1
|
El-Husseini ZW, Karp T, Lan A, Gillett TE, Qi C, Khalenkow D, van der Molen T, Brightling C, Papi A, Rabe KF, Siddiqui S, Singh D, Kraft M, Beghé B, Joubert P, Bossé Y, Sin D, Cordero AH, Timens W, Brandsma CA, Hao K, Nickle DC, Vonk JM, Nawijn MC, van den Berge M, Gosens R, Faiz A, Koppelman GH. Improved Annotation of Asthma Gene Variants with Cell Type Deconvolution of Nasal and Lung Expression Quantitative Trait Loci. Am J Respir Cell Mol Biol 2025; 72:607-614. [PMID: 39836087 DOI: 10.1165/rcmb.2024-0251ma] [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: 05/30/2024] [Accepted: 01/21/2025] [Indexed: 01/22/2025] Open
Abstract
Asthma is a genetically complex inflammatory airway disease associated with more than 200 SNPs. However, the functional effects of many asthma-associated SNPs in lung and airway epithelial samples are unknown. Here, we aimed to conduct expression quantitative trait loci (eQTL) analysis using a meta-analysis of nasal and lung samples. We hypothesize that incorporating cell type proportions of airway and lung samples enhances eQTL analysis outcomes. Nasal brush (n = 792) and lung tissue (n = 1,087) samples were investigated separately. Initially, a general eQTL analysis identified genetic variants associated with gene expression levels. Estimated cell type proportions were adjusted based on the Human Lung Cell Atlas. In addition, the presence of significant interaction effects between asthma-associated SNPs and each cell type proportion was explored and considered evidence for cell type-associated eQTL. In nasal brush and lung parenchyma samples, 44 and 116 asthma-associated SNPs were identified as eQTL. Adjusting for cell type proportions revealed eQTL for an additional 17 genes (e.g., FCER1G, CD200R1, and GABBR2) and 16 genes (e.g., CYP2C8, SLC9A2, and SGCD) in nose and lung, respectively. Moreover, we identified eQTL for nine SNPs annotated to genes such as VASP, FOXA3, and PCDHB12 displayed significant interactions with cell type proportions of club, goblet, and alveolar macrophages. Our findings demonstrate increased power for identifying eQTL among asthma-associated SNPs by considering cell type proportion of the bulk RNA-sequencing data from nasal and lung tissues. Integration of cell type deconvolution and eQTL analysis enhances our understanding of asthma genetics and cellular mechanisms, uncovering potential therapeutic targets for personalized interventions.
Collapse
Affiliation(s)
- Zaid W El-Husseini
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen, the Netherlands
| | - Tatiana Karp
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Department of Pulmonary Diseases
| | - Andy Lan
- Respiratory Bioinformatics and Molecular Biology, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Tessa E Gillett
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Department of Pathology and Medical Biology
| | - Cancan Qi
- Microbiome Medicine Center, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Dmitry Khalenkow
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Laboratory of Genome Structure and Ageing, European Research Institute for the Biology of Ageing, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Chris Brightling
- Department of Infection, Immunity, and Inflammation, Institute for Lung Health, University of Leicester, Leicester, United Kingdom
| | - Alberto Papi
- Department of Respiratory Medicine, University of Ferrara, Ferrara, Italy
| | - Klaus F Rabe
- Department of Medicine, Christian Albrechts University Kiel, Kiel and Lungen Clinic Grosshansdorf, Grosshansdorf, Germany (Members of the German Center for Lung Research [DZL])
| | - Salman Siddiqui
- National Heart and Lung Institute, Imperial College and Imperial NIHR Biomedical Research Centre, London, United Kingdom
| | - Dave Singh
- Medicines Evaluation Unit, Manchester University NHS Foundation Hospital Trust, University of Manchester, Manchester, United Kingdom
| | - Monica Kraft
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Bianca Beghé
- Section of Respiratory Diseases, Department of Oncology, Haematology, and Respiratory Diseases, University of Modena and Reggio Emilia, Policlinico di Modena, Modena, Italy
| | - Philippe Joubert
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Yohan Bossé
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Don Sin
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada; and
| | - Ana H Cordero
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada; and
| | - Wim Timens
- Department of Pathology and Medical Biology
| | - Corry-Anke Brandsma
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Department of Pathology and Medical Biology
| | - Ke Hao
- Merck Research Laboratories, Boston, Massachusetts
| | | | - Judith M Vonk
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Department of Epidemiology, and
| | - Martijn C Nawijn
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Department of Pathology and Medical Biology
| | - Maarten van den Berge
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Department of Pulmonary Diseases
| | - Reinoud Gosens
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen, the Netherlands
| | - Alen Faiz
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Respiratory Bioinformatics and Molecular Biology, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Gerard H Koppelman
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, and
| |
Collapse
|
2
|
Kachroo P, Shutta KH, Maiorino E, Moll M, Hecker J, Carey V, McGeachie MJ, Litonjua AA, Celedón JC, National Heart, Lung, and Blood Institute Trans-Omics for Precision Medicine (TOPMed) Consortium, Weiss ST, DeMeo DL. DNA-methylation markers associated with lung function at birth and childhood reveal early life programming of inflammatory pathways. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.05.12.653131. [PMID: 40462996 PMCID: PMC12132251 DOI: 10.1101/2025.05.12.653131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/16/2025]
Abstract
Rationale Lung function deficits may be caused by early life epigenetic programming. Early childhood studies are necessary to understand life-course trends in lung diseases. Objectives We aimed to examine whether DNA-methylation at birth and childhood is associated with lung function growth. Methods We measured DNA-methylation in leukocytes from participants in two childhood asthma cohorts (CAMP [n=703, mean-age 12.9 years] and GACRS [n=788, mean-age 9.3 years]) and cord blood from participants in the VDAART study (n=572) to identify CpGs and pathways associated with lung function. Results We identified 1,049 consistent differentially methylated CpGs (608 relatively hypermethylated) across all three studies (FDR-P<0.05). Relatively hypomethylated CpGs were enriched for gluconeogenesis, cell adhesion and VEGF signaling. Relatively hypermethylated CpGs were enriched for Hippo, B-cell and growth hormone receptor signaling. Functional enrichment suggested potential regulatory roles for active enhancers and histone modifications. Additionally, enrichment in PI3K/AKT and Notch pathways in males and enrichment in hormonal pathways in females was identified. Gaussian graphical models identified sex-differential DNA-methylation nodes and hub scores at birth and childhood. Integrating with previously identified polygenic risk scores for asthma and drug-target enrichment identified seven robust genes including MPO , CHCHD3, CACNA1S, PI4KA, EP400, CREBBP and KCNA10 with known associations as biomarkers for asthma severity and drug targets for airway inflammation. Conclusions Epigenetic variability from birth through puberty provides mechanistic insights into fetal programming of developmental and immune pathways associated with lung function. These early life observations reveal potential targets for mitigating risk for lung function decline and asthma progression in later life. Key messages We identified consistent DNA methylation signatures between birth and childhood in critical metabolic, lung development and immune pathways that were associated with lung function and may be influenced by sex and genetics.Our integrative findings provide a deeper understanding for accelerated lung function decline across the life-course and could pave the way for translational interventions for lung diseases based on epigenetic plasticity.
Collapse
Affiliation(s)
- Priyadarshini Kachroo
- School of Health Professions, Department of Health Informatics, Rutgers The State University of New Jersey, NJ, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Katherine H. Shutta
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Enrico Maiorino
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Matthew Moll
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Section on Pulmonary, Allergy, Sleep, and Critical Care Medicine, Department of Veterans Affairs, West Roxbury, MA, USA
| | - Julian Hecker
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Vincent Carey
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Michael J. McGeachie
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Augusto A. Litonjua
- Department of Pediatrics, Golisano Children’s Hospital and University of Rochester Medical Center, Rochester, NY, USA
| | - Juan C. Celedón
- Division of Pulmonary Medicine, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
| | | | - Scott T. Weiss
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Dawn L. DeMeo
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| |
Collapse
|
3
|
Donoghue LJ, Benner C, Chang D, Irudayanathan FJ, Pendergrass RK, Yaspan BL, Mahajan A, McCarthy MI. Integration of biobank-scale genetics and plasma proteomics reveals evidence for causal processes in asthma risk and heterogeneity. CELL GENOMICS 2025; 5:100840. [PMID: 40187354 DOI: 10.1016/j.xgen.2025.100840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 01/20/2025] [Accepted: 03/07/2025] [Indexed: 04/07/2025]
Abstract
Hundreds of genetic associations for asthma have been identified, yet translating these findings into mechanistic insights remains challenging. We leveraged plasma proteomics from the UK Biobank Pharma Proteomics Project (UKB-PPP) to identify biomarkers and effectors of asthma risk or heterogeneity using genetic causal inference approaches. We identified 609 proteins associated with asthma status (269 proteins after controlling for body mass index [BMI] and smoking). Analysis of genetically predicted protein levels identified 70 proteins with putative causal roles in asthma risk, including known drug targets and proteins without prior genetic evidence in asthma (e.g., GCHFR, TDRKH, and CLEC7A). The genetic architecture of causally associated proteins provided evidence for a Toll-like receptor (TLR)1-interleukin (IL)-27 asthma axis. Lastly, we identified evidence of causal relationships between proteins and heterogeneous aspects of asthma biology, including between TSPAN8 and neutrophil counts. These findings illustrate that integrating biobank-scale genetics and plasma proteomics can provide a framework to identify therapeutic targets and mechanisms underlying disease risk and heterogeneity.
