1
|
Tutino M, Yu NYL, Hatzikotoulas K, Park YC, Kreitmaier P, Katsoula G, Berner R, Casteels K, Elding Larsson H, Kordonouri O, Ołtarzewski M, Szypowska A, Ott R, Weiss A, Winkler C, Zapardiel-Gonzalo J, Petrera A, Hauck SM, Bonifacio E, Ziegler AG, Zeggini E. Genetics of circulating proteins in newborn babies at high risk of type 1 diabetes. Nat Commun 2025; 16:3750. [PMID: 40263317 PMCID: PMC12015297 DOI: 10.1038/s41467-025-58972-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 04/04/2025] [Indexed: 04/24/2025] Open
Abstract
Type 1 diabetes is a chronic, autoimmune disease characterized by the destruction of insulin-producing β-cells in the pancreas. Early detection can facilitate timely intervention, potentially delaying or preventing disease onset. Circulating proteins reflect dysregulated biological processes and offer insights into early disease mechanisms. Here, we construct a genome-wide pQTL map of 1985 proteins in 695 newborn babies (median age 2 days) at increased genetic risk of developing Type 1 diabetes. We identify 535 pQTLs (352 cis-pQTLs, 183 trans-pQTLs), 62 of which characteristic of newborns. We show colocalization of pQTLs for CTRB1, APOBR, IL7R, CPA1, and PNLIPRP1 with Type 1 diabetes GWAS signals, and Mendelian randomization causally implicates each of these five proteins in the aetiology of Type 1 diabetes. Our study illustrates the utility of newborn molecular profiles for discovering potential drug targets for childhood diseases of significant concern.
Collapse
Affiliation(s)
- Mauro Tutino
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Nancy Yiu-Lin Yu
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Konstantinos Hatzikotoulas
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Young-Chan Park
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Peter Kreitmaier
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Technical University of Munich, TUM School of Medicine and Health, Graduate School of Experimental Medicine, Munich, Germany
- Technical University of Munich and Klinikum Rechts der Isar, TUM School of Medicine and Health, 81675, Munich, Germany
| | - Georgia Katsoula
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Technical University of Munich, TUM School of Medicine and Health, Graduate School of Experimental Medicine, Munich, Germany
- Technical University of Munich and Klinikum Rechts der Isar, TUM School of Medicine and Health, 81675, Munich, Germany
| | - Reinhard Berner
- Department of Pediatrics, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Kristina Casteels
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Helena Elding Larsson
- Unit for Pediatric Endocrinology, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
- Department of Paediatrics, Skane University Hospital, Malmö/Lund, Lund, Sweden
| | - Olga Kordonouri
- Kinder- und Jugendkrankenhaus AUF DER BULT, Hannover, Germany
| | - Mariusz Ołtarzewski
- Department of Screening and Metabolic Diagnostics, Institute of Mother and Child, Warsaw, Poland
| | - Agnieszka Szypowska
- Department of Paediatric Diabetology and Paediatrics, Medical University of Warsaw, Warsaw, Poland
| | - Raffael Ott
- Institute of Diabetes Research, Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| | - Andreas Weiss
- Institute of Diabetes Research, Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| | - Christiane Winkler
- Institute of Diabetes Research, Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
- Forschergruppe Diabetes e.V. at Helmholtz Munich, Munich, Germany
| | - Jose Zapardiel-Gonzalo
- Institute of Diabetes Research, Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
| | - Agnese Petrera
- Metabolomics and Proteomics Core, Helmholtz Zentrum München - German Research Center for Environmental Health, Munich, Germany
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum München - German Research Center for Environmental Health, Munich, Germany
| | - Ezio Bonifacio
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Munich at University Hospital Carl Gustav Carus and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany
- Forschergruppe Diabetes e.V. at Helmholtz Munich, Munich, Germany
- Forschergruppe Diabetes, School of Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.
- Technical University of Munich and Klinikum Rechts der Isar, TUM School of Medicine and Health, 81675, Munich, Germany.
| |
Collapse
|
2
|
Cui Z, Qiu J, Lin J, Fu Y, Lin L. Discovering genetically-supported drug targets for multisite chronic pain through multi-omics Mendelian randomization and single-cell RNA-sequencing. Neuroscience 2025; 572:254-268. [PMID: 39993665 DOI: 10.1016/j.neuroscience.2025.02.038] [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: 10/05/2024] [Revised: 01/14/2025] [Accepted: 02/17/2025] [Indexed: 02/26/2025]
Abstract
Multisite chronic pain (MCP) is a highly prevalent disorder with substantial unmet therapeutic needs.We conducted multi-omics Mendelian randomization and Bayesian colocalization to identify potential therapeutic targets for MCP. Summary-level data of gene expressions and protein abundance levels were obtained from corresponding quantitative trait loci studies, respectively. Summary-level data for MCP was leveraged from the UK Biobank. The transcriptome-wide association study (TWAS), Mendelian randomization, and Bayesian colocalization approaches were applied to investigate the potential causal effects of gene expressions and protein levels on MCP in both blood and brain tissues. Phenome-wide Mendelian randomization analysis (MR-PheWAS), single-cell sequencing data, protein-protein interaction (PPI), and reaction pathway analysis were further conducted to digging the underlying mechanisms. Our analysis identified and validated two plasma targets for MCP, namely KLC1 and LANCL1, at both gene expression levels and protein levels across multi-methodologies. Moreover, MR-PheWAS observed additional benefits associated with these two targets. Through analyses based on single-cell sequencing data, we identified critical cell types for KLC1, primarily megakaryocytes, and neurons, notably linked to the axon guidance pathway, while LANCL1 showed associations with B lymphocytes, neurons, and the electron transport pathway. In dorsal root ganglions, we identified enrichments of both LANCL1 and KLC1 in putative silent nociceptors. The effects are possibly mediated through axonal transport and the activation of NMDARs, supported by PPI and reaction pathway analysis. Our multi-dimensional analysis suggests that genetically determined KLC1 and LANCL1 are causally linked to MCP risk, holding promise as appealing drug targets for MCP.
Collapse
Affiliation(s)
- Ziyang Cui
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Dermatology and Venereology, Peking University First Hospital, Beijing, China.
| | - Junxiong Qiu
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Jianwei Lin
- Big Data Laboratory, Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, Guangdong, China.
| | - Yanni Fu
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Liling Lin
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
3
|
Tong H, Zhao Y, Cui Y, Yao J, Zhang T. Multi-omic studies on the pathogenesis of Sepsis. J Transl Med 2025; 23:361. [PMID: 40128726 PMCID: PMC11934817 DOI: 10.1186/s12967-025-06366-w] [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: 11/20/2024] [Accepted: 03/08/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Sepsis is a life-threatening inflammatory condition, and its underlying genetic mechanisms are not yet fully elucidated. We applied methods such as Mendelian randomization (MR), genetic correlation analysis, and colocalization analysis to integrate multi-omics data and explore the relationship between genetically associated genes and sepsis, as well as sepsis-related mortality, with the goal of identifying key genetic factors and their potential mechanistic pathways. METHODS To identify therapeutic targets for sepsis and sepsis-related mortality, we conducted an MR analysis on 11,643 sepsis cases and 1,896 cases of 28-day sepsis mortality from the UK Biobank cohort. The exposure data consisted of 15,944 potential druggable genes (expression quantitative trait loci, eQTL) and 4,907 plasma proteins (protein quantitative trait loci, pQTL). We then performed sensitivity analysis, SMR analysis, reverse MR analysis, genetic correlation analysis, colocalization analysis, enrichment analysis, and protein-protein interaction network analysis on the overlapping genes. Validation was conducted using 17,133 sepsis cases from FinnGen R12. Drug prediction and molecular docking were subsequently used to further assess the therapeutic potential of the identified drug targets, while PheWAS was used to evaluate potential side effects. Finally, mediation analysis was conducted to identify the mediating role of related metabolites. RESULTS The MR analysis results identified a significant causal relationship between 24 genes and sepsis. The robustness of these causal associations was further strengthened by SMR analysis, sensitivity analysis, and reverse MR analysis. Genetic correlation analysis revealed that only two of these genes were genetically correlated with sepsis. Colocalization analysis showed that only one gene was closely associated with sepsis, while validation using the FinnGen dataset identified three genes. In the MR analysis of 28-day sepsis mortality, seven genes were found to have significant associations, with reverse MR analysis excluding one gene. The remaining genes passed sensitivity analysis, with no significant genes identified in genetic correlation and colocalization analyses. Molecular docking demonstrated excellent binding affinity between drugs and proteins with available structural data. PheWAS at the gene level did not reveal any potential side effects of the related drugs. CONCLUSIONS The identified drug targets, associated pathways, and metabolites have enhanced our understanding of the complex relationships between genes and sepsis. These genes and metabolites can serve as effective targets for sepsis treatment, paving new pathways in this field and laying a foundation for future research.
