins Room 2021, Atlanta, GA, 30322, USA.
| |
Collapse
|
35
|
Lea A, Subramaniam M, Ko A, Lehtimäki T, Raitoharju E, Kähönen M, Seppälä I, Mononen N, Raitakari OT, Ala-Korpela M, Pajukanta P, Zaitlen N, Ayroles JF. Genetic and environmental perturbations lead to regulatory decoherence. eLife 2019; 8:e40538. [PMID: 30834892 PMCID: PMC6400502 DOI: 10.7554/elife.40538] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 02/14/2019] [Indexed: 01/24/2023] Open
Abstract
Correlation among traits is a fundamental feature of biological systems that remains difficult to study. To address this problem, we developed a flexible approach that allows us to identify factors associated with inter-individual variation in correlation. We use data from three human cohorts to study the effects of genetic and environmental variation on correlations among mRNA transcripts and among NMR metabolites. We first show that environmental exposures (infection and disease) lead to a systematic loss of correlation, which we define as 'decoherence'. Using longitudinal data, we show that decoherent metabolites are better predictors of whether someone will develop metabolic syndrome than metabolites commonly used as biomarkers of this disease. Finally, we demonstrate that correlation itself is under genetic control by mapping hundreds of 'correlation quantitative trait loci (QTLs)'. Together, this work furthers our understanding of how and why coordinated biological processes break down, and points to a potential role for decoherence in disease. Editorial note This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).
Collapse
Affiliation(s)
- Amanda Lea
- Department of Ecology and EvolutionPrinceton UniversityPrincetonUnited States
- Lewis-Sigler Institute for Integrative GenomicsPrinceton UniversityPrincetonUnited States
| | - Meena Subramaniam
- Department of Medicine, Lung Biology CenterUniversity of California, San FranciscoSan FranciscoUnited States
| | - Arthur Ko
- Department of Medicine, David Geffen School of Medicine at UCLAUniversity of California, Los AngelesLos AngelesUnited States
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
- Finnish Cardiovascular Research Center, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Emma Raitoharju
- Finnish Cardiovascular Research Center, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Mika Kähönen
- Finnish Cardiovascular Research Center, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
- Department of Clinical PhysiologyTampere University, Tampere University HospitalTampereFinland
| | - Ilkka Seppälä
- Finnish Cardiovascular Research Center, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Nina Mononen
- Finnish Cardiovascular Research Center, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Olli T Raitakari
- Research Centre of Applied and Preventive Cardiovascular MedicineUniversity of TurkuTurkuFinland
- Department of Clinical Physiology and Nuclear MedicineTurku University HospitalTurkuFinland
| | - Mika Ala-Korpela
- Systems Epidemiology, Baker Heart and Diabetes InstituteMelbourneAustralia
- Computational Medicine, Faculty of Medicine, Biocenter OuluUniversity of OuluOuluFinland
- NMR Metabolomics Laboratory, School of PharmacyUniversity of Eastern FinlandKuopioFinland
- Population Health Science, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
- Medical Research Council Integrative Epidemiology UnitUniversity of BristolBristolUnited Kingdom
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health SciencesThe Alfred Hospital, Monash UniversityMelbourneAustralia
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLAUniversity of California, Los AngelesLos AngelesUnited States
| | - Noah Zaitlen
- Department of Medicine, Lung Biology CenterUniversity of California, San FranciscoSan FranciscoUnited States
| | - Julien F Ayroles
- Department of Ecology and EvolutionPrinceton UniversityPrincetonUnited States
- Lewis-Sigler Institute for Integrative GenomicsPrinceton UniversityPrincetonUnited States
| |
Collapse
|
36
|
Hughes MF, Lenighan YM, Godson C, Roche HM. Exploring Coronary Artery Disease GWAs Targets With Functional Links to Immunometabolism. Front Cardiovasc Med 2018; 5:148. [PMID: 30460244 PMCID: PMC6232936 DOI: 10.3389/fcvm.2018.00148] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 10/01/2018] [Indexed: 12/24/2022] Open
Abstract
Finding genetic variants that cause functional disruption or regulatory change among the many implicated GWAs variants remains a key challenge to translating the findings from GWAs to therapeutic treatments. Defining the causal mechanisms behind the variants require functional screening experiments that can be complex and costly. Prioritizing variants for functional characterization using techniques that capture important functional and regulatory elements can assist this. The genetic architecture of complex traits such as cardiovascular disease and type II diabetes comprise an enormously large number of variants of small effect contributing to heritability and spread throughout the genome. This makes it difficult to distinguish which variants or core genes are most relevant for prioritization and how they contribute to the regulatory networks that become dysregulated leading to disease. Despite these challenges, recent GWAs for CAD prioritized genes associated with lipid metabolism, coagulation and adhesion along with novel signals related to innate immunity, adipose tissue and, vascular function as important core drivers of risk. We focus on three examples of novel signals associated with CAD which affect risk through missense or UTR mutations indicating their potential for therapeutic modification. These variants play roles in adipose tissue function vascular function and innate immunity which form the cornerstones of immuno-metabolism. In addition we have explored the putative, but potentially important interactions between the environment, specifically food and nutrition, with respect to key processes.