Collapse
Affiliation(s)
| | | | - Diana Chang
- Genentech, Inc., South San Francisco, CA, USA
| | | | | | | | | | | |
Collapse
|
4
|
Henches L, Kim J, Yang Z, Rubinacci S, Pires G, Albiñana C, Boetto C, Julienne H, Frouin A, Auvergne A, Suzuki Y, Djebali S, Delaneau O, Ganna A, Vilhjálmsson B, Privé F, Aschard H. Polygenic risk score prediction accuracy convergence. HGG ADVANCES 2025; 6:100457. [PMID: 40375557 PMCID: PMC12167061 DOI: 10.1016/j.xhgg.2025.100457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 05/12/2025] [Accepted: 05/12/2025] [Indexed: 05/18/2025] Open
Abstract
Polygenic risk scores (PRSs) models trained from genome-wide association study (GWAS) results are set to play a pivotal role in biomedical research addressing multifactorial human diseases. The prospect of using these risk scores in clinical care and public health is generating both enthusiasm and controversy, with varying opinions among experts about their strengths and limitations. The performance of existing polygenic scores is still limited but is expected to improve with increasing GWAS sample sizes and the development of new, more powerful methods. Theoretically, the variance explained by PRS can be as high as the total additive genetic variance, but it is unclear how much of that variance has already been captured by PRS. Here, we conducted a retrospective analysis to assess progress in PRS prediction accuracy since the publication of the first large-scale GWASs, using data from six common human diseases with sufficient GWAS information. We show that although PRS accuracy has grown rapidly over the years, the pace of improvement from recent GWAS has decreased substantially, suggesting that merely increasing GWAS sample sizes may lead to only modest improvements in risk discrimination. We next investigated the factors influencing the maximum achievable prediction using whole-genome sequencing data from 125,000 UK Biobank participants and state-of-the-art modeling of polygenic outcomes. Our analyses suggest that increasing the variant coverage of PRS, using either more imputed variants or sequencing data, is a key component for future improvements in prediction accuracy.
Collapse
Affiliation(s)
- Léo Henches
- Institut Pasteur, Université de Paris, Department of Computational Biology, 75015 Paris, France
| | - Jihye Kim
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Zhiyu Yang
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Simone Rubinacci
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Gabriel Pires
- Institut Pasteur, Université de Paris, Department of Computational Biology, 75015 Paris, France
| | - Clara Albiñana
- National Centre for Register-Based Research, Aarhus University, 8210 Aarhus, Denmark
| | - Christophe Boetto
- Institut Pasteur, Université de Paris, Department of Computational Biology, 75015 Paris, France
| | - Hanna Julienne
- Institut Pasteur, Université de Paris, Department of Computational Biology, 75015 Paris, France
| | - Arthur Frouin
- Institut Pasteur, Université de Paris, Department of Computational Biology, 75015 Paris, France
| | - Antoine Auvergne
- Institut Pasteur, Université de Paris, Department of Computational Biology, 75015 Paris, France
| | - Yuka Suzuki
- Institut Pasteur, Université de Paris, Department of Computational Biology, 75015 Paris, France
| | - Sarah Djebali
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, University Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Olivier Delaneau
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Andrea Ganna
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Bjarni Vilhjálmsson
- National Centre for Register-Based Research, Aarhus University, 8210 Aarhus, Denmark; Bioinformatics Research Centre, Aarhus University, 8000 Aarhus, Denmark
| | - Florian Privé
- National Centre for Register-Based Research, Aarhus University, 8210 Aarhus, Denmark
| | - Hugues Aschard
- Institut Pasteur, Université de Paris, Department of Computational Biology, 75015 Paris, France; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
5
|
Pedersen CET, Hoang TT, Jin J, Starnawska A, Granell R, Elliott HR, Huels A, Zar HJ, Stein DJ, Zhang Y, Dekker HTD, Duijts L, Felix JF, Sangüesa J, Bustamante M, Casas M, Vrijheid M, Kadalayil L, Rezwan FI, Arshad H, Holloway JW, Röder S, Zenclussen AC, Herberth G, Staunstrup NH, Horsdal HT, Mill J, Hannon E, Annesi-Maesano I, Pesce G, Baïz N, Heude B, Hosseinian-Mohazzab S, Breton CV, Harlid S, Harbs J, Domellof M, West C, Yeung E, Zeng X, Nystad W, Håberg SE, Magnus MC, Schendel D, London SJ, Bønnelykke K. Maternal asthma and newborn DNA methylation. Clin Epigenetics 2025; 17:79. [PMID: 40349045 PMCID: PMC12065361 DOI: 10.1186/s13148-025-01858-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 03/11/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Prenatal exposure to maternal asthma may influence DNA methylation patterns in offspring, potentially affecting their susceptibility to later diseases including asthma. OBJECTIVE To investigate the relationship between parental asthma and newborn blood DNA methylation. METHODS Epigenome-wide association analyses were conducted in 13 cohorts on 7433 newborns with blood methylation data from the Illumina450K or EPIC array. We used fixed effects meta-analyses to identify differentially methylated CpGs (DMCs) and comb-p to identify differentially methylated regions (DMRs) associated with maternal asthma during pregnancy and maternal asthma ever. Paternal asthma was analyzed for comparison. Models were adjusted for covariates and cell-type composition. We examined whether implicated sites related to gene expression analyses in publicly available data for childhood blood and adult lung. RESULTS We identified 27 CpGs associated with maternal asthma during pregnancy at False Discovery Rate < 0.05 but none for maternal asthma ever. Two distinct CpGs were associated with paternal asthma. We observed 5 DMRs associated with maternal asthma during pregnancy 3 associated with maternal asthma ever and 13 DMRs associated with paternal asthma. Gene expression analysis using data in blood from 832 children and lung from 424 adults showed associations between identified DMCs using maternal asthma and expression of several genes, including HLA genes and HOXA5, previously implicated in asthma or lung function. CONCLUSION Parental asthma, especially maternal asthma during pregnancy, may be associated with alterations in newborn DNA methylation. These findings might shed light on underlying mechanisms for asthma susceptibility.
Collapse
Affiliation(s)
- Casper-Emil Tingskov Pedersen
- Copenhagen Prospective Studies On Asthma in Childhood, Herlev and Gentofte Hospital, COPSAC, University of Copenhagen, Ledreborg Alle 34 Gentofte, 2820, Copenhagen, Denmark
| | - Thanh T Hoang
- Division of Intramural Research, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), MD A3-05, PO 12233, Research Triangle Park, NC, 27709, USA
- Department of Pediatrics, Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Cancer and Hematology Center, Texas Children's Hospital, Houston, TX, USA
| | - Jianping Jin
- Division of Intramural Research, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), MD A3-05, PO 12233, Research Triangle Park, NC, 27709, USA
| | - Anna Starnawska
- Department of Biomedicine, Aarhus University, 8000, Aarhus, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, CGPM, and Center for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
| | - Raquel Granell
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, BS8 2BN, UK
| | - Hannah R Elliott
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, BS8 2BN, UK
| | - Anke Huels
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Ganagarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Heather J Zar
- SAMRC Unit On Child & Adolescent Health, Dept of Paediatrics, University of Cape Town, Cape Town, South Africa
| | - Dan J Stein
- SAMRC Unit On Child & Adolescent Health, Dept of Paediatrics, University of Cape Town, Cape Town, South Africa
| | - Yining Zhang
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Herman T den Dekker
- The Generation R Study Group and Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Liesbeth Duijts
- The Generation R Study Group and Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Neonatal and Pedicatric Intensive Care, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Janine F Felix
- The Generation R Study Group and Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | | | | | - Martine Vrijheid
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública, Madrid, Spain
| | - Latha Kadalayil
- School of Chemistry, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK
| | - Faisal I Rezwan
- Department of Computer Science, Aberystwyth University, University, Aberystwyth, SY23 3DB, UK
- Faculty of Medicine, Human Development and Health, University of Southampton, Southampton, UK
| | - Hasan Arshad
- David Hide Asthma and Allergy Research Centre, Isle of Wight, UK
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, Southampton General Hospital, Southampton, UK
| | - John W Holloway
- Faculty of Medicine, Human Development and Health, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, Southampton General Hospital, Southampton, UK
| | - Stefan Röder
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
| | - Ana C Zenclussen
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
| | - Gunda Herberth
- Department of Environmental Immunology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
| | - Nicklas Heine Staunstrup
- Department of Biomedicine, Aarhus University, 8000, Aarhus, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, CGPM, and Center for Integrative Sequencing, iSEQ, Aarhus University, Aarhus, Denmark
| | - Henriette Thisted Horsdal
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
| | - Jonathan Mill
- Department of Clinical & Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Eilis Hannon
- Department of Clinical & Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Isabella Annesi-Maesano
- Institute Desbrest of Epidemiology and Public Health, University of Montpellier and INSERM, Montpellier, France
| | - Giancarlo Pesce
- Institute Desbrest of Epidemiology and Public Health, University of Montpellier and INSERM, Montpellier, France
| | - Nour Baïz
- Institute Desbrest of Epidemiology and Public Health, University of Montpellier and INSERM, Montpellier, France
| | - Barbara Heude
- Inserm, INRAE, Center for Research in Epidemiology and StatisticS (CRESS), Université Paris Cité and Université Sorbonne Paris Nord, 75004, Paris, France
| | - Sahra Hosseinian-Mohazzab
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | - Carrie V Breton
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | - Sophia Harlid
- Department of Diagnostics and Intervention, Oncology, Umeå University, Umeå, Sweden
| | - Justin Harbs
- Department of Diagnostics and Intervention, Oncology, Umeå University, Umeå, Sweden
| | - Magnus Domellof
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| | - Christina West
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| | - Edwina Yeung
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, 20817, USA
| | - Xuehuo Zeng
- Glotech Inc., 1801 Research Blvd #605, Rockville, MD, 20850, USA
| | - Wenche Nystad
- Department of Chronic Diseases, Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Siri E Håberg
- Centre for Fertility and Health, Norwegian Institute of Public Health, Skøyen, P.O. Box 222, 0213, Oslo, Norway
| | - Maria C Magnus
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
- Centre for Fertility and Health, Norwegian Institute of Public Health, Skøyen, P.O. Box 222, 0213, Oslo, Norway
- MRC Integrative Epidemiology Unit at the University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
| | - Diana Schendel
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
- AJ Drexel Autism Institute, Drexel University, Philadelphia, USA
| | - Stephanie J London
- Division of Intramural Research, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), MD A3-05, PO 12233, Research Triangle Park, NC, 27709, USA.
| | - Klaus Bønnelykke
- Copenhagen Prospective Studies On Asthma in Childhood, Herlev and Gentofte Hospital, COPSAC, University of Copenhagen, Ledreborg Alle 34 Gentofte, 2820, Copenhagen, Denmark.
| |
Collapse
|
6
|
Huang M, Wen J, Lu C, Cai X, Ou C, Deng Z, Huang X, Zhang E, Chung KF, Yan J, Zhong N, Zhang Q. Residential greenness, genetic susceptibility, and asthma risk: Mediating roles of air pollution in UK and Chinese populations. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 296:118199. [PMID: 40267880 DOI: 10.1016/j.ecoenv.2025.118199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 04/12/2025] [Accepted: 04/13/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND The relationship between residential greenness and asthma remains a topic of interest, especially in understanding the pathways involved and how genetic factors might influence this association. This study aimed to explore the association between residential greenness and asthma incidence, while also examining potential mediating pathways and the role of genetic susceptibility. METHODS Data were analyzed from two independent cohorts: the UK Biobank and the Chinese Biomarkers for the Prediction of Respiratory Disease Outcomes (C-BIOPRED) study. Greenness was measured by normalized difference vegetation index (NDVI). Polygenic risk scores were constructed from 145 asthma-associated single nucleotide polymorphisms. Cox proportional hazard models and logistics regression models were used to assess the association between residential greenness and asthma incidence, and mediation analysis was conducted to explore potential mediators. RESULTS Over a median follow-up of 11.85 years in UK Biobank, higher NDVI exposure was associated with reduced asthma incidence (hazard ratio per IQR increase in NDVI300 m: 0.965, 95 % CI: 0.949-0.982). The association was more pronounced among non-smokers and individuals with highest genetic risk. PM2.5 mediated 40.4 % (95 % CI: 5.1 %-76.4 %) of the protective effect. In the C-BIOPRED study, greenness was inversely associated with severe asthma (odd ratio: 0.645, 95 % CI: 0.441-0.943) and improved clinical outcomes. CONCLUSION Residential greenness is associated with a lower risk of asthma, particularly in genetically susceptible and socioeconomically disadvantaged populations, partially through improving air quality. Our findings advocate for integrating green space optimization into urban planning as a precision public health strategy.