Collapse
Affiliation(s)
- Hongjie Tong
- Department of Critical Care Medicine, Jinhua Hospital Affiliated to Zhejiang University, Jinhua, Zhejiang, China
- Zhejiang University School of Medicine, Hangzhou, China
| | - Yuhang Zhao
- Department of Neurology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Ying Cui
- Department of Critical Care Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Jiali Yao
- Department of Critical Care Medicine, Jinhua Hospital Affiliated to Zhejiang University, Jinhua, Zhejiang, China.
| | - Tianlong Zhang
- Department of Critical Care Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
| |
Collapse
|
4
|
Wang B, Pozarickij A, Mazidi M, Wright N, Yao P, Said S, Iona A, Kartsonaki C, Fry H, Lin K, Chen Y, Du H, Avery D, Schmidt-Valle D, Yu C, Sun D, Lv J, Hill M, Li L, Bennett DA, Collins R, Walters RG, Clarke R, Millwood IY, Chen Z. Comparative studies of 2168 plasma proteins measured by two affinity-based platforms in 4000 Chinese adults. Nat Commun 2025; 16:1869. [PMID: 39984443 PMCID: PMC11845630 DOI: 10.1038/s41467-025-56935-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 01/27/2025] [Indexed: 02/23/2025] Open
Abstract
Proteomics offers unique insights into human biology and drug development, but few studies have directly compared the utility of different proteomics platforms. We measured plasma levels of 2168 proteins in 3976 Chinese adults using both Olink Explore and SomaScan platforms. The correlation of protein levels between platforms was modest (median rho = 0.29), with protein abundance and data quality parameters being key factors influencing correlation. For 1694 proteins with one-to-one matched reagents, 765 Olink and 513 SomaScan proteins had cis-pQTLs, including 400 with colocalising cis-pQTLs. Moreover, 1096 Olink and 1429 SomaScan proteins were associated with BMI, while 279 and 154 proteins were associated with risk of ischaemic heart disease, respectively. Addition of Olink and SomaScan proteins to conventional risk factors for ischaemic heart disease improved C-statistics from 0.845 to 0.862 (NRI: 12.2%) and 0.863 (NRI: 16.4%), respectively. These results demonstrate the utility of these platforms and could inform the design and interpretation of future studies.
Collapse
Grants
- 82192901, 82192904, 82192900 National Natural Science Foundation of China (National Science Foundation of China)
- CH/1996001/9454 British Heart Foundation (BHF)
- MC-PC-13049, MC-PC-14135 RCUK | Medical Research Council (MRC)
- FS/18/23/33512 British Heart Foundation (BHF)
- C16077/A29186, C500/A16896 Cancer Research UK (CRUK)
- Wellcome Trust
- 212946/Z/18/Z, 202922/Z/16/Z, 104085/Z/14/Z, 088158/Z/09/Z Wellcome Trust (Wellcome)
- The CKB baseline survey and the first re-survey were supported by the Kadoorie Charitable Foundation in Hong Kong. The long-term follow-up and subsequent resurveys have been supported by Wellcome grants to Oxford University (212946/Z/18/Z, 202922/Z/16/Z, 104085/Z/14/Z, 088158/Z/09/Z) and grants from the National Natural Science Foundation of China (82192901, 82192904, 82192900) and from the National Key Research and Development Program of China (2016YFC0900500).The UK Medical Research Council (MC_UU_00017/1, MC_UU_12026/2, MC_U137686851), Cancer Research UK (C16077/A29186, C500/A16896) and British Heart Foundation (CH/1996001/9454), provide core funding to the Clinical Trial Service Unit and Epidemiological Studies Unit, Oxford University for the project. The proteomic assays were supported by BHF (FS/18/23/33512), Novo Nordisk, Olink, SomaScan and NDPH. DNA extraction and genotyping were supported by GlaxoSmithKline and the UK Medical Research Council (MC-PC-13049, MC-PC-14135). Computation used the Oxford Biomedical Research Computing (BMRC) facility, a joint development between the Wellcome Centre for Human Genetics and the Big Data Institute supported by Health Data Research UK and the NIHR Oxford Biomedical Research Centre. The views expressed are those of the author(s) and not necessarily those of the NHS, the NIHR or the Department of Health.
Collapse
Affiliation(s)
- Baihan Wang
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Alfred Pozarickij
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Mohsen Mazidi
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Neil Wright
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Pang Yao
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Saredo Said
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Andri Iona
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Christiana Kartsonaki
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Hannah Fry
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Kuang Lin
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Yiping Chen
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Huaidong Du
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Daniel Avery
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Dan Schmidt-Valle
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Canqing Yu
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Dianjianyi Sun
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Jun Lv
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Michael Hill
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Liming Li
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Derrick A Bennett
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Rory Collins
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Robin G Walters
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Robert Clarke
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Iona Y Millwood
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| | - Zhengming Chen
- Clinical Trial Service Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK.
| |
Collapse
|
5
|
Valdés-Mas R, Leshem A, Zheng D, Cohen Y, Kern L, Zmora N, He Y, Katina C, Eliyahu-Miller S, Yosef-Hevroni T, Richman L, Raykhel B, Allswang S, Better R, Shmueli M, Saftien A, Cullin N, Slamovitz F, Ciocan D, Ouyang KS, Mor U, Dori-Bachash M, Molina S, Levin Y, Atarashi K, Jona G, Puschhof J, Harmelin A, Stettner N, Chen M, Suez J, Honda K, Lieb W, Bang C, Kori M, Maharshak N, Merbl Y, Shibolet O, Halpern Z, Shouval DS, Shamir R, Franke A, Abdeen SK, Shapiro H, Savidor A, Elinav E. Metagenome-informed metaproteomics of the human gut microbiome, host, and dietary exposome uncovers signatures of health and inflammatory bowel disease. Cell 2025; 188:1062-1083.e36. [PMID: 39837331 DOI: 10.1016/j.cell.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/08/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025]
Abstract
Host-microbiome-dietary interactions play crucial roles in regulating human health, yet their direct functional assessment remains challenging. We adopted metagenome-informed metaproteomics (MIM), in mice and humans, to non-invasively explore species-level microbiome-host interactions during commensal and pathogen colonization, nutritional modification, and antibiotic-induced perturbation. Simultaneously, fecal MIM accurately characterized the nutritional exposure landscape in multiple clinical and dietary contexts. Implementation of MIM in murine auto-inflammation and in human inflammatory bowel disease (IBD) characterized a "compositional dysbiosis" and a concomitant species-specific "functional dysbiosis" driven by suppressed commensal responses to inflammatory host signals. Microbiome transfers unraveled early-onset kinetics of these host-commensal cross-responsive patterns, while predictive analyses identified candidate fecal host-microbiome IBD biomarker protein pairs outperforming S100A8/S100A9 (calprotectin). Importantly, a simultaneous fecal nutritional MIM assessment enabled the determination of IBD-related consumption patterns, dietary treatment compliance, and small intestinal digestive aberrations. Collectively, a parallelized dietary-bacterial-host MIM assessment functionally uncovers trans-kingdom interactomes shaping gastrointestinal ecology while offering personalized diagnostic and therapeutic insights into microbiome-associated disease.
Collapse
Affiliation(s)
- Rafael Valdés-Mas
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Avner Leshem
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Department of Surgery, Tel Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Danping Zheng
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yotam Cohen
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Lara Kern
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Niv Zmora
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Yiming He
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Corine Katina
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), Weizmann Institute of Science, Rehovot, Israel
| | | | - Tal Yosef-Hevroni
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Liron Richman
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Barbara Raykhel
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Shira Allswang
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Reut Better
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Merav Shmueli
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Nyssa Cullin
- Division of Microbiome & Cancer, DKFZ, Heidelberg, Germany
| | - Fernando Slamovitz
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Dragos Ciocan
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Uria Mor
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Mally Dori-Bachash
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Shahar Molina
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Yishai Levin
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), Weizmann Institute of Science, Rehovot, Israel
| | - Koji Atarashi
- RIKEN Center for Integrative Medical Sciences (IMS), Tsurumi, Yokohama, Kanagawa, Japan; Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Ghil Jona
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Jens Puschhof
- Division of Microbiome & Cancer, DKFZ, Heidelberg, Germany
| | - Alon Harmelin
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Noa Stettner
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Minhu Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jotham Suez
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kenya Honda
- RIKEN Center for Integrative Medical Sciences (IMS), Tsurumi, Yokohama, Kanagawa, Japan; Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Wolfgang Lieb
- Institute of Epidemiology and Biobank Popgen, University Hospital of Schleswig-Holstein (UKSH), Kiel, Germany
| | - Corinna Bang
- Institute of Clinical Molecular Biology, Christian-Albrechts-Universität Zu Kiel, Kiel, Germany; University Hospital of Schleswig-Holstein (UKSH), Kiel, Germany
| | - Michal Kori
- Pediatric Gastroenterology Unit, Kaplan Medical Center, Rehovot, Israel; Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nitsan Maharshak
- School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Gastroenterology and Hepatology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Yifat Merbl
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Oren Shibolet
- School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Gastroenterology and Hepatology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Zamir Halpern
- School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Gastroenterology and Hepatology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Dror S Shouval
- School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Institute of Gastroenterology, Nutrition, and Liver Diseases, Schneider Children's Medical Centre, Petach-Tikva, Israel
| | - Raanan Shamir
- School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Institute of Gastroenterology, Nutrition, and Liver Diseases, Schneider Children's Medical Centre, Petach-Tikva, Israel
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-Universität Zu Kiel, Kiel, Germany; University Hospital of Schleswig-Holstein (UKSH), Kiel, Germany
| | - Suhaib K Abdeen
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Hagit Shapiro
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Savidor
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Division of Microbiome & Cancer, DKFZ, Heidelberg, Germany.