Collapse
Affiliation(s)
- Maria F Hughes
- UCD Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.,Nutrigenomics Research Group, UCD Institute of Food and Health, School of Public Health Physiotherapy and Sports Science, University College Dublin, Dublin, Ireland.,Centre of Excellence for Public Health, Queen's University Belfast, Belfast, United Kingdom.,UCD Institute of Food and Health, School of Public Health Physiotherapy and Sports Science, University College Dublin, Dublin, Ireland
| | - Yvonne M Lenighan
- UCD Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.,UCD Institute of Food and Health, School of Public Health Physiotherapy and Sports Science, University College Dublin, Dublin, Ireland
| | - Catherine Godson
- UCD Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.,School of Medicine, University College Dublin, Dublin, Ireland
| | - Helen M Roche
- UCD Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland.,Nutrigenomics Research Group, UCD Institute of Food and Health, School of Public Health Physiotherapy and Sports Science, University College Dublin, Dublin, Ireland.,UCD Institute of Food and Health, School of Public Health Physiotherapy and Sports Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
37
|
Petrie JR, Guzik TJ, Touyz RM. Diabetes, Hypertension, and Cardiovascular Disease: Clinical Insights and Vascular Mechanisms. Can J Cardiol 2018; 34:575-584. [PMID: 29459239 PMCID: PMC5953551 DOI: 10.1016/j.cjca.2017.12.005] [Citation(s) in RCA: 993] [Impact Index Per Article: 141.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 12/06/2017] [Accepted: 12/07/2017] [Indexed: 12/11/2022] Open
Abstract
Hypertension and type 2 diabetes are common comorbidities. Hypertension is twice as frequent in patients with diabetes compared with those who do not have diabetes. Moreover, patients with hypertension often exhibit insulin resistance and are at greater risk of diabetes developing than are normotensive individuals. The major cause of morbidity and mortality in diabetes is cardiovascular disease, which is exacerbated by hypertension. Accordingly, diabetes and hypertension are closely interlinked because of similar risk factors, such as endothelial dysfunction, vascular inflammation, arterial remodelling, atherosclerosis, dyslipidemia, and obesity. There is also substantial overlap in the cardiovascular complications of diabetes and hypertension related primarily to microvascular and macrovascular disease. Common mechanisms, such as upregulation of the renin-angiotensin-aldosterone system, oxidative stress, inflammation, and activation of the immune system likely contribute to the close relationship between diabetes and hypertension. In this article we discuss diabetes and hypertension as comorbidities and discuss the pathophysiological features of vascular complications associated with these conditions. We also highlight some vascular mechanisms that predispose to both conditions, focusing on advanced glycation end products, oxidative stress, inflammation, the immune system, and microRNAs. Finally, we provide some insights into current therapies targeting diabetes and cardiovascular complications and introduce some new agents that may have vasoprotective therapeutic potential in diabetes.
Collapse
Affiliation(s)
- John R Petrie
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom.
| |
Collapse
|