Collapse
Affiliation(s)
- Mingkai Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, PR China
| | - Junjie Wen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, PR China
| | - Chenyang Lu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, PR China
| | - Xuliang Cai
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, PR China
| | - Changxing Ou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, PR China
| | - Zhenan Deng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, PR China
| | - Xinyi Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, PR China
| | - Enli Zhang
- Xingyi People's Hospital, Xingyi, Guizhou 562400, PR China
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London SW3, UK.
| | - Jie Yan
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, Guangdong 510260, PR China.
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, PR China.
| | - Qingling Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, PR China; Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong 510005, PR China.
| |
Collapse
|
7
|
Starshinova AA, Savchenko AA, Borisov A, Kudryavtsev I, Rubinstein A, Dovgalyuk I, Kulpina A, Churilov LP, Sobolevskaia P, Fedotkina T, Kudlay D, Shlyakhto EV. Immunological Disorders: Gradations and the Current Approach in Laboratory Diagnostics. PATHOPHYSIOLOGY 2025; 32:17. [PMID: 40265442 PMCID: PMC12015883 DOI: 10.3390/pathophysiology32020017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 04/06/2025] [Accepted: 04/15/2025] [Indexed: 04/24/2025] Open
Abstract
Currently, understanding the immune response, its abnormalities, and its diagnostic possibilities is a key point in the management of patients with various diseases, from infectious to oncological ones. The aim of this review was to analyze the data presented in the current literature on immune disorders and the possibility of their laboratory diagnostics in combination with clinical manifestations. We have performed a systematic analysis of the literature presented in international databases over the last ten years. We have presented data on the possibility of diagnosing immunopathological processes due to changes in immune cells and soluble molecules involved in the pathogenesis of a wide range of diseases, as well as the determination of antibodies to detect autoimmune processes. By applying laboratory techniques such as hematology, flow cytometry, ELISA, etc., available to most clinical laboratories worldwide, clinical data on immune system dysfunction in a wide range of diseases are being collected. This process is unfortunately still very far from being completed. However, with all the diversity of accumulated knowledge, we can currently state that the pathogenesis of the vast majority of immune-mediated diseases is not yet known. At the same time, the current success in dividing immune-mediated diseases into distinct clusters based on different types of inflammatory responses that are based on the involvement of different populations of T helper cells and cytokine molecules represents significant progress. Further research in this direction seems very promising, as it allows the identification of new target cells and target molecules for both improved diagnostics and targeted therapies.
Collapse
Affiliation(s)
- Anna A. Starshinova
- Department of Mathematics Computer Science, St. Petersburg State University, 199034 St. Petersburg, Russia;
- Medicine Department, St. Petersburg State University, 199034 St. Petersburg, Russia; (L.P.C.); (P.S.)
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (I.K.); (A.R.); (T.F.); (E.V.S.)
| | - Andrey An. Savchenko
- Federal Research Center «Krasnoyarsk Science Center» of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660036 Krasnoyarsk, Russia; (A.A.S.); (A.B.)
| | - Alexander Borisov
- Federal Research Center «Krasnoyarsk Science Center» of the Siberian Branch of the Russian Academy of Sciences, Scientific Research Institute of Medical Problems of the North, 660036 Krasnoyarsk, Russia; (A.A.S.); (A.B.)
| | - Igor Kudryavtsev
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (I.K.); (A.R.); (T.F.); (E.V.S.)
- Department of Immunology, Institution of Experimental Medicine, 197376 St. Petersburg, Russia
| | - Artem Rubinstein
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (I.K.); (A.R.); (T.F.); (E.V.S.)
- Department of Immunology, Institution of Experimental Medicine, 197376 St. Petersburg, Russia
| | - Irina Dovgalyuk
- Research Institute of Phthisiopulmonology, 190961 St. Petersburg, Russia;
| | - Anastasia Kulpina
- Department of Mathematics Computer Science, St. Petersburg State University, 199034 St. Petersburg, Russia;
- Medicine Department, St. Petersburg State University, 199034 St. Petersburg, Russia; (L.P.C.); (P.S.)
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (I.K.); (A.R.); (T.F.); (E.V.S.)
| | - Leonid P. Churilov
- Medicine Department, St. Petersburg State University, 199034 St. Petersburg, Russia; (L.P.C.); (P.S.)
| | - Polina Sobolevskaia
- Medicine Department, St. Petersburg State University, 199034 St. Petersburg, Russia; (L.P.C.); (P.S.)
| | - Tamara Fedotkina
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (I.K.); (A.R.); (T.F.); (E.V.S.)
- Laboratory of Comparative Sensory Physiology, Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, 194223 St. Petersburg, Russia
| | - Dmitry Kudlay
- Medical Department, I.M. Sechenov First Moscow State Medical University, 197022 Moscow, Russia;
- Department of Pharmacology, I.M. Sechenov First Moscow State Medical University, 197022 Moscow, Russia
- Institute of Immunology FMBA of Russia, 115478 Moscow, Russia
- Department of Pharmacognosy and Industrial Pharmacy, Faculty of Fundamental Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Evgeny V. Shlyakhto
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (I.K.); (A.R.); (T.F.); (E.V.S.)
| |
Collapse
|
8
|
Ruan J, Yi X. Genomic structural equation modeling elucidates the shared genetic architecture of allergic disorders. J Transl Med 2025; 23:445. [PMID: 40234965 PMCID: PMC12001568 DOI: 10.1186/s12967-025-06465-8] [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: 03/06/2025] [Accepted: 04/07/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND The intricate shared genetic architecture underlying allergic disorders-including allergic asthma, atopic dermatitis, contact dermatitis, allergic rhinitis, allergic conjunctivitis, allergic urticaria, anaphylaxis, and eosinophilic esophagitis-remains incompletely characterized. METHODS Our study employed genomic structural equation modeling (Genomic SEM) to define the common factor representing the shared genetic architecture of allergic disorders. Coupled with diverse post-GWAS analytical methods, we aimed to discover susceptible loci and investigate genetic associations with external traits. Furthermore, we explored enriched genetic pathways, cellular layers, and genomic elements, and investigated putative plasma protein biomarkers. Polygenic risk score (PRS) analyses, leveraging our integrated GWAS data, were conducted to assess chromosomal-level risk associations for allergic disorders. RESULTS A well-fitted genomic SEM integrated GWAS data, revealing the shared genetic architecture of allergic disorders. We identified a total of 2038 genome-wide significant SNP loci (p < 5e-8), including 31 previously unreported loci. Fine-mapping of variants and gene sets pinpointed 2 causal variants and 31 candidate susceptible genes. Genetic correlation analyses further illuminated the shared genetic architecture underlying multiple traits, notably psychiatric disorders. Preliminary findings identified four putative causal plasma protein biomarkers. CONCLUSION Notably, this study presents the first comprehensive genetic characterization of allergic disorders through a GWAS analysis of an unmeasured composite phenotype, providing novel insights into shared etiological pathways across these conditions.
Collapse
Affiliation(s)
- Jingsheng Ruan
- Department of Thoracic, Jinshan Hospital of Fudan University, Fudan University, Shanghai, China
| | - Xinglin Yi
- Department of Respiratory and Critical Care Medicine, Third Military Medical University, Chongqing, China.
| |
Collapse
|
9
|
Oliva M, Sarkar MK, March ME, Saeidian AH, Mentch FD, Hsieh CL, Tang F, Uppala R, Patrick MT, Li Q, Bogle R, Kahlenberg JM, Watson D, Glessner JT, Youssefian L, Vahidnezhad H, Tsoi LC, Hakonarson H, Gudjonsson JE, Smith KM, Riley-Gillis B. Integration of GWAS, QTLs and keratinocyte functional assays reveals molecular mechanisms of atopic dermatitis. Nat Commun 2025; 16:3101. [PMID: 40164604 PMCID: PMC11958703 DOI: 10.1038/s41467-025-58310-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 03/18/2025] [Indexed: 04/02/2025] Open
Abstract
Atopic dermatitis is a highly heritable and common inflammatory skin condition affecting children and adults worldwide. Multi-ancestry approaches to atopic dermatitis genetic association studies are poised to boost power to detect genetic signal and identify loci contributing to atopic dermatitis risk. Here, we present a multi-ancestry GWAS meta-analysis of twelve atopic dermatitis cohorts from five ancestral populations totaling 56,146 cases and 602,280 controls. We report 101 genomic loci associated with atopic dermatitis, including 16 loci that have not been previously associated with atopic dermatitis or eczema. Fine-mapping, QTL colocalization, and cell-type enrichment analyses identified genes and cell types implicated in atopic dermatitis pathophysiology. Functional analyses in keratinocytes provide evidence for genes that could play a role in atopic dermatitis through epidermal barrier function. Our study provides insights into the etiology of atopic dermatitis by harnessing multiple genetic and functional approaches to unveil the mechanisms by which atopic dermatitis-associated variants impact genes and cell types.