| |
Collapse
|
6
|
Deng YT, You J, He Y, Zhang Y, Li HY, Wu XR, Cheng JY, Guo Y, Long ZW, Chen YL, Li ZY, Yang L, Zhang YR, Chen SD, Ge YJ, Huang YY, Shi LM, Dong Q, Mao Y, Feng JF, Cheng W, Yu JT. Atlas of the plasma proteome in health and disease in 53,026 adults. Cell 2025; 188:253-271.e7. [PMID: 39579765 DOI: 10.1016/j.cell.2024.10.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 07/17/2024] [Accepted: 10/24/2024] [Indexed: 11/25/2024]
Abstract
Large-scale proteomics studies can refine our understanding of health and disease and enable precision medicine. Here, we provide a detailed atlas of 2,920 plasma proteins linking to diseases (406 prevalent and 660 incident) and 986 health-related traits in 53,026 individuals (median follow-up: 14.8 years) from the UK Biobank, representing the most comprehensive proteome profiles to date. This atlas revealed 168,100 protein-disease associations and 554,488 protein-trait associations. Over 650 proteins were shared among at least 50 diseases, and over 1,000 showed sex and age heterogeneity. Furthermore, proteins demonstrated promising potential in disease discrimination (area under the curve [AUC] > 0.80 in 183 diseases). Finally, integrating protein quantitative trait locus data determined 474 causal proteins, providing 37 drug-repurposing opportunities and 26 promising targets with favorable safety profiles. These results provide an open-access comprehensive proteome-phenome resource (https://proteome-phenome-atlas.com/) to help elucidate the biological mechanisms of diseases and accelerate the development of disease biomarkers, prediction models, and therapeutic targets.
Collapse
Affiliation(s)
- Yue-Ting Deng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jia You
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China; Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Yu He
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hai-Yun Li
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xin-Rui Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ji-Yun Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu Guo
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zi-Wen Long
- Department of Gastric Cancer Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Lin Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ze-Yu Li
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China; Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Liu Yang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Ru Zhang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Jun Ge
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Yuan Huang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Le-Ming Shi
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital Fudan University, Shanghai, China.
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China; Department of Computer Science, University of Warwick, Coventry, UK.
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China; Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Crouch DJM, Inshaw JRJ, Robertson CC, Ng E, Zhang J, Chen W, Onengut‐Gumuscu S, Cutler AJ, Sidore C, Cucca F, Pociot F, Concannon P, Rich SS, Todd JA. Bayesian Effect Size Ranking to Prioritise Genetic Risk Variants in Common Diseases for Follow-Up Studies. Genet Epidemiol 2025; 49:e22608. [PMID: 39749473 PMCID: PMC11696485 DOI: 10.1002/gepi.22608] [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: 03/28/2024] [Revised: 09/10/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025]
Abstract
Biological datasets often consist of thousands or millions of variables, e.g. genetic variants or biomarkers, and when sample sizes are large it is common to find many associated with an outcome of interest, for example, disease risk in a GWAS, at high levels of statistical significance, but with very small effects. The False Discovery Rate (FDR) is used to identify effects of interest based on ranking variables according to their statistical significance. Here, we develop a complementary measure to the FDR, the priorityFDR, that ranks variables by a combination of effect size and significance, allowing further prioritisation among a set of variables that pass a significance or FDR threshold. Applying to the largest GWAS of type 1 diabetes to date (15,573 cases and 158,408 controls), we identified 26 independent genetic associations, including two newly-reported loci, with qualitatively lower priorityFDRs than the remaining 175 signals. We detected putatively causal type 1 diabetes risk genes using Mendelian Randomisation, and found that these were located disproportionately close to low priorityFDR signals (p = 0.005), as were genes in the IL-2 pathway (p = 0.003). Selecting variables on both effect size and significance can lead to improved prioritisation for mechanistic follow-up studies from genetic and other large biological datasets.
Collapse
Grants
- DP3 DK111906 NIDDK NIH HHS
- T32 LM012416 NLM NIH HHS
- U01 DK062418 NIDDK NIH HHS
- Wellcome Trust
- This work was funded by JDRF grants 9-2011-253, 5-SRA-2015-130-A-N and 4-SRA-2017-473-A-N, and Wellcome grants 091157/Z/10/Z and 107212/Z/15/Z, to the Diabetes and Inflammation Laboratory, University of Oxford, Fondazione di Sardegna grant U1301.2015/AI.1157.BE to Francesco Cucca, NIDDK grants U01 DK062418 and DP3 DK111906 and US National Library of Medicine grant T32 LM012416.
- This work was funded by JDRF grants 9‐2011‐253, 5‐SRA‐2015‐130‐A‐N and 4‐SRA‐2017‐473‐A‐N, and Wellcome grants 091157/Z/10/Z and 107212/Z/15/Z, to the Diabetes and Inflammation Laboratory, University of Oxford, Fondazione di Sardegna grant U1301.2015/AI.1157.BE to Francesco Cucca, NIDDK grants U01 DK062418 and DP3 DK111906 and US National Library of Medicine grant T32 LM012416.
Collapse
Affiliation(s)
- Daniel J. M. Crouch
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of MedicineCentre for Human Genetics, NIHR Oxford Biomedical Research CentreUniversity of OxfordOxfordUK
| | - Jamie R. J. Inshaw
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of MedicineCentre for Human Genetics, NIHR Oxford Biomedical Research CentreUniversity of OxfordOxfordUK
| | | | - Esther Ng
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of MedicineCentre for Human Genetics, NIHR Oxford Biomedical Research CentreUniversity of OxfordOxfordUK
- Nuffield Department of OrthopaedicsKennedy Institute of Rheumatology, Rheumatology and Musculoskeletal SciencesUniversity of OxfordOxfordUK
| | - Jia‐Yuan Zhang
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of MedicineCentre for Human Genetics, NIHR Oxford Biomedical Research CentreUniversity of OxfordOxfordUK
| | - Wei‐Min Chen
- Center for Public Health GenomicsUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Public Health SciencesUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Suna Onengut‐Gumuscu
- Center for Public Health GenomicsUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Public Health SciencesUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Antony J. Cutler
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of MedicineCentre for Human Genetics, NIHR Oxford Biomedical Research CentreUniversity of OxfordOxfordUK
| | - Carlo Sidore
- Institute for Research in Genetics and Biomedicine (IRGB)SardiniaItaly
| | - Francesco Cucca
- Institute for Research in Genetics and Biomedicine (IRGB)SardiniaItaly
| | - Flemming Pociot
- Department of PediatricsHerlev University HospitalCopenhagenDenmark
- Institute of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Department of Clinical ResearchSteno Diabetes Center Copenhagen, Type 1 Diabetes BiologyGentofteDenmark
| | - Patrick Concannon
- Department of Pathology, Immunology, and Laboratory MedicineUniversity of FloridaGainesvilleFloridaUSA
- Genetics InstituteUniversity of FloridaGainesvilleFloridaUSA
| | - Stephen S. Rich
- Center for Public Health GenomicsUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Public Health SciencesUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - John A. Todd
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of MedicineCentre for Human Genetics, NIHR Oxford Biomedical Research CentreUniversity of OxfordOxfordUK
| |
Collapse
|
8
|
Geyer PE, Hornburg D, Pernemalm M, Hauck SM, Palaniappan KK, Albrecht V, Dagley LF, Moritz RL, Yu X, Edfors F, Vandenbrouck Y, Mueller-Reif JB, Sun Z, Brun V, Ahadi S, Omenn GS, Deutsch EW, Schwenk JM. The Circulating Proteome─Technological Developments, Current Challenges, and Future Trends. J Proteome Res 2024; 23:5279-5295. [PMID: 39479990 PMCID: PMC11629384 DOI: 10.1021/acs.jproteome.4c00586] [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: 07/09/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 11/02/2024]
Abstract
Recent improvements in proteomics technologies have fundamentally altered our capacities to characterize human biology. There is an ever-growing interest in using these novel methods for studying the circulating proteome, as blood offers an accessible window into human health. However, every methodological innovation and analytical progress calls for reassessing our existing approaches and routines to ensure that the new data will add value to the greater biomedical research community and avoid previous errors. As representatives of HUPO's Human Plasma Proteome Project (HPPP), we present our 2024 survey of the current progress in our community, including the latest build of the Human Plasma Proteome PeptideAtlas that now comprises 4608 proteins detected in 113 data sets. We then discuss the updates of established proteomics methods, emerging technologies, and investigations of proteoforms, protein networks, extracellualr vesicles, circulating antibodies and microsamples. Finally, we provide a prospective view of using the current and emerging proteomics tools in studies of circulating proteins.