Collapse
Affiliation(s)
| | | | - Michael E March
- Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | | | - Frank D Mentch
- Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | | | | | | | | | - Qinmengge Li
- University of Michigan, Ann Arbor, MI, 48109, USA
| | | | | | - Deborah Watson
- Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | | | - Leila Youssefian
- Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- City of Hope National Medical Center, Irwindale, CA, 91706, USA
| | - Hassan Vahidnezhad
- Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Lam C Tsoi
- University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hakon Hakonarson
- Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | | | | | | |
Collapse
|
10
|
Wu DC, Zhang XY, Li AD, Wang T, Wang ZY, Song SY, Chen MZ. Neuroticism and asthma: Mendelian randomization analysis reveals causal link with mood swings and BMI mediation. J Asthma 2025; 62:674-683. [PMID: 39620646 DOI: 10.1080/02770903.2024.2434516] [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: 09/18/2024] [Revised: 11/13/2024] [Accepted: 11/21/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Neuroticism has been associated with asthma, but the nature of this relationship remains unclear due to limited understanding of the impact of psychological factors on asthma risk. While Neuroticism is known to affect various health outcomes, its specific role in respiratory conditions like asthma is not fully understood. METHODS We conducted Mendelian randomization (MR) analyses using genome-wide association studies (GWAS) to explore the causal link between 12 Neuroticism traits and asthma. Various MR approaches, including MR-PRESSO, were employed, with validation through independent GWAS and the FinnGen dataset. RESULTS MR-PRESSO revealed a significant causal relationship between mood swings and asthma (OR: 1.927, 95% CI: 1.641-2.263), surpassing the Bonferroni-corrected threshold (p < 4.167 × 10-³). Mood swings emerged as the only significant trait associated with asthma, with reverse MR analyses showing no causal links for other traits. Secondary analyses supported these findings. Multivariate analysis showed mood swings increased asthma risk, independent of smoking, BMI, and air pollution. Mediation analysis indicated that BMI partially mediates the mood swing-asthma relationship, accounting for 9.87% of the effect (95% CI: 4.54%-15.2%, p = 2.850 × 10-4). CONCLUSION Mood swings elevate asthma risk, with BMI partially mediating this effect, highlighting a potentially significant pathway through which psychological traits influence asthma.
Collapse
Affiliation(s)
- Dong-Cai Wu
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xin-Yue Zhang
- Artemisia annua Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - An-Dong Li
- Department of Respiratory, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Tan Wang
- Department of Respiratory, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Zi-Yuan Wang
- Department of Respiratory, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Si-Yu Song
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Meng-Zhu Chen
- Department of Respiratory, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
11
|
Kottyan LC, Richards S, Tracy ME, Lawson LP, Cobb B, Esslinger S, Gerwe M, Morgan J, Chandel A, Travitz L, Huang Y, Black C, Sobowale A, Akintobi T, Mitchell M, Beck AF, Unaka N, Seid M, Fairbanks S, Adams M, Mersha T, Namjou B, Pauciulo MW, Strawn JR, Ammerman RT, Santel D, Pestian J, Glauser T, Prows CA, Martin LJ, Muglia L, Harley JB, Chepelev I, Kaufman KM. Sequencing and health data resource of children of African ancestry. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.22.25324419. [PMID: 40196241 PMCID: PMC11974803 DOI: 10.1101/2025.03.22.25324419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Purpose Individuals who self-report as Black or African American are historically underrepresented in genome-wide studies of disease risk, a disparity particularly evident in pediatric disease research. To address this gap, Cincinnati Children's Hospital Medical Center (CCHMC) established a biorepository and developed a comprehensive DNA sequencing resource including 15,684 individuals who self-identified as African American or Black and received care at CCHMC. Methods Participants were enrolled through the CCHMC Discover Together Biobank and sequenced. Admixture analyses confirmed the genetic ancestry of the cohort, which was then linked to electronic medical records. Results High-quality genome-wide genotypes from common variants accompanied by medical recordsourced data are available through the Genomic Information Commons. This dataset performs well in genetic studies. Specifically, we replicated known associations in sickle cell disease (HBB, p = 4.05 × 10-1), anxiety (PLAA3, p = 6.93 × 10-), and asthma (PCDH15, p = 5.6 × 10-1), while also identifying novel loci associated with asthma severity. Conclusion We present the acquisition and quality of genetic and disease-associated data and present an analytical framework for using this resource. In partnership with a community advisory council, we have co-developed a valuable framework for data use and future research.
Collapse
Affiliation(s)
- Leah C. Kottyan
- Department of Pediatrics. College of Medicine. University of Cincinnati. Cincinnati, Ohio
- Division of Allergy & Immunology. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
- Division of Human Genetics. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
- Center for Autoimmune Genomics and Etiology. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Scott Richards
- Division of Human Genetics. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
- Center for Autoimmune Genomics and Etiology. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Morgan E. Tracy
- Division of Human Genetics. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
- Discover Together Biobank. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Lucinda P. Lawson
- Division of Allergy & Immunology. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Beth Cobb
- Center for Autoimmune Genomics and Etiology. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
- Center for Stem Cell & Organoid Medicine (CuSTOM), Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Steve Esslinger
- Division of Human Genetics. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
- Discover Together Biobank. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Margaret Gerwe
- Division of Human Genetics. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
- Discover Together Biobank. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - James Morgan
- Division of Human Genetics. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
- Discover Together Biobank. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Alka Chandel
- Information Services for Research (IS4R). Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Leksi Travitz
- Department of Pediatrics. College of Medicine. University of Cincinnati. Cincinnati, Ohio
- Center for Autoimmune Genomics and Etiology. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Yongbo Huang
- Department of Pediatrics. College of Medicine. University of Cincinnati. Cincinnati, Ohio
- Center for Autoimmune Genomics and Etiology. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Catherine Black
- Division of Human Genetics. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
- Center for Autoimmune Genomics and Etiology. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Agboade Sobowale
- Division of Human Genetics. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
- Center for Autoimmune Genomics and Etiology. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
- Office of Community Relations. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Tinuke Akintobi
- Office of Community Relations. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Monica Mitchell
- Department of Pediatrics. College of Medicine. University of Cincinnati. Cincinnati, Ohio
- Office of Community Relations. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
- Division of Behavioral Medicine and Clinical Psychology. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Andrew F. Beck
- Department of Pediatrics. College of Medicine. University of Cincinnati. Cincinnati, Ohio
- Division of General & Community Pediatrics. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
- Division of Hospital Medicine. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
- Office of Population Health and Michael Fisher Child Health Equity Center. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
- Anderson Center. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Ndidi Unaka
- Department of Pediatrics. College of Medicine. University of Cincinnati. Cincinnati, Ohio
- Division of General & Community Pediatrics. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
- Department of Pediatrics, Stanford University School of Medicine. Stanford, California
| | - Michael Seid
- Department of Pediatrics. College of Medicine. University of Cincinnati. Cincinnati, Ohio
- Anderson Center. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
- Division of Pulmonary Medicine. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Sonja Fairbanks
- Division of Hospital Medicine. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Michelle Adams
- Cincinnati Children’s Research Foundation. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Tesfaye Mersha
- Department of Pediatrics. College of Medicine. University of Cincinnati. Cincinnati, Ohio
- Division of Asthma Research. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Bahram Namjou
- Department of Pediatrics. College of Medicine. University of Cincinnati. Cincinnati, Ohio
- Division of Human Genetics. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
- Center for Autoimmune Genomics and Etiology. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Michael W. Pauciulo
- Department of Pediatrics. College of Medicine. University of Cincinnati. Cincinnati, Ohio
- Division of Human Genetics. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
- Discover Together Biobank. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Jeffrey R. Strawn
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati School of Medicine. Cincinnati, Ohio
| | - Robert T. Ammerman
- Division of Behavioral Medicine and Clinical Psychology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati. Cincinnati, Ohio
| | - Daniel Santel
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center
| | - John Pestian
- Department of Pediatrics. College of Medicine. University of Cincinnati. Cincinnati, Ohio
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center
- Computational Medicine Center, Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Tracy Glauser
- Department of Pediatrics. College of Medicine. University of Cincinnati. Cincinnati, Ohio
- Division of Neurology. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Cynthia A. Prows
- Division of Human Genetics. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Lisa J. Martin
- Department of Pediatrics. College of Medicine. University of Cincinnati. Cincinnati, Ohio
- Division of Human Genetics. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - Louis Muglia
- Department of Pediatrics. College of Medicine. University of Cincinnati. Cincinnati, Ohio
- Division of Human Genetics. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
| | - John B. Harley
- US Department of Veterans Affairs Medical Center, Cincinnati, Ohio. Cincinnati, Ohio
| | - Iouri Chepelev
- US Department of Veterans Affairs Medical Center, Cincinnati, Ohio. Cincinnati, Ohio
- Research Service, US Department of Veterans Affairs Medical Center, Cincinnati, Ohio
| | - Kenneth M. Kaufman
- Department of Pediatrics. College of Medicine. University of Cincinnati. Cincinnati, Ohio
- Division of Human Genetics. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
- Center for Autoimmune Genomics and Etiology. Cincinnati Children’s Hospital Medical Center. Cincinnati, Ohio
- US Department of Veterans Affairs Medical Center, Cincinnati, Ohio. Cincinnati, Ohio
| |
Collapse
|
12
|
Zakarya R, Chan YL, Wang B, Thorpe A, Xenaki D, Ho KF, Guo H, Chen H, Oliver BG, O'Neill C. Developmental air pollution exposure augments airway hyperreactivity, alters transcriptome, and DNA methylation in female adult progeny. Commun Biol 2025; 8:400. [PMID: 40057553 PMCID: PMC11890619 DOI: 10.1038/s42003-025-07835-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 02/26/2025] [Indexed: 05/13/2025] Open
Abstract
Maternal exposure to particulate air pollution increases the incidence and severity of asthma in offspring, yet the mechanisms for this are unclear. Known susceptibility loci are a minor component of this effect. We interrogate a mouse allergic airway disease model to assess epigenetic associations between maternal air pollution exposure and asthma responses in offspring. Maternal air pollution exposure increased allergic airway disease severity in adult offspring associated with a suppressed transcriptomic response. Control progeny showed differential expression of 2842 genes across several important pathways, whilst air pollutant progeny showed an 80% reduction in differentially expressed genes and abrogation of many pathway associations. Whole genome CpG methylome analysis following allergen challenge detected differential methylation regions across the genome. Differentially methylated regions were markedly reduced in air pollutant offspring, and this was most evident in intronic regions and some transposable element classes. This study shows that asthma in adult offspring of PM2.5 exposed mothers had a markedly repressed transcriptomic response, a proportion of which was associated with identifiable changes in the lung's methylome. The results point to an epigenetic contribution to the severity of asthma in offspring of mothers exposed to particulate air pollution.
Collapse
Affiliation(s)
- Razia Zakarya
- School of Life Sciences, University of Technology Sydney, Sydney, Australia.
- Epigenetics of Chronic Disease Group, Woolcock Institute of Medical Research, Macquarie University, Sydney, Australia.
| | - Yik Lung Chan
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
- Respiratory Cell and Molecular Biology Group, Woolcock Institute of Medical Research, Macquarie University, Sydney, Australia
| | - Baoming Wang
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
- Respiratory Cell and Molecular Biology Group, Woolcock Institute of Medical Research, Macquarie University, Sydney, Australia
| | - Andrew Thorpe
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
- Respiratory Cell and Molecular Biology Group, Woolcock Institute of Medical Research, Macquarie University, Sydney, Australia
| | - Dikaia Xenaki
- Respiratory Cell and Molecular Biology Group, Woolcock Institute of Medical Research, Macquarie University, Sydney, Australia
| | - Kin Fai Ho
- Jockey Club School of Public Health and Primary, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of the People's Republic of China, Hong Kong, China
| | - Hai Guo
- Air Quality Studies, Department of Civil and Environmental Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Hui Chen
- School of Life Sciences, University of Technology Sydney, Sydney, Australia
| | - Brian G Oliver
- School of Life Sciences, University of Technology Sydney, Sydney, Australia.
- Respiratory Cell and Molecular Biology Group, Woolcock Institute of Medical Research, Macquarie University, Sydney, Australia.
| | - Christopher O'Neill
- School of Life Sciences, University of Technology Sydney, Sydney, Australia.
- Epigenetics of Chronic Disease Group, Woolcock Institute of Medical Research, Macquarie University, Sydney, Australia.
| |
Collapse
|
13
|
Su H, Xie H. Associations between lifestyle habits, environmental factors and respiratory diseases: a cross-sectional study from southwest China. Front Public Health 2025; 13:1513926. [PMID: 40109413 PMCID: PMC11919831 DOI: 10.3389/fpubh.2025.1513926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 02/07/2025] [Indexed: 03/22/2025] Open
Abstract
Background Numerous studies have demonstrated that lifestyle habits and environmental factors influence the incidence and progression of respiratory diseases. However, there is a paucity of similar research conducted in southwest China. Objective This study aims to investigate the prevalence and primary influencing factors of respiratory diseases among residents in a specific region of southwest China, and to identify vulnerable populations. Method From February 2024 to May 2024, a multi-stage stratified random sampling method was employed in a specific region of southwest China. Three monitoring points were randomly selected from six jurisdictions within this region, resulting in the collection of relevant information from a total of 4,507 residents through offline interviews. Lasso-logistic regression was conducted using R version 4.3.0 to develop a nomogram for estimating disease probabilities. Interaction analysis was performed with gender and age group serving as grouping variables, while other dimensional factors were utilized as analysis variables. Result A total of 4,507 respondents participated in this study, of whom 956 (21.21%) were identified as sick. The older adult group (>65 years) exhibited the highest prevalence (30.3%). Results from the Lasso-logistic model indicated that current smoking, alcohol abuse, passive smoking, coupled with poor indoor and outdoor environments were significant risk factors. Additionally, a history of respiratory disease, a family history of respiratory issues, negative emotions, and high stress levels may also contribute to the risk of the disease. Protective factors identified include regular exercise, adequate indoor lighting, frequent ventilation, and regular disinfection practices. The nomogram developed in this study demonstrated good discrimination, calibration, and clinical efficacy. Multiplicative interaction analysis indicated that gender and age group exhibited varying degrees of interaction with factors such as smoking, passive smoking, alcohol abuse, regular exercise, household smoke, house disinfection, dust mites, history of respiratory allergies, use of velvet products, and family history of respiratory conditions. Notably, females, adolescents, and the older adult were identified as particularly susceptible and at-risk groups for these interactions. Conclusion The prevalence of respiratory diseases is notably higher among the permanent population in southwest China. High-risk lifestyles, coupled with poor indoor and outdoor environments, pose particularly significant threats to women, adolescents, and the older adult. Consequently, improving living habits, renovating aging communities, enhancing the quality of the living environment, and prioritizing vulnerable populations remain central to the objectives of primary health services.
Collapse
Affiliation(s)
| | - Huifang Xie
- School of Public Health, Xinjiang Medical University, Ürümqi, China
| |
Collapse
|
14
|
Peng B, Ye W, Liu S, Jiang Y, Meng Z, Guo M, Zhi L, Chang X, Shao L. Sex differences in asthma: omics evidence and future directions. Front Genet 2025; 16:1560276. [PMID: 40110046 PMCID: PMC11920188 DOI: 10.3389/fgene.2025.1560276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 02/11/2025] [Indexed: 03/22/2025] Open
Abstract
Asthma is a common and complex heterogeneous disease, with prevalence and severity varying across different age groups and sexes. Over the past few decades, with the development of high-throughput technologies, various "omics" analyses have emerged and been applied to asthma research, providing us with significant opportunities to study the genetic mechanisms underlying asthma. However, despite these advancements, the differences and specificities in the genetic mechanisms of asthma between sexes remain to be fully explored. Moreover, clinical guidelines have yet to incorporate or recommend sex-specific asthma management based on high-quality omics evidence. In this article, we review recent omics-level findings on sex differ-ences in asthma and discuss how to better integrate these multidimensional findings to generate further insights and advance the precision and effectiveness of asthma treatment.
Collapse
Affiliation(s)
- Bichen Peng
- College of Medical Information and Artificial Intelligence, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Weiyi Ye
- College of Medical Information and Artificial Intelligence, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Shuai Liu
- Agricultural Products Quality and Safety Center of Ji'nan, Jinan, Shandong, China
| | - Yue Jiang
- College of Medical Information and Artificial Intelligence, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ziang Meng
- College of Medical Information and Artificial Intelligence, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Miao Guo
- School of Life Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Lili Zhi
- Department of Allergy, Shandong Institute of Respiratory Diseases, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Xiao Chang
- College of Medical Information and Artificial Intelligence, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Lei Shao
- Department of infectious Disease, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
15
|
Jin Z, Sun W, Huang J, Zhou M, Zhang C, Zhao B, Wang G. Association between triglyceride glucose index and asthma exacerbation: A population-based study. Heart Lung 2025; 70:1-7. [PMID: 39531988 DOI: 10.1016/j.hrtlng.2024.11.005] [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: 09/13/2024] [Revised: 10/20/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Metabolic dysfunction is associated with respiratory diseases, and the triglyceride-glucose (TyG) index is an important indicator of metabolic dysfunction. OBJECTIVES The purpose of this study was to explore the possible relationship between TyG and asthma exacerbation, while also investigating potential subgroup differences in this relationship. METHODS Data from the 2009-2018 National Health and Nutrition Examination Survey (NHANES) were included. Multifactorial logistic regression, subgroup analysis, smoothed curve fitting, and threshold effect analysis models were used to explore the relationship between TyG and asthma exacerbations. RESULTS A total of 964 participants were included in the analysis (34.13 % male, 65.87 % female, 45.4 % Non-Hispanic White, 25.3 % Non-Hispanic Black), with a mean age of 50.57 ± 17.32 years. We found a nonlinear positive relationship between TyG and asthma exacerbation, which was maintained in all three models. In the fully adjusted model, the risk of asthma exacerbation increased by 25 % with each unit increase in the patient's TyG level (OR:1.25, 95 %CI: 1.21-1.30). Subgroup analysis showed significant associations between TyG and asthma exacerbations among females, as well as in individuals aged 20-59, body mass index (BMI) <25 or BMI≥30. Furthermore, a U-shaped relationship between TyG and asthma exacerbation was identified in males using smoothed curve fitting, with an inflection point at the TyG level of 9.15. CONCLUSIONS We found a nonlinear positive association between TyG and asthma exacerbation. Our study highlights the potential clinical value of TyG in managing asthma exacerbations, particularly emphasizing the need for gender-specific risk management strategies.
Collapse
Affiliation(s)
- Zhou Jin
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China
| | - Wen Sun
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China
| | - Junjun Huang
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China
| | - Mengyun Zhou
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China
| | - Chunbo Zhang
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China
| | - Bangchao Zhao
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China
| | - Guangfa Wang
- Department of Respiratory and Critical Care Medicine, Peking University First Hospital, Beijing, China.
| |
Collapse
|
16
|
Wang H, Concannon P, Ge Y. Roles of TULA-family proteins in T cells and autoimmune diseases. Genes Immun 2025; 26:54-62. [PMID: 39558087 DOI: 10.1038/s41435-024-00300-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/28/2024] [Accepted: 10/01/2024] [Indexed: 11/20/2024]
Abstract
The T cell Ubiquitin Ligand (TULA) protein family contains two members, UBASH3A and UBASH3B, that display similarities in protein sequence and domain structure. Both TULA proteins act to repress T cell activation via a combination of overlapping and nonredundant functions. UBASH3B acts mainly as a phosphatase that suppresses proximal T cell receptor (TCR) signaling. In contrast, UBASH3A acts primarily as an adaptor protein, interacting with other proteins (including UBASH3B) in T cells upon TCR stimulation and resulting in downregulation of TCR signaling and NF-κB signaling. Human genetic and functional studies have revealed another notable distinction between UBASH3A and UBASH3B: numerous genome-wide association studies have identified statistically significant associations between genetic variants in and around the UBASH3A gene and at least seven different autoimmune diseases, suggesting a key role of UBASH3A in autoimmunity. However, the evidence for an independent role of UBASH3B in autoimmune disease is limited. This review summarizes key findings regarding the roles of TULA proteins in T cell biology and autoimmunity, highlights the commonalities and differences between UBASH3A and UBASH3B, and speculates on the individual and joint effects of TULA proteins on T cell signaling.