Collapse
Affiliation(s)
- Philipp E. Geyer
- Department
of Proteomics and Signal Transduction, Max
Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Daniel Hornburg
- Seer,
Inc., Redwood City, California 94065, United States
- Bruker
Scientific, San Jose, California 95134, United States
| | - Maria Pernemalm
- Department
of Oncology and Pathology/Science for Life Laboratory, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Stefanie M. Hauck
- Metabolomics
and Proteomics Core, Helmholtz Zentrum München
GmbH, German Research Center for Environmental Health, 85764 Oberschleissheim,
Munich, Germany
| | | | - Vincent Albrecht
- Department
of Proteomics and Signal Transduction, Max
Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Laura F. Dagley
- The
Walter and Eliza Hall Institute for Medical Research, Parkville, VIC 3052, Australia
- Department
of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Robert L. Moritz
- Institute
for Systems Biology, Seattle, Washington 98109, United States
| | - Xiaobo Yu
- State
Key Laboratory of Medical Proteomics, Beijing
Proteome Research Center, National Center for Protein Sciences-Beijing
(PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Fredrik Edfors
- Science
for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, 17121 Solna, Sweden
| | | | - Johannes B. Mueller-Reif
- Department
of Proteomics and Signal Transduction, Max
Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Zhi Sun
- Institute
for Systems Biology, Seattle, Washington 98109, United States
| | - Virginie Brun
- Université Grenoble
Alpes, CEA, Leti, Clinatec, Inserm UA13
BGE, CNRS FR2048, Grenoble, France
| | - Sara Ahadi
- Alkahest, Inc., Suite
D San Carlos, California 94070, United States
| | - Gilbert S. Omenn
- Institute
for Systems Biology, Seattle, Washington 98109, United States
- Departments
of Computational Medicine & Bioinformatics, Internal Medicine,
Human Genetics and Environmental Health, University of Michigan, Ann Arbor, Michigan 48109-2218, United States
| | - Eric W. Deutsch
- Institute
for Systems Biology, Seattle, Washington 98109, United States
| | - Jochen M. Schwenk
- Science
for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, 17121 Solna, Sweden
| |
Collapse
|
9
|
Samet M, Mahdiabadi PR, Tajamolian M, Jelodar MG, Monshizadeh K, Javazm RR, Yazdi M, Abessi P, Hoseini SM. ABO gene polymorphism and COVID-19 severity: The impact on haematological complications, inflammatory markers, and lung lesions. Hum Immunol 2024; 85:111184. [PMID: 39566435 DOI: 10.1016/j.humimm.2024.111184] [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: 08/12/2024] [Revised: 11/01/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024]
Abstract
PURPOSE The study aimed to investigate the connection between an intronic variant in the ABO gene (rs657152) and the severity of COVID-19 in terms of clinical symptoms, haematological complications, inflammatory markers, and lung lesions. METHODS After applying exclusion criteria, the study included 240 patients divided into 3 groups: 88 Outpatients, 84 Ward-hospitalized, and 68 ICU-admitted/failed patients. The tetra-ARMS PCR method was used to genotype ABO polymorphism in the patient. Paraclinical tests of patients at the time of admission (before receiving conventional treatments) included levels of C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR), as well as a complete blood count (CBC). Also, the severity of lung lesions was evaluated based on the results of spiral computed tomography (CT) of the chest during admission. RESULTS The statistical analysis using the ANOVA test revealed significant differences in the mean values of allele frequencies (p-value = 0.0020) and genotype proportions (p-value = 0.0017) among clinical groups. The study also found a notable difference in ABO polymorphism across different levels of the inflammatory marker CRP, but not with the ESR levels. Furthermore, the study showed a significant difference in the distribution of lung lesion severity and ABO polymorphism among different clinical groups. CONCLUSION To conclude, our findings supported the substantial impact of ABO polymorphism rs657152 on the severity of COVID-19 in Iranian patients, specifically concerning haematological complications, inflammatory markers, and lung lesions. The study underscored the protective effect of the AC genotype and the detrimental impact of the CC genotype on clinical manifestations.
Collapse
Affiliation(s)
- Mohammad Samet
- Departments of Internal Medicine, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Parvane Raeesi Mahdiabadi
- Departments of Internal Medicine, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Masoud Tajamolian
- Medical Genetics Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohsen Gholinataj Jelodar
- Departments of Internal Medicine, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Kimia Monshizadeh
- Medical Genetics Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Reza Rafiei Javazm
- Biotechnology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mehran Yazdi
- Departments of Internal Medicine, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Panteha Abessi
- Biotechnology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyed Mehdi Hoseini
- Hematology and Oncology Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
10
|
Wang QS, Hasegawa T, Namkoong H, Saiki R, Edahiro R, Sonehara K, Tanaka H, Azekawa S, Chubachi S, Takahashi Y, Sakaue S, Namba S, Yamamoto K, Shiraishi Y, Chiba K, Tanaka H, Makishima H, Nannya Y, Zhang Z, Tsujikawa R, Koike R, Takano T, Ishii M, Kimura A, Inoue F, Kanai T, Fukunaga K, Ogawa S, Imoto S, Miyano S, Okada Y. Statistically and functionally fine-mapped blood eQTLs and pQTLs from 1,405 humans reveal distinct regulation patterns and disease relevance. Nat Genet 2024; 56:2054-2067. [PMID: 39317738 PMCID: PMC11525184 DOI: 10.1038/s41588-024-01896-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 08/06/2024] [Indexed: 09/26/2024]
Abstract
Studying the genetic regulation of protein expression (through protein quantitative trait loci (pQTLs)) offers a deeper understanding of regulatory variants uncharacterized by mRNA expression regulation (expression QTLs (eQTLs)) studies. Here we report cis-eQTL and cis-pQTL statistical fine-mapping from 1,405 genotyped samples with blood mRNA and 2,932 plasma samples of protein expression, as part of the Japan COVID-19 Task Force (JCTF). Fine-mapped eQTLs (n = 3,464) were enriched for 932 variants validated with a massively parallel reporter assay. Fine-mapped pQTLs (n = 582) were enriched for missense variations on structured and extracellular domains, although the possibility of epitope-binding artifacts remains. Trans-eQTL and trans-pQTL analysis highlighted associations of class I HLA allele variation with KIR genes. We contrast the multi-tissue origin of plasma protein with blood mRNA, contributing to the limited colocalization level, distinct regulatory mechanisms and trait relevance of eQTLs and pQTLs. We report a negative correlation between ABO mRNA and protein expression because of linkage disequilibrium between distinct nearby eQTLs and pQTLs.
Collapse
Affiliation(s)
- Qingbo S Wang
- Department of Genome Informatics, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan.
| | - Takanori Hasegawa
- M&D Data Science Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ho Namkoong
- Department of Infectious Diseases, Keio University School of Medicine, Tokyo, Japan.
| | - Ryunosuke Saiki
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | - Ryuya Edahiro
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kyuto Sonehara
- Department of Genome Informatics, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hiromu Tanaka
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shuhei Azekawa
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shotaro Chubachi
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | | | - Saori Sakaue
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shinichi Namba
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Kenichi Yamamoto
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory of Children's Health and Genetics, Division of Health Science, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yuichi Shiraishi
- Division of Genome Analysis Platform Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Kenichi Chiba
- Division of Genome Analysis Platform Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Hiroko Tanaka
- M&D Data Science Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hideki Makishima
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | - Yasuhito Nannya
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | - Zicong Zhang
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Rika Tsujikawa
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Ryuji Koike
- Health Science Research and Development Center (HeRD), Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomomi Takano
- Laboratory of Veterinary Infectious Disease, Department of Veterinary Medicine, Kitasato University, Tokyo, Japan
| | - Makoto Ishii
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akinori Kimura
- Institute of Research, Tokyo Medical and Dental University, Tokyo, Japan
| | - Fumitaka Inoue
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, the Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Satoru Miyano
- M&D Data Science Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yukinori Okada
- Department of Genome Informatics, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan.