Collapse
Affiliation(s)
- Hua Wang
- International Center for Genetic Engineering and Biotechnology, China Regional Research Center, Taizhou, Jiangsu Province, China
| | - Patrick Concannon
- Genetics Institute, University of Florida, Gainesville, FL, USA
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Yan Ge
- International Center for Genetic Engineering and Biotechnology, China Regional Research Center, Taizhou, Jiangsu Province, China.
| |
Collapse
|
17
|
Plichta J, Panek M. Role of the TGF-β cytokine and its gene polymorphisms in asthma etiopathogenesis. FRONTIERS IN ALLERGY 2025; 6:1529071. [PMID: 39949968 PMCID: PMC11821632 DOI: 10.3389/falgy.2025.1529071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Transforming growth factor beta (TGF-β) is a pluripotent cytokine expressed by all cells of the human body which plays important roles in maintaining homeostasis and allowing for proper individual development. Disturbances in TGF-β signaling contribute to the development of many diseases and disorders, including cancer and organ fibrosis. One of the diseases with the best-characterized correlation between TGF-β action and etiopathogenesis is asthma. Asthma is the most common chronic inflammatory disease of the lower and upper respiratory tract, characterized by bronchial hyperresponsiveness to a number of environmental factors, leading to bronchospasm and reversible limitation of expiratory flow. TGF-β, in particular TGF-β1, is a key factor in the etiopathogenesis of asthma. TGF-β1 concentration in bronchoalveolar lavage fluid samples is elevated in atopic asthma, and TGF-β expression is increased in asthmatic bronchial samples. The expression of all TGF-β isoforms is affected by a number of single nucleotide polymorphisms found in the genes encoding these cytokines. Some of the SNPs that alter the level of TGF-β expression may be associated with the occurrence and severity of symptoms of asthma and other diseases. The TGF-β gene polymorphisms, which are the subject of this paper, are potential diagnostic factors. If properly used, these polymorphisms can facilitate the early and precise diagnosis of asthma, allowing for the introduction of appropriate therapy and reduction of asthma exacerbation frequency.
Collapse
Affiliation(s)
- Jacek Plichta
- Department of Internal Medicine, Asthma and Allergology, Medical University of Lodz, Lodz, Poland
| | | |
Collapse
|
18
|
Chen F, Yang Y, Yu L, Song L, Zhang C, He Y, Wu L, Ma W, Zhang B. The association between type 2 diabetes and asthma incidence: a longitudinal analysis considering genetic susceptibility. BMC Public Health 2025; 25:166. [PMID: 39815260 PMCID: PMC11734334 DOI: 10.1186/s12889-024-21266-2] [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: 05/14/2024] [Accepted: 12/31/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND The prevalence of type 2 diabetes (T2D) and asthma is rising, yet evidence regarding the relationship between T2D and asthma, particularly in the context of genetic predispositions, remains limited. METHODS This study utilized data from the UK Biobank longitudinal cohort, involving 388,775 participants. A polygenic risk score (PRS) for asthma was derived from genome-wide association studies summary. Cox regression models were used to assess the association between T2D and asthma, incorporating the asthma PRS. RESULTS Over a median follow-up of 13.62 years, 10,211 asthma cases were documented. After adjusting for age, sex, current smoking status, and other confounding variables, T2D was significantly associated with an increased risk of developing asthma (Hazard Ratios [HR] 1.16, 95% confidence interval [CI] 1.06-1.26). This association remained significant after further adjustments for genetic susceptibility to asthma. Furthermore, T2D increased the risk of developing asthma across both high and low genetic risk groups. CONCLUSIONS T2D is associated with an increased risk of developing asthma, irrespective of genetic susceptibility. These findings underscore the importance of incorporating glucose regulation strategies into asthma prevention efforts.
Collapse
Affiliation(s)
- Fei Chen
- Department of Endocrinology, China-Japan Friendship Hospital, No.2 Yinghuayuan East Street, Hepingli, Chaoyang District, 100029, Beijing, China
| | - Ying Yang
- Department of Endocrinology, China-Japan Friendship Hospital, No.2 Yinghuayuan East Street, Hepingli, Chaoyang District, 100029, Beijing, China
| | - Liping Yu
- Department of Endocrinology, China-Japan Friendship Hospital, No.2 Yinghuayuan East Street, Hepingli, Chaoyang District, 100029, Beijing, China
| | - Lulu Song
- Department of Endocrinology, China-Japan Friendship Hospital, No.2 Yinghuayuan East Street, Hepingli, Chaoyang District, 100029, Beijing, China
| | - Cong Zhang
- Department of Endocrinology, China-Japan Friendship Hospital, No.2 Yinghuayuan East Street, Hepingli, Chaoyang District, 100029, Beijing, China
| | - Yifan He
- Department of Endocrinology, China-Japan Friendship Hospital, No.2 Yinghuayuan East Street, Hepingli, Chaoyang District, 100029, Beijing, China
| | - Lili Wu
- Department of Endocrinology, China-Japan Friendship Hospital, No.2 Yinghuayuan East Street, Hepingli, Chaoyang District, 100029, Beijing, China
| | - Wanlu Ma
- Department of Endocrinology, China-Japan Friendship Hospital, No.2 Yinghuayuan East Street, Hepingli, Chaoyang District, 100029, Beijing, China
| | - Bo Zhang
- Department of Endocrinology, China-Japan Friendship Hospital, No.2 Yinghuayuan East Street, Hepingli, Chaoyang District, 100029, Beijing, China.
| |
Collapse
|
19
|
Pariès M, Bougeard S, Eslami A, Li Z, Laviolette M, Boulet LP, Vigneau E, Bossé Y. The clinical value and most informative threshold of polygenic risk score in the Quebec City Case-Control Asthma Cohort. BMC Pulm Med 2025; 25:21. [PMID: 39815278 PMCID: PMC11734400 DOI: 10.1186/s12890-025-03486-3] [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: 10/20/2023] [Accepted: 01/07/2025] [Indexed: 01/18/2025] Open
Abstract
Genome-wide association studies (GWAS) have identified genetic variants robustly associated with asthma. A potential near-term clinical application is to calculate polygenic risk score (PRS) to improve disease risk prediction. The value of PRS, as part of numerous multi-source variables used to define asthma, remains unclear. This study aims to evaluate PRS and define most informative thresholds in relation to conventional clinical and physiological criteria of asthma using a multivariate statistical method. Clinical and genome-wide genotyping data were obtained from the Quebec City Case-Control Asthma Cohort (QCCCAC), which is an independent cohort from previous GWAS. PRS was derived using LDpred2 and integrated with other asthma phenotypes by means of Principal Component Analysis with Optimal Scaling (PCAOS). PRS was considered using 'ordinal level of scaling' to account for non-linear information. In two dimensional PCAOS space, the first component delineated individuals with and without asthma, whereas the severity of asthma was discerned on the second component. The positioning of high vs. low PRS in this space matched the presence and absence of airway hyperresponsiveness, showing that PRS delineated cases and controls at the same extent as a positive bronchial challenge test. The top 10% and the bottom 5% of the PRS were the most informative thresholds to define individuals at high and low genetic risk of asthma in this cohort. PRS used in a multivariate method offers a decision-making space similar to hyperresponsiveness in this cohort and highlights the most informative and asymmetrical thresholds to define high and low genetic risk of asthma.
Collapse
Affiliation(s)
- Martin Pariès
- Oniris, INRAE, StatSC, Nantes, 44300, France
- Anses (French Agency for Food, Environmental and Occupational Health and Safety), Ploufragan, 22440, France
| | - Stéphanie Bougeard
- Anses (French Agency for Food, Environmental and Occupational Health and Safety), Ploufragan, 22440, France
| | - Aida Eslami
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada
- Department of Social and Preventive Medicine, Université Laval, Quebec City, Canada
| | - Zhonglin Li
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada
| | - Michel Laviolette
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada
| | - Louis-Philippe Boulet
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada
| | | | - Yohan Bossé
- Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval, Quebec City, Canada.
- Department of Molecular Medicine, Université Laval, Quebec City, Canada.
| |
Collapse
|
20
|
Zhou S, Xiao H, Gao M, Wang M, He W, Shu Y, Wang X. Causal role of immune cells in asthma: a Mendelian randomization study. J Asthma 2025; 62:84-90. [PMID: 39087928 DOI: 10.1080/02770903.2024.2387758] [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: 04/05/2024] [Revised: 07/16/2024] [Accepted: 07/30/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Immune cells may have a significant role in the pathophysiology of asthma, according to increasing evidence, although it is yet unclear how immune cells cause asthma. Therefore, we aimed to use Mendelian randomization (MR) methods to investigate this causal relationship. METHODS This study explored the causal effects between immune cells and asthma using a two-sample MR technique. Using publicly available genetic data, the causal connection between asthma risk and 731 immune cell phenotypes was investigated. Sensitivity analysis guaranteed the results' stability. To further evaluate the existence of reverse causality, we employed reverse MR analysis. RESULTS According to the inverse-variance weighted (IVW) method, five immune cell phenotypes were found to be statistically significantly associated with asthma risk (p < 0.001). Among them, TCRgd %T cell (OR = 0.968, 95%CI = 0.951 - 0.986), TCRgd %lymphocyte (OR = 0.978, 95%CI = 0.965 - 0.991), HLA DR + NK AC (OR = 0.966, 95% CI = 0.947 - 0.986) and CD3 on CD4 Treg (OR = 0.956, 95%CI= 0.931 - 0.981), four phenotypes that resulted in a decreased risk of asthma. CD25 on transitional (OR = 1.033, 95%CI = 1.014 - 1.052) resulted in an increased risk of asthma. Reverse MR analysis revealed that asthma increases HLA DR + NK AC levels (p < 0.05). CONCLUSION The results of MR analysis showed a causal relationship between immune cell phenotype and asthma risk, which provides a direction for future asthma treatment.
Collapse
Affiliation(s)
- Siding Zhou
- Department of Medical College of Yangzhou University, Yangzhou, China
| | - Hongbi Xiao
- Department of Medical College of Yangzhou University, Yangzhou, China
| | - Mingjun Gao
- Department of Dalian Medical University, Dalian, China
| | - Mengmeng Wang
- Department of Dalian Medical University, Dalian, China
| | - Wenbo He
- Department of Medical College of Yangzhou University, Yangzhou, China
| | - Yusheng Shu
- Department of Thoracic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Xiaolin Wang
- Department of Thoracic Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| |
Collapse
|
21
|
Nishida Y, Terkawi MA, Matsumae G, Yokota S, Tokuhiro T, Ogawa Y, Ishizu H, Shiota J, Endo T, Alhasan H, Ebata T, Kitahara K, Shimizu T, Takahashi D, Takahata M, Kadoya K, Iwasaki N. Dynamic transcriptome analysis of osteal macrophages identifies a distinct subset with senescence features in experimental osteoporosis. JCI Insight 2024; 9:e182418. [PMID: 39480497 PMCID: PMC11623942 DOI: 10.1172/jci.insight.182418] [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: 04/25/2024] [Accepted: 10/25/2024] [Indexed: 11/02/2024] Open
Abstract
Given the potential fundamental function of osteal macrophages in bone pathophysiology, we study here their precise function in experimental osteoporosis. Gene profiling of osteal macrophages from ovariectomized mice demonstrated the upregulation of genes that were involved in oxidative stress, cell senescence, and apoptotic process. A single-cell RNA-Seq analysis revealed that osteal macrophages were heterogeneously clustered into 6 subsets that expressed proliferative, inflammatory, antiinflammatory, and efferocytosis gene signatures. Importantly, postmenopausal mice exhibited an increase in subset 3 that showed a typical gene signature of cell senescence and inflammation. These findings suggest that the decreased production of estrogen due to postmenopausal condition altered the osteal macrophage subsets, resulting in a shift toward cell senescence and inflammatory conditions in the bone microenvironment. Furthermore, adoptive macrophage transfer onto calvarial bone was performed, and mice that received oxidatively stressed macrophages exhibited greater osteolytic lesions than control macrophages, suggesting the role of these cells in the development of inflammaging in the bone microenvironment. Consistently, depletion of senescent cells and the oxidatively stressed macrophage subset alleviated the excessive bone loss in postmenopausal mice. Our data provided insight into the pathogenesis of osteoporosis and shed light on a therapeutic approach for the treatment or prevention of postmenopausal osteoporosis.