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan.
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Japan.
| |
Collapse
|
11
|
Sarnowski C, Ma J, Nguyen NQH, Hoogeveen RC, Ballantyne CM, Coresh J, Morrison AC, Chatterjee N, Boerwinkle E, Yu B. Ancestrally diverse genome-wide association analysis highlights ancestry-specific differences in genetic regulation of plasma protein levels. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.27.24314500. [PMID: 39399032 PMCID: PMC11469718 DOI: 10.1101/2024.09.27.24314500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Fully characterizing the genetic architecture of circulating proteins in multi-ancestry populations provides an unprecedented opportunity to gain insights into the etiology of complex diseases. We characterized and contrasted the genetic associations of plasma proteomes in 9,455 participants of European and African (19.8%) ancestry from the Atherosclerosis Risk in Communities Study. Of 4,651 proteins, 1,408 and 2,565 proteins had protein-quantitative trait loci (pQTLs) identified in African and European ancestry respectively, and twelve unreported potentially causal protein-disease relationships were identified. Shared pQTLs across the two ancestries were detected in 1,113 aptamer-region pairs pQTLs, where 53 of them were not previously reported (all trans pQTLs). Sixteen unique protein-cardiovascular trait pairs were colocalized in both European and African ancestry with the same candidate causal variants. Our systematic cross-ancestry comparison provided a reliable set of pQTLs, highlighted the shared and distinct genetic architecture of proteome in two ancestries, and demonstrated possible biological mechanisms underlying complex diseases.
Collapse
Affiliation(s)
- Chloé Sarnowski
- Department of Epidemiology, The University of Texas Health Science Center at Houston, School of Public Health, Houston, TX
| | - Jianzhong Ma
- Department of Epidemiology, The University of Texas Health Science Center at Houston, School of Public Health, Houston, TX
| | - Ngoc Quynh H. Nguyen
- Department of Epidemiology, The University of Texas Health Science Center at Houston, School of Public Health, Houston, TX
| | - Ron C Hoogeveen
- Department of Medicine, Baylor College of Medicine, Houston, TX
| | | | - Josef Coresh
- Optimal Aging Institute, New York University Grossman School of Medicine, New York, NY
- Department of Population Health, New York University Grossman School of Medicine, New York, NY
| | - Alanna C Morrison
- Department of Epidemiology, The University of Texas Health Science Center at Houston, School of Public Health, Houston, TX
| | - Nilanjan Chatterjee
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Eric Boerwinkle
- Department of Epidemiology, The University of Texas Health Science Center at Houston, School of Public Health, Houston, TX
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX
| | - Bing Yu
- Department of Epidemiology, The University of Texas Health Science Center at Houston, School of Public Health, Houston, TX
| |
Collapse
|
12
|
Tahir UA, Barber JL, Cruz DE, Kars ME, Deng S, Tuftin B, Gillman MG, Benson MD, Robbins JM, Chen ZZ, Rao P, Katz DH, Farrell L, Sofer T, Hall ME, Ekunwe L, Tracy RP, Durda P, Taylor KD, Liu Y, Johnson WC, Guo X, Chen YDI, Manichaikul AW, Jain D, Wang TJ, Reiner AP, Natarajan P, Itan Y, Rich SS, Rotter JI, Wilson JG, Raffield LM, Gerszten RE. Proteogenomic analysis integrated with electronic health records data reveals disease-associated variants in Black Americans. J Clin Invest 2024; 134:e181802. [PMID: 39316441 PMCID: PMC11527441 DOI: 10.1172/jci181802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 09/11/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUNDMost GWAS of plasma proteomics have focused on White individuals of European ancestry, limiting biological insight from other ancestry-enriched protein quantitative loci (pQTL).METHODSWe conducted a discovery GWAS of approximately 3,000 plasma proteins measured by the antibody-based Olink platform in 1,054 Black adults from the Jackson Heart Study (JHS) and validated our findings in the Multi-Ethnic Study of Atherosclerosis (MESA). The genetic architecture of identified pQTLs was further explored through fine mapping and admixture association analysis. Finally, using our pQTL findings, we performed a phenome-wide association study (PheWAS) across 2 large multiethnic electronic health record (EHR) systems in All of Us and BioMe.RESULTSWe identified 1,002 pQTLs for 925 protein assays. Fine mapping and admixture analyses suggested allelic heterogeneity of the plasma proteome across diverse populations. We identified associations for variants enriched in African ancestry, many in diseases that lack precise biomarkers, including cis-pQTLs for cathepsin L (CTSL) and Siglec-9, which were linked with sarcoidosis and non-Hodgkin's lymphoma, respectively. We found concordant associations across clinical diagnoses and laboratory measurements, elucidating disease pathways, including a cis-pQTL associated with circulating CD58, WBC count, and multiple sclerosis.CONCLUSIONSOur findings emphasize the value of leveraging diverse populations to enhance biological insights from proteomics GWAS, and we have made this resource readily available as an interactive web portal.FUNDINGNIH K08 HL161445-01A1; 5T32HL160522-03; HHSN268201600034I; HL133870.
Collapse
Affiliation(s)
- Usman A. Tahir
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Jacob L. Barber
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel E. Cruz
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Shuliang Deng
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Madeline G. Gillman
- University of North Carolina School of Medicine, Raleigh, North Carolina, USA
| | - Mark D. Benson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Jeremy M. Robbins
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Zsu-Zsu Chen
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Prashant Rao
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Laurie Farrell
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Tamar Sofer
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael E. Hall
- University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Lynette Ekunwe
- University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Russell P. Tracy
- Department of Pathology Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Peter Durda
- Department of Pathology Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation, Torrance, California, USA
| | - Yongmei Liu
- Department of Medicine, Division of Cardiology, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - W. Craig Johnson
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation, Torrance, California, USA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation, Torrance, California, USA
| | - Ani W. Manichaikul
- Center for Public Health Genomics and
- Division of Biostatistics and Epidemiology, Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, USA
| | - Deepti Jain
- University of Washington, Seattle, Washington
| | | | - Thomas J. Wang
- Department of Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | | | - Pradeep Natarajan
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Yuval Itan
- University of North Carolina School of Medicine, Raleigh, North Carolina, USA
| | | | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation, Torrance, California, USA
| | - James G. Wilson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Laura M. Raffield
- University of North Carolina School of Medicine, Raleigh, North Carolina, USA
| | - Robert E. Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| |
Collapse
|
13
|
Gurung RL, Zheng H, Lee BTK, Liu S, Liu JJ, Chan C, Ang K, Subramaniam T, Sum CF, Coffman TM, Lim SC. Proteomics profiling and association with cardiorenal complications in type 2 diabetes subtypes in Asian population. Diabetes Res Clin Pract 2024; 214:111790. [PMID: 39059739 DOI: 10.1016/j.diabres.2024.111790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/09/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024]
Abstract
AIM Among multi-ethnic Asians, type 2 diabetes (T2D) clustered in three subtypes; mild obesity-related diabetes (MOD), mild age-related diabetes with insulin insufficiency (MARD-II) and severe insulin-resistant diabetes with relative insulin insufficiency (SIRD-RII) had differential cardio-renal complication risk. We assessed the proteomic profiles to identify subtype specific biomarkers and its association with diabetes complications. METHODS 1448 plasma proteins at baseline were measured and compared across the T2D subtypes. Multivariable cox regression was used to assess associations between significant proteomics features and cardio-renal complications. RESULTS Among 645 T2D participants (SIRD-RII [19%], MOD [45%], MARD-II [36%]), 295 proteins expression differed significantly across the groups. These proteins were enriched in cell adhesion, neurogenesis and inflammatory response processes. In SIRD-RII group, ADH4, ACY1, THOP1, IGFBP2, NEFL, ENTPD2, CALB1, HAO1, CTSV, ITGAV, SCLY, EDA2R, ERBB2 proteins significantly associated with progressive CKD and LILRA5 protein with incident heart failure (HF). In MOD group, TAFA5, RSPO3, EDA2R proteins significantly associated with incident HF. In MARD-II group, FABP4 protein significantly associated with progressive CKD and PTPRN2 protein with major adverse cardiovascular events. Genetically determined NEFL and CALB1 were associated with kidney function decline. CONCLUSIONS Each T2D subtype has unique proteomics signature and association with clinical outcomes and underlying mechanisms.