Collapse
|
22
|
Ochoa S, Rasquel-Oliveira FS, McKinnon B, Haro M, Subramaniam S, Yu P, Coetzee S, Anglesio MS, Wright KN, Meyer R, Gargett CE, Mortlock S, Montgomery GW, Rogers MS, Lawrenson K. M2 Macrophages are Major Mediators of Germline Risk of Endometriosis and Explain Pleiotropy with Comorbid Traits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.21.624726. [PMID: 39605445 PMCID: PMC11601670 DOI: 10.1101/2024.11.21.624726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Endometriosis is a common gynecologic condition that causes chronic life-altering symptoms including pain, infertility, and elevated cancer risk. There is an urgent need for new non-hormonal targeted therapeutics to treat endometriosis, but until very recently, the cellular and molecular signatures of endometriotic lesions were undefined, severely hindering the development of clinical advances. Integrating inherited risk data from analyses of >450,000 individuals with ∼350,000 single cell transcriptomes from 21 patients, we uncover M2-macrophages as candidate drivers of disease susceptibility, and nominate IL1 signaling as a central hub impacted by germline genetic variation associated with endometriosis. Extensive functional follow-up confirmed these associations and revealed a pleiotropic role for this pathway in endometriosis. Population-scale expression quantitative trail locus analysis demonstrated that genetic variation controlling IL1A expression is also associated with endometriosis risk variants. Manipulation of IL1 signaling in state-of-the-art in vitro decidualized assembloids impacted epithelial differentiation, and in an in vivo endometriosis model, treatment with anakinra (an interleukin-1 receptor antagonist) resulted in a significant, dose-dependent reduction in both spontaneous pain and evoked pain. Together these studies highlight non-diagnostic cell types as central to endometriosis susceptibility and support IL1 signaling as an important actionable pathway for this disease.
Collapse
|
23
|
Veerasubramanian PK, Wynn TA, Quan J, Karlsson FJ. Targeting TNF/TNFR superfamilies in immune-mediated inflammatory diseases. J Exp Med 2024; 221:e20240806. [PMID: 39297883 PMCID: PMC11413425 DOI: 10.1084/jem.20240806] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/19/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Abstract
Dysregulated signaling from TNF and TNFR proteins is implicated in several immune-mediated inflammatory diseases (IMIDs). This review centers around seven IMIDs (rheumatoid arthritis, systemic lupus erythematosus, Crohn's disease, ulcerative colitis, psoriasis, atopic dermatitis, and asthma) with substantial unmet medical needs and sheds light on the signaling mechanisms, disease relevance, and evolving drug development activities for five TNF/TNFR signaling axes that garner substantial drug development interest in these focus conditions. The review also explores the current landscape of therapeutics, emphasizing the limitations of the approved biologics, and the opportunities presented by small-molecule inhibitors and combination antagonists of TNF/TNFR signaling.
Collapse
Affiliation(s)
| | - Thomas A. Wynn
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA, USA
| | - Jie Quan
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA, USA
| | | |
Collapse
|
24
|
Van Asselt AJ, Beck JJ, Finnicum CT, Johnson BN, Kallsen N, Viet S, Huizenga P, Ligthart L, Hottenga JJ, Pool R, der Zee AHMV, Vijverberg SJ, de Geus E, Boomsma DI, Ehli EA, van Dongen J. Epigenetic signatures of asthma: a comprehensive study of DNA methylation and clinical markers. Clin Epigenetics 2024; 16:151. [PMID: 39488688 PMCID: PMC11531182 DOI: 10.1186/s13148-024-01765-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/18/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Asthma, a complex respiratory disease, presents with inflammatory symptoms in the lungs, blood, and other tissues. We investigated the relationship between DNA methylation and 35 clinical markers of asthma. METHODS The Illumina Infinium EPIC v1 methylation array was used to evaluate 742,442 CpGs in whole blood from 319 participants from 94 families. They were part of the Netherlands Twin Register from families with at least one member suffering from severe asthma. Repeat blood samples were taken after 10 years from 182 individuals. Principal component analysis on the clinical asthma markers yielded ten principal components (PCs) that explained 92.8% of the total variance. We performed epigenome-wide association studies (EWAS) for each of the ten PCs correcting for familial structure and other covariates. RESULTS 221 unique CpGs reached genome-wide significance at timepoint 1 after Bonferroni correction. PC7, which correlated with loadings of eosinophil counts and immunoglobulin levels, accounted for the majority of associations (204). Enrichment analysis via the EWAS Atlas identified 190 of these CpGs to be previously identified in EWASs of asthma and asthma-related traits. Proximity assessment to previously identified SNPs associated with asthma identified 17 unique SNPs within 1 MB of two of the 221 CpGs. EWAS in 182 individuals with epigenetic data at a second timepoint identified 49 significant CpGs. EWAS Atlas enrichment analysis indicated that 4 of the 49 were previously associated with asthma or asthma-related traits. Comparing the estimates of all the significant associations identified across the two time points yielded a correlation of 0.81. CONCLUSION We identified 270 unique CpGs that were associated with PC scores generated from 35 clinical markers of asthma, either cross-sectionally or 10 years later. A strong correlation was present between effect sizes at the 2 timepoints. Most associations were identified for PC7, which captured blood eosinophil counts and immunoglobulin levels and many of these CpGs have previous associations in earlier studies of asthma and asthma-related traits. The results point to a robust DNA methylation profile as a new, stable biomarker for asthma.
Collapse
Affiliation(s)
- Austin J Van Asselt
- Avera McKennan Hospital and University Health Center, Sioux Falls, SD, USA.
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| | - Jeffrey J Beck
- Avera McKennan Hospital and University Health Center, Sioux Falls, SD, USA
| | - Casey T Finnicum
- Avera McKennan Hospital and University Health Center, Sioux Falls, SD, USA
| | - Brandon N Johnson
- Avera McKennan Hospital and University Health Center, Sioux Falls, SD, USA
| | - Noah Kallsen
- Avera McKennan Hospital and University Health Center, Sioux Falls, SD, USA
| | - Sarah Viet
- Avera McKennan Hospital and University Health Center, Sioux Falls, SD, USA
| | - Patricia Huizenga
- Avera McKennan Hospital and University Health Center, Sioux Falls, SD, USA
| | - Lannie Ligthart
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Jouke-Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - René Pool
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Anke H Maitland-van der Zee
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Department Pulmonary Medicine, Amsterdam University Medical Center, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - S J Vijverberg
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Department Pulmonary Medicine, Amsterdam University Medical Center, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Eco de Geus
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development (AR&D) Research Institute, Amsterdam, The Netherlands
- Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Erik A Ehli
- Avera McKennan Hospital and University Health Center, Sioux Falls, SD, USA
| | - Jenny van Dongen
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development (AR&D) Research Institute, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Ramessur R, Saklatvala J, Budu-Aggrey A, Ostaszewski M, Möbus L, Greco D, Ndlovu M, Mahil SK, Barker JN, Brown S, Paternoster L, Dand N, Simpson MA, Smith CH. Exploring the Link Between Genetic Predictors of Cardiovascular Disease and Psoriasis. JAMA Cardiol 2024; 9:1009-1017. [PMID: 39292496 PMCID: PMC11411451 DOI: 10.1001/jamacardio.2024.2859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 06/13/2024] [Indexed: 09/19/2024]
Abstract
Importance The epidemiological link between immune-mediated diseases (IMIDs) and cardiovascular disease has often been attributed to systemic inflammation. However, the direction of causality and the biological mechanisms linking cardiovascular disease with IMIDs are incompletely understood. Given the robust epidemiological association and the growing body of supportive mechanistic evidence, psoriasis is an exemplary IMID model for exploring this relationship. Objective To assess the bidirectional relationships between genetic predictors of psoriasis and the 2 major forms of cardiovascular disease, coronary artery disease (CAD) and stroke, and to evaluate the association between genetic predictors of cardiovascular disease with 9 other IMIDs. Design, Setting, and Participants This was a genetic association study using mendelian randomization (MR), a powerful genetic tool to help distinguish causation from associations observed in epidemiological studies, to provide supportive evidence for causality between traits. The study conducted 2-sample MR analyses using summary-level data from large-scale genome-wide association meta-analysis studies (GWAS) for each trait. The analysis focused on individuals of European descent from GWAS meta-analyses, involving CAD, stroke, psoriasis, and 9 other IMIDs. Data were analyzed from January 2023 to May 2024. Exposures Genetic predictors of CAD, stroke, psoriasis, and 9 other IMIDs. Main Outcomes and Measures The primary outcomes were the associations of genetic predictors of CAD and stroke with the risk of psoriasis and 9 other IMIDs, determined using inverse-variance weighted (IVW) MR estimates. Results This study included 181 249 cases and 1 165 690 controls with CAD, 110 182 cases and 1 503 898 controls with stroke, 36 466 cases and 458 078 controls with psoriasis, for a total of approximately 3 400 000 individuals, and 9 other IMIDs. In contrast to previous assumptions, genetic predictors of psoriasis were found to have no association with CAD or stroke. In the reverse direction, genetic predictors of both CAD (MR estimate IVW odds ratio [OR], 1.07; 95% CI, 1.04-1.10; P = .003) and stroke (IVW OR, 1.22; 95% CI, 1.05-1.41; P = .01) were found to have risk-increasing associations with psoriasis. Adjusting for stroke rendered the associations of genetically predicted CAD with psoriasis risk nonsignificant (and vice versa), suggesting that a shared effect underlying genetic risk for CAD and stroke associates with increased psoriasis risk. No risk-increasing associations were observed for genetic predictors of cardiovascular disease with other common IMIDs, including rheumatoid arthritis and inflammatory bowel disease. Conclusions and Relevance Findings of this mendelian randomization study indicate that genetic predictors of cardiovascular disease were associated with increased psoriasis risk with no reciprocal effect or association with other IMIDs. Elucidating mechanisms underpinning this association could lead to novel therapeutic approaches in both diseases.