Collapse
Affiliation(s)
- Resham Lal Gurung
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore; Cardiovascular and Metabolic Disorders Signature Research Program, Duke-NUS Medical School, Singapore
| | - Huili Zheng
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore
| | | | - Sylvia Liu
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore
| | - Jian-Jun Liu
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore
| | - Clara Chan
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore
| | - Keven Ang
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore
| | | | - Chee Fang Sum
- Diabetes Centre, Admiralty Medical Centre, Singapore
| | - Thomas M Coffman
- Cardiovascular and Metabolic Disorders Signature Research Program, Duke-NUS Medical School, Singapore
| | - Su Chi Lim
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore; Lee Kong Chian School of Medicine, Nanyang Technology University, Singapore; Diabetes Centre, Admiralty Medical Centre, Singapore; Saw Swee Hock School of Public Heath, Singapore.
| |
Collapse
|
14
|
Nam Y, Kim J, Jung SH, Woerner J, Suh EH, Lee DG, Shivakumar M, Lee ME, Kim D. Harnessing Artificial Intelligence in Multimodal Omics Data Integration: Paving the Path for the Next Frontier in Precision Medicine. Annu Rev Biomed Data Sci 2024; 7:225-250. [PMID: 38768397 PMCID: PMC11972123 DOI: 10.1146/annurev-biodatasci-102523-103801] [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: 05/22/2024]
Abstract
The integration of multiomics data with detailed phenotypic insights from electronic health records marks a paradigm shift in biomedical research, offering unparalleled holistic views into health and disease pathways. This review delineates the current landscape of multimodal omics data integration, emphasizing its transformative potential in generating a comprehensive understanding of complex biological systems. We explore robust methodologies for data integration, ranging from concatenation-based to transformation-based and network-based strategies, designed to harness the intricate nuances of diverse data types. Our discussion extends from incorporating large-scale population biobanks to dissecting high-dimensional omics layers at the single-cell level. The review underscores the emerging role of large language models in artificial intelligence, anticipating their influence as a near-future pivot in data integration approaches. Highlighting both achievements and hurdles, we advocate for a concerted effort toward sophisticated integration models, fortifying the foundation for groundbreaking discoveries in precision medicine.
Collapse
Affiliation(s)
- Yonghyun Nam
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Jaesik Kim
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sang-Hyuk Jung
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Jakob Woerner
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Erica H Suh
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Dong-Gi Lee
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Manu Shivakumar
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Matthew E Lee
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Dokyoon Kim
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| |
Collapse
|
15
|
Ma S, Xu F, Fu Y, Zheng JS. Genetic mapping of plasma proteome on brain structure. J Genet Genomics 2024; 51:774-777. [PMID: 38608937 DOI: 10.1016/j.jgg.2024.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/28/2024] [Accepted: 03/31/2024] [Indexed: 04/14/2024]
Affiliation(s)
- Shengyi Ma
- Fudan University, Shanghai 200433, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310030, China
| | - Fengzhe Xu
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310030, China
| | - Yuanqing Fu
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310030, China.
| | - Ju-Sheng Zheng
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310030, China.
| |
Collapse
|
16
|
Carrasco-Zanini J, Pietzner M, Koprulu M, Wheeler E, Kerrison ND, Wareham NJ, Langenberg C. Proteomic prediction of diverse incident diseases: a machine learning-guided biomarker discovery study using data from a prospective cohort study. Lancet Digit Health 2024; 6:e470-e479. [PMID: 38906612 DOI: 10.1016/s2589-7500(24)00087-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/03/2024] [Accepted: 04/19/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Broad-capture proteomic technologies have the potential to improve disease prediction, enabling targeted prevention and management, but studies have so far been limited to very few selected diseases and have not evaluated predictive performance across multiple conditions. We aimed to evaluate the potential of serum proteins to improve risk prediction over and above health-derived information and polygenic risk scores across a diverse set of 24 outcomes. METHODS We designed multiple case-cohorts nested in the EPIC-Norfolk prospective study, from participants with available serum samples and genome-wide genotype data, with more than 32 974 person-years of follow-up. Participants were middle-aged individuals (aged 40-79 years at baseline) of European ancestry who were recruited from the general population of Norfolk, England, between March, 1993 and December, 1997. We selected participants who developed one of ten less common diseases within 10 years of follow-up; we also subsampled a randomly drawn control subcohort, which also served to investigate 14 more common outcomes (n>70), including all-cause premature mortality (death before the age of 75 years; case numbers 71-437; controls 608-1556). Individuals were excluded from the current study owing to failed genotyping or proteomic quality control, relatedness, or missing information on age, sex, BMI, or smoking status. We used a machine learning framework to derive sparse predictive protein models for the onset of the the 23 individual diseases and all-cause premature mortality, and to derive a single common sparse multimorbidity signature that was predictive across multiple diseases from 2923 serum proteins. FINDINGS Participants who developed one of ten less common diseases within 10 years of follow-up included 482 women and 507 men, with a mean age at baseline of 64·56 years (8·08). The random subcohort included 990 women and 769 men, with a mean age of 58·79 years (9·31). As few as five proteins alone outperformed polygenic risk scores for 17 of 23 outcomes (median dfference in concordance index [C-index] 0·13 [0·10-0·17]) and improved predictive performance when added over basic patient-derived information models for seven outcomes, achieving a median C-index of 0·82 (IQR 0·77-0·82). This included diseases with poor prognosis such as lung cancer (C-index 0·85 [+/- cross-validation error 0·83-0·87]), for which we identified unreported biomarkers such as C-X-C motif chemokine ligand 17. A sparse multimorbidity signature of ten proteins improved prediction across seven outcomes over patient-derived information models, achieving performances (median C-index 0·81 [IQR 0·80-0·82]) similar to those of disease-specific signatures. INTERPRETATION We show the value of broad-capture proteomic biomarker discovery studies across multiple diseases of diverse causes, pointing to those that might benefit the most from proteomic approaches, and the potential to derive common sparse biomarker panels for prediction of multiple diseases at once. This framework could enable follow-up studies to explore the generalisability of proteomic models and to benchmark these against clinical assays, which are required to understand the translational potential of these findings. FUNDING Medical Research Council, Health Data Research UK, UK Research and Innovation-National Institute for Health and Care Research, Cancer Research UK, and Wellcome Trust.
Collapse
Affiliation(s)
- Julia Carrasco-Zanini
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK; Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
| | - Maik Pietzner
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK; Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany; Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
| | - Mine Koprulu
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Eleanor Wheeler
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Nicola D Kerrison
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK; Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany; Precision Healthcare University Research Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
17
|
Pietzner M, Uluvar B, Kolnes KJ, Jeppesen PB, Frivold SV, Skattebo Ø, Johansen EI, Skålhegg BS, Wojtaszewski JFP, Kolnes AJ, Yeo GSH, O'Rahilly S, Jensen J, Langenberg C. Systemic proteome adaptions to 7-day complete caloric restriction in humans. Nat Metab 2024; 6:764-777. [PMID: 38429390 PMCID: PMC7617311 DOI: 10.1038/s42255-024-01008-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 02/01/2024] [Indexed: 03/03/2024]
Abstract
Surviving long periods without food has shaped human evolution. In ancient and modern societies, prolonged fasting was/is practiced by billions of people globally for religious purposes, used to treat diseases such as epilepsy, and recently gained popularity as weight loss intervention, but we still have a very limited understanding of the systemic adaptions in humans to extreme caloric restriction of different durations. Here we show that a 7-day water-only fast leads to an average weight loss of 5.7 kg (±0.8 kg) among 12 volunteers (5 women, 7 men). We demonstrate nine distinct proteomic response profiles, with systemic changes evident only after 3 days of complete calorie restriction based on in-depth characterization of the temporal trajectories of ~3,000 plasma proteins measured before, daily during, and after fasting. The multi-organ response to complete caloric restriction shows distinct effects of fasting duration and weight loss and is remarkably conserved across volunteers with >1,000 significantly responding proteins. The fasting signature is strongly enriched for extracellular matrix proteins from various body sites, demonstrating profound non-metabolic adaptions, including extreme changes in the brain-specific extracellular matrix protein tenascin-R. Using proteogenomic approaches, we estimate the health consequences for 212 proteins that change during fasting across ~500 outcomes and identified putative beneficial (SWAP70 and rheumatoid arthritis or HYOU1 and heart disease), as well as adverse effects. Our results advance our understanding of prolonged fasting in humans beyond a merely energy-centric adaptions towards a systemic response that can inform targeted therapeutic modulation.
Collapse
Affiliation(s)
- Maik Pietzner
- Computational Medicine, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK.
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK.
| | - Burulça Uluvar
- Computational Medicine, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Kristoffer J Kolnes
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
| | - Per B Jeppesen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - S Victoria Frivold
- Institute of Health and Society, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Øyvind Skattebo
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Egil I Johansen
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Bjørn S Skålhegg
- Department of Nutrition, Division for Molecular Nutrition, University of Oslo, Oslo, Norway
| | - Jørgen F P Wojtaszewski
- August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Anders J Kolnes
- Section of Specialized Endocrinology, Department of Endocrinology, Oslo University Hospital, Oslo, Norway
| | - Giles S H Yeo
- Metabolic Research Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Stephen O'Rahilly
- Metabolic Research Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Jørgen Jensen
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Claudia Langenberg
- Computational Medicine, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK.
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK.
| |
Collapse
|
18
|
Saint-André V, Charbit B, Biton A, Rouilly V, Possémé C, Bertrand A, Rotival M, Bergstedt J, Patin E, Albert ML, Quintana-Murci L, Duffy D. Smoking changes adaptive immunity with persistent effects. Nature 2024; 626:827-835. [PMID: 38355791 PMCID: PMC10881394 DOI: 10.1038/s41586-023-06968-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/13/2023] [Indexed: 02/16/2024]
Abstract
Individuals differ widely in their immune responses, with age, sex and genetic factors having major roles in this inherent variability1-6. However, the variables that drive such differences in cytokine secretion-a crucial component of the host response to immune challenges-remain poorly defined. Here we investigated 136 variables and identified smoking, cytomegalovirus latent infection and body mass index as major contributors to variability in cytokine response, with effects of comparable magnitudes with age, sex and genetics. We find that smoking influences both innate and adaptive immune responses. Notably, its effect on innate responses is quickly lost after smoking cessation and is specifically associated with plasma levels of CEACAM6, whereas its effect on adaptive responses persists long after individuals quit smoking and is associated with epigenetic memory. This is supported by the association of the past smoking effect on cytokine responses with DNA methylation at specific signal trans-activators and regulators of metabolism. Our findings identify three novel variables associated with cytokine secretion variability and reveal roles for smoking in the short- and long-term regulation of immune responses. These results have potential clinical implications for the risk of developing infections, cancers or autoimmune diseases.
Collapse
Affiliation(s)
- Violaine Saint-André
- Translational Immunology Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, Paris, France.
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, France.
| | - Bruno Charbit
- Cytometry and Biomarkers UTechS, Center for Translational Research, Institut Pasteur, Université Paris Cité, Paris, France
| | - Anne Biton
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, France
| | | | - Céline Possémé
- Translational Immunology Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Anthony Bertrand
- Translational Immunology Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, Paris, France
- Frontiers of Innovation in Research and Education PhD Program, LPI Doctoral School, Université Paris Cité, Paris, France
| | - Maxime Rotival
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Human Evolutionary Genetics Unit, Paris, France
| | - Jacob Bergstedt
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Human Evolutionary Genetics Unit, Paris, France
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Etienne Patin
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Human Evolutionary Genetics Unit, Paris, France
| | | | - Lluis Quintana-Murci
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Human Evolutionary Genetics Unit, Paris, France
- Chair Human Genomics and Evolution, Collège de France, Paris, France
| | - Darragh Duffy
- Translational Immunology Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, Paris, France.
- Cytometry and Biomarkers UTechS, Center for Translational Research, Institut Pasteur, Université Paris Cité, Paris, France.
| |
Collapse
|
19
|
Tian W, Shi D, Zhang Y, Wang H, Tang H, Han Z, Wong CCL, Cui L, Zheng J, Chen Y. Deep proteomic analysis of obstetric antiphospholipid syndrome by DIA-MS of extracellular vesicle enriched fractions. Commun Biol 2024; 7:99. [PMID: 38225453 PMCID: PMC10789860 DOI: 10.1038/s42003-024-05789-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 01/03/2024] [Indexed: 01/17/2024] Open
Abstract
Proteins in the plasma/serum mirror an individual's physiology. Circulating extracellular vesicles (EVs) proteins constitute a large portion of the plasma/serum proteome. Thus, deep and unbiased proteomic analysis of circulating plasma/serum extracellular vesicles holds promise for discovering disease biomarkers as well as revealing disease mechanisms. We established a workflow for simple, deep, and reproducible proteome analysis of both serum large and small EVs enriched fractions by ultracentrifugation plus 4D-data-independent acquisition mass spectrometry (4D-DIA-MS). In our cohort study of obstetric antiphospholipid syndrome (OAPS), 4270 and 3328 proteins were identified from large and small EVs enriched fractions respectively. Both of them revealed known or new pathways related to OAPS. Increased levels of von Willebrand factor (VWF) and insulin receptor (INSR) were identified as candidate biomarkers, which shed light on hypercoagulability and abnormal insulin signaling in disease progression. Our workflow will significantly promote our understanding of plasma/serum-based disease mechanisms and generate new biomarkers.
Collapse
Affiliation(s)
- Wenmin Tian
- Department of Biochemistry and Biophysics, Center for Precision Medicine Multi-Omics Research, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Dongxue Shi
- Department of Biochemistry and Biophysics, Center for Precision Medicine Multi-Omics Research, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yinmei Zhang
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, P R China
| | - Hongli Wang
- Department of Biochemistry and Biophysics, Center for Precision Medicine Multi-Omics Research, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Haohao Tang
- Department of Biochemistry and Biophysics, Center for Precision Medicine Multi-Omics Research, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Zhongyu Han
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, P R China
| | - Catherine C L Wong
- Department of Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, 100730, Beijing, China
- Tsinghua University-Peking University Joint Center for Life Sciences, Peking University, 100084, Beijing, China
| | - Liyan Cui
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, P R China.
| | - Jiajia Zheng
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, P R China.
| | - Yang Chen
- Department of Biochemistry and Biophysics, Center for Precision Medicine Multi-Omics Research, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
20
|
Eldjarn GH, Ferkingstad E, Lund SH, Helgason H, Magnusson OT, Gunnarsdottir K, Olafsdottir TA, Halldorsson BV, Olason PI, Zink F, Gudjonsson SA, Sveinbjornsson G, Magnusson MI, Helgason A, Oddsson A, Halldorsson GH, Magnusson MK, Saevarsdottir S, Eiriksdottir T, Masson G, Stefansson H, Jonsdottir I, Holm H, Rafnar T, Melsted P, Saemundsdottir J, Norddahl GL, Thorleifsson G, Ulfarsson MO, Gudbjartsson DF, Thorsteinsdottir U, Sulem P, Stefansson K. Large-scale plasma proteomics comparisons through genetics and disease associations. Nature 2023; 622:348-358. [PMID: 37794188 PMCID: PMC10567571 DOI: 10.1038/s41586-023-06563-x] [Citation(s) in RCA: 124] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 08/22/2023] [Indexed: 10/06/2023]
Abstract
High-throughput proteomics platforms measuring thousands of proteins in plasma combined with genomic and phenotypic information have the power to bridge the gap between the genome and diseases. Here we performed association studies of Olink Explore 3072 data generated by the UK Biobank Pharma Proteomics Project1 on plasma samples from more than 50,000 UK Biobank participants with phenotypic and genotypic data, stratifying on British or Irish, African and South Asian ancestries. We compared the results with those of a SomaScan v4 study on plasma from 36,000 Icelandic people2, for 1,514 of whom Olink data were also available. We found modest correlation between the two platforms. Although cis protein quantitative trait loci were detected for a similar absolute number of assays on the two platforms (2,101 on Olink versus 2,120 on SomaScan), the proportion of assays with such supporting evidence for assay performance was higher on the Olink platform (72% versus 43%). A considerable number of proteins had genomic associations that differed between the platforms. We provide examples where differences between platforms may influence conclusions drawn from the integration of protein levels with the study of diseases. We demonstrate how leveraging the diverse ancestries of participants in the UK Biobank helps to detect novel associations and refine genomic location. Our results show the value of the information provided by the two most commonly used high-throughput proteomics platforms and demonstrate the differences between them that at times provides useful complementarity.
Collapse
Affiliation(s)
| | | | - Sigrun H Lund
- deCODE Genetics/Amgen, Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | - Hannes Helgason
- deCODE Genetics/Amgen, Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | | | - Bjarni V Halldorsson
- deCODE Genetics/Amgen, Reykjavik, Iceland
- School of Technology, Reykjavik University, Reykjavik, Iceland
| | | | | | | | | | | | - Agnar Helgason
- deCODE Genetics/Amgen, Reykjavik, Iceland
- Department of Anthropology, University of Iceland, Reykjavik, Iceland
| | | | | | - Magnus K Magnusson
- deCODE Genetics/Amgen, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Saedis Saevarsdottir
- deCODE Genetics/Amgen, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | | | - Ingileif Jonsdottir
- deCODE Genetics/Amgen, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Hilma Holm
- deCODE Genetics/Amgen, Reykjavik, Iceland
| | | | - Pall Melsted
- deCODE Genetics/Amgen, Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | | | - Magnus O Ulfarsson
- deCODE Genetics/Amgen, Reykjavik, Iceland
- Faculty of Electrical and Computer Engineering, University of Iceland, Reykjavik, Iceland
| | - Daniel F Gudbjartsson
- deCODE Genetics/Amgen, Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | - Unnur Thorsteinsdottir
- deCODE Genetics/Amgen, Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | - Kari Stefansson
- deCODE Genetics/Amgen, Reykjavik, Iceland.