Collapse
Affiliation(s)
- Ravi Ramessur
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Jake Saklatvala
- Department of Medical and Molecular Genetics, School of Basic & Medical Biosciences, King’s College London, London, United Kingdom
| | - Ashley Budu-Aggrey
- MRC Integrative Epidemiology Unit at University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, Bristol, United Kingdom
| | - Marek Ostaszewski
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Lena Möbus
- Finnish Hub for Development and Validation of Integrated Approaches, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Dario Greco
- Finnish Hub for Development and Validation of Integrated Approaches, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Uusimaa, Finland
| | - Matladi Ndlovu
- Department of Immunology Research, UCB, Brussels, Belgium
| | - Satveer K. Mahil
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Jonathan N. Barker
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Sara Brown
- Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Department of Dermatology, NHS Lothian, Edinburgh, Scotland, United Kingdom
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit at University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, Bristol, United Kingdom
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, Bristol, United Kingdom
| | - Nick Dand
- Department of Medical and Molecular Genetics, School of Basic & Medical Biosciences, King’s College London, London, United Kingdom
| | - Michael A. Simpson
- Department of Medical and Molecular Genetics, School of Basic & Medical Biosciences, King’s College London, London, United Kingdom
| | - Catherine H. Smith
- St John’s Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
26
|
Melgaard ME, Jensen SK, Eliasen A, Pedersen CET, Thorsen J, Mikkelsen M, Vahman N, Schoos AMM, Gern J, Brix S, Stokholm J, Chawes BL, Bønnelykke K. Asthma development is associated with low mucosal IL-10 during viral infections in early life. Allergy 2024; 79:2981-2992. [PMID: 39221476 DOI: 10.1111/all.16276] [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: 03/12/2024] [Revised: 06/21/2024] [Accepted: 07/03/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Viral infection is a common trigger of severe respiratory illnesses in early life and a risk factor for later asthma development. The mechanism leading to asthma could involve an aberrant airway immune response to viral infections, but this has rarely been studied in a human setting. OBJECTIVES To investigate in situ virus-specific differences in upper airway immune mediator levels during viral episodes of respiratory illnesses and the association with later asthma. METHODS We included 493 episodes of acute respiratory illnesses in 277 children aged 0-3 years from the COPSAC2010 mother-child cohort. Levels of 18 different immune mediators were assessed in nasal epithelial lining fluid using high-sensitivity MesoScale Discovery kits and compared between children with and without viral PCR-identification in nasopharyngeal samples. Finally, we investigated whether the virus-specific immune response was associated with asthma by age 6 years. RESULTS Viral detection were associated with upregulation of several Type 1 and regulatory immune mediators, including IFN-ɣ, TNF-α, CCL4, CXCL10 and IL-10 and downregulation of Type 2 and Type 17 immune mediators, including CCL13, and CXCL8 (FDR <0.05). Children developing asthma had decreased levels of IL-10 (FDR <0.05) during viral episodes compared to children not developing asthma. CONCLUSION We described the airway immune mediator profile during viral respiratory illnesses in early life and showed that children developing asthma by age 6 years have a reduced regulatory (IL-10) immune mediator level. This provides insight into the interplay between early-life viral infections, airway immunity and asthma development.
Collapse
Affiliation(s)
- Mathias Elsner Melgaard
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Signe Kjeldgaard Jensen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Anders Eliasen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Health Technology, Section for Bioinformatics, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Casper-Emil Tingskov Pedersen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jonathan Thorsen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Marianne Mikkelsen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Nilofar Vahman
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Ann-Marie Malby Schoos
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Pediatrics, Slagelse Sygehus, Slagelse, Denmark
| | - James Gern
- Department of Pediatrics and Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin, USA
| | - Susanne Brix
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Jakob Stokholm
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Pediatrics, Slagelse Sygehus, Slagelse, Denmark
- Department of Food Science, University of Copenhagen, Frederiksberg C, Denmark
| | - Bo Lund Chawes
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Klaus Bønnelykke
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
27
|
Hilser JR, Spencer NJ, Afshari K, Gilliland FD, Hu H, Deb A, Lusis AJ, Wilson Tang W, Hartiala JA, Hazen SL, Allayee H. COVID-19 Is a Coronary Artery Disease Risk Equivalent and Exhibits a Genetic Interaction With ABO Blood Type. Arterioscler Thromb Vasc Biol 2024; 44:2321-2333. [PMID: 39381876 PMCID: PMC11495539 DOI: 10.1161/atvbaha.124.321001] [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: 03/25/2024] [Accepted: 08/08/2024] [Indexed: 10/10/2024]
Abstract
BACKGROUND COVID-19 is associated with acute risk of major adverse cardiac events (MACE), including myocardial infarction, stroke, and mortality (all-cause). However, the duration and underlying determinants of heightened risk of cardiovascular disease and MACE post-COVID-19 are not known. METHODS Data from the UK Biobank was used to identify COVID-19 cases (n=10 005) who were positive for polymerase chain reaction (PCR+)-based tests for SARS-CoV-2 infection (n=8062) or received hospital-based International Classification of Diseases version-10 (ICD-10) codes for COVID-19 (n=1943) between February 1, 2020 and December 31, 2020. Population controls (n=217 730) and propensity score-matched controls (n=38 860) were also drawn from the UK Biobank during the same period. Proportional hazard models were used to evaluate COVID-19 for association with long-term (>1000 days) risk of MACE and as a coronary artery disease risk equivalent. Additional analyses examined whether COVID-19 interacted with genetic determinants to affect the risk of MACE and its components. RESULTS The risk of MACE was elevated in COVID-19 cases at all levels of severity (HR, 2.09 [95% CI, 1.94-2.25]; P<0.0005) and to a greater extent in cases hospitalized for COVID-19 (HR, 3.85 [95% CI, 3.51-4.24]; P<0.0005). Hospitalization for COVID-19 represented a coronary artery disease risk equivalent since incident MACE risk among cases without history of cardiovascular disease was even higher than that observed in patients with cardiovascular disease without COVID-19 (HR, 1.21 [95% CI, 1.08-1.37]; P<0.005). A significant genetic interaction was observed between the ABO locus and hospitalization for COVID-19 (Pinteraction=0.01), with risk of thrombotic events being increased in subjects with non-O blood types (HR, 1.65 [95% CI, 1.29-2.09]; P=4.8×10-5) to a greater extent than subjects with blood type O (HR, 0.96 [95% CI, 0.66-1.39]; P=0.82). CONCLUSIONS Hospitalization for COVID-19 represents a coronary artery disease risk equivalent, with post-acute myocardial infarction and stroke risk particularly heightened in non-O blood types. These results may have important clinical implications and represent, to our knowledge, one of the first examples of a gene-pathogen exposure interaction for thrombotic events.
Collapse
Affiliation(s)
- James R. Hilser
- Department of Population and Public Health Sciences (J.R.H., N.J.S., K.A., F.D.G., H.H., J.A.H., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
- Department of Biochemistry and Molecular Medicine (J.R.H., N.J.S., K.A., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
| | - Neal J. Spencer
- Department of Population and Public Health Sciences (J.R.H., N.J.S., K.A., F.D.G., H.H., J.A.H., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
- Department of Biochemistry and Molecular Medicine (J.R.H., N.J.S., K.A., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
| | - Kimia Afshari
- Department of Population and Public Health Sciences (J.R.H., N.J.S., K.A., F.D.G., H.H., J.A.H., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
- Department of Biochemistry and Molecular Medicine (J.R.H., N.J.S., K.A., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
| | - Frank D. Gilliland
- Department of Population and Public Health Sciences (J.R.H., N.J.S., K.A., F.D.G., H.H., J.A.H., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
| | - Howard Hu
- Department of Population and Public Health Sciences (J.R.H., N.J.S., K.A., F.D.G., H.H., J.A.H., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
| | - Arjun Deb
- Department of Medicine (A.D., A.J.L.), Keck School of Medicine, University of Southern California, Los Angeles
| | - Aldons J. Lusis
- Department of Medicine (A.D., A.J.L.), Keck School of Medicine, University of Southern California, Los Angeles
- Department of Microbiology, Immunology, and Molecular Genetics (A.J.L.), David Geffen School of Medicine of UCLA, CA
- Department of Human Genetics (A.J.L.), David Geffen School of Medicine of UCLA, CA
| | - W.H. Wilson Tang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute (W.H.W.T., S.L.H.), Cleveland Clinic, OH
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute (W.H.W.T., S.L.H.), Cleveland Clinic, OH
- Center for Microbiome and Human Health (W.H.W.T., S.L.H.), Cleveland Clinic, OH
| | - Jaana A. Hartiala
- Department of Population and Public Health Sciences (J.R.H., N.J.S., K.A., F.D.G., H.H., J.A.H., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
| | - Stanley L. Hazen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute (W.H.W.T., S.L.H.), Cleveland Clinic, OH
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute (W.H.W.T., S.L.H.), Cleveland Clinic, OH
- Center for Microbiome and Human Health (W.H.W.T., S.L.H.), Cleveland Clinic, OH
| | - Hooman Allayee
- Department of Population and Public Health Sciences (J.R.H., N.J.S., K.A., F.D.G., H.H., J.A.H., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
- Department of Biochemistry and Molecular Medicine (J.R.H., N.J.S., K.A., H.A.), Keck School of Medicine, University of Southern California, Los Angeles
| |
Collapse
|
28
|
Li M, Hao X, Shi D, Cheng S, Zhong Z, Cai L, Jiang M, Ding L, Ding L, Wang C, Yu X. Identification of susceptibility loci and relevant cell type for IgA nephropathy in Han Chinese by integrative genome-wide analysis. Front Med 2024; 18:862-877. [PMID: 39343836 DOI: 10.1007/s11684-024-1086-2] [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: 11/22/2023] [