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.
| |
Collapse
|
21
|
Sun BB, Chiou J, Traylor M, Benner C, Hsu YH, Richardson TG, Surendran P, Mahajan A, Robins C, Vasquez-Grinnell SG, Hou L, Kvikstad EM, Burren OS, Davitte J, Ferber KL, Gillies CE, Hedman ÅK, Hu S, Lin T, Mikkilineni R, Pendergrass RK, Pickering C, Prins B, Baird D, Chen CY, Ward LD, Deaton AM, Welsh S, Willis CM, Lehner N, Arnold M, Wörheide MA, Suhre K, Kastenmüller G, Sethi A, Cule M, Raj A, Burkitt-Gray L, Melamud E, Black MH, Fauman EB, Howson JMM, Kang HM, McCarthy MI, Nioi P, Petrovski S, Scott RA, Smith EN, Szalma S, Waterworth DM, Mitnaul LJ, Szustakowski JD, Gibson BW, Miller MR, Whelan CD. Plasma proteomic associations with genetics and health in the UK Biobank. Nature 2023; 622:329-338. [PMID: 37794186 PMCID: PMC10567551 DOI: 10.1038/s41586-023-06592-6] [Citation(s) in RCA: 361] [Impact Index Per Article: 180.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/31/2023] [Indexed: 10/06/2023]
Abstract
The Pharma Proteomics Project is a precompetitive biopharmaceutical consortium characterizing the plasma proteomic profiles of 54,219 UK Biobank participants. Here we provide a detailed summary of this initiative, including technical and biological validations, insights into proteomic disease signatures, and prediction modelling for various demographic and health indicators. We present comprehensive protein quantitative trait locus (pQTL) mapping of 2,923 proteins that identifies 14,287 primary genetic associations, of which 81% are previously undescribed, alongside ancestry-specific pQTL mapping in non-European individuals. The study provides an updated characterization of the genetic architecture of the plasma proteome, contextualized with projected pQTL discovery rates as sample sizes and proteomic assay coverages increase over time. We offer extensive insights into trans pQTLs across multiple biological domains, highlight genetic influences on ligand-receptor interactions and pathway perturbations across a diverse collection of cytokines and complement networks, and illustrate long-range epistatic effects of ABO blood group and FUT2 secretor status on proteins with gastrointestinal tissue-enriched expression. We demonstrate the utility of these data for drug discovery by extending the genetic proxied effects of protein targets, such as PCSK9, on additional endpoints, and disentangle specific genes and proteins perturbed at loci associated with COVID-19 susceptibility. This public-private partnership provides the scientific community with an open-access proteomics resource of considerable breadth and depth to help to elucidate the biological mechanisms underlying proteo-genomic discoveries and accelerate the development of biomarkers, predictive models and therapeutics1.
Collapse
Affiliation(s)
- Benjamin B Sun
- Translational Sciences, Research & Development, Biogen, Cambridge, MA, USA.
| | - Joshua Chiou
- Internal Medicine Research Unit, Worldwide Research, Development and Medical, Pfizer, Cambridge, MA, USA
| | - Matthew Traylor
- Human Genetics Centre of Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
| | | | | | - Tom G Richardson
- Human Genetics Centre of Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
- Genomic Sciences, GlaxoSmithKline, Stevenage, UK
| | | | | | - Chloe Robins
- Genomic Sciences, GlaxoSmithKline, Collegeville, PA, USA
| | | | - Liping Hou
- Population Analytics, Janssen Research & Development, Spring House, PA, USA
| | | | - Oliver S Burren
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | - Kyle L Ferber
- Biostatistics, Research and Development, Biogen, Cambridge, MA, USA
| | | | - Åsa K Hedman
- External Science and Innovation Target Sciences, Worldwide Research, Development and Medical, Pfizer, Stockholm, Sweden
| | - Sile Hu
- Human Genetics Centre of Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - Tinchi Lin
- Analytics and Data Sciences, Biogen, Cambridge, MA, USA
| | - Rajesh Mikkilineni
- Data Science Institute, Takeda Development Center Americas, Cambridge, MA, USA
| | | | | | - Bram Prins
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Denis Baird
- Translational Sciences, Research & Development, Biogen, Cambridge, MA, USA
| | - Chia-Yen Chen
- Translational Sciences, Research & Development, Biogen, Cambridge, MA, USA
| | - Lucas D Ward
- Alnylam Human Genetics, Discovery & Translational Research, Alnylam Pharmaceuticals, Cambridge, MA, USA
| | - Aimee M Deaton
- Alnylam Human Genetics, Discovery & Translational Research, Alnylam Pharmaceuticals, Cambridge, MA, USA
| | | | - Carissa M Willis
- Alnylam Human Genetics, Discovery & Translational Research, Alnylam Pharmaceuticals, Cambridge, MA, USA
| | - Nick Lehner
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Matthias Arnold
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC, USA
| | - Maria A Wörheide
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Karsten Suhre
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | | | | | - Anil Raj
- Calico Life Sciences, San Francisco, CA, USA
| | | | | | - Mary Helen Black
- Population Analytics, Janssen Research & Development, Spring House, PA, USA
| | - Eric B Fauman
- Internal Medicine Research Unit, Worldwide Research, Development and Medical, Pfizer, Cambridge, MA, USA
| | - Joanna M M Howson
- Human Genetics Centre of Excellence, Novo Nordisk Research Centre Oxford, Oxford, UK
| | | | | | - Paul Nioi
- Alnylam Human Genetics, Discovery & Translational Research, Alnylam Pharmaceuticals, Cambridge, MA, USA
| | - Slavé Petrovski
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
- Department of Medicine, University of Melbourne, Austin Health, Melbourne, Victoria, Australia
| | | | - Erin N Smith
- Takeda Development Center Americas, San Diego, CA, USA
| | - Sándor Szalma
- Takeda Development Center Americas, San Diego, CA, USA
| | | | | | | | | | - Melissa R Miller
- Internal Medicine Research Unit, Worldwide Research, Development and Medical, Pfizer, Cambridge, MA, USA
| | - Christopher D Whelan
- Translational Sciences, Research & Development, Biogen, Cambridge, MA, USA.
- Neuroscience Data Science, Janssen Research & Development, Cambridge, MA, USA.
| |
Collapse
|
22
|
Langenberg C, Hingorani AD, Whitty CJM. Biological and functional multimorbidity-from mechanisms to management. Nat Med 2023; 29:1649-1657. [PMID: 37464031 DOI: 10.1038/s41591-023-02420-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/23/2023] [Indexed: 07/20/2023]
Abstract
Globally, the number of people with multiple co-occurring diseases will increase substantially over the coming decades, with important consequences for patients, carers, healthcare systems and society. Addressing this challenge requires a shift in the prevailing clinical, educational and scientific thinking and organization-with a strong emphasis on the maintenance of generalist skills to balance the specialization trends of medical education and research. Multimorbidity is not a single entity but differs quantitively and qualitatively across life stages, ethnicities, sexes, socioeconomic groups and geographies. Data-driven science that quantifies the impact of disease co-occurrence-beyond the small number of currently well-studied long-term conditions (such as cardiometabolic diseases)-can help illuminate the pathological diversity of multimorbidity and identify common, mechanistically related, and prognostically relevant clusters. Broader access to data opportunities across modalities and disciplines will catalyze vertical and horizontal integration of multimorbidity research, to enable reconfiguring of medical services, clinical trials, guidelines and research in a way that accounts for the complexity of multimorbidity-and provides efficient, joined-up services for patients.
Collapse
Affiliation(s)
- Claudia Langenberg
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK.
- Computational Medicine, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge, UK.
| | - Aroon D Hingorani
- UCL BHF Research Accelerator, University College London, London, UK
- Institute of Cardiovascular Science, University College London, London, UK
- University College London Hospitals NIHR Biomedical Research Centre, London, UK
| | - Christopher J M Whitty
- Department of Health and Social Care, London, UK
- London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
23
|
Coral DE, Franks PW. Proteogenomic mapping sets stage for precision medicine. Nat Metab 2023; 5:366-367. [PMID: 36823472 DOI: 10.1038/s42255-023-00759-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Affiliation(s)
- Daniel E Coral
- Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Malmo, Sweden
| | - Paul W Franks
- Lund University Diabetes Centre, Department of Clinical Sciences, Lund University, Malmo, Sweden.
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
- Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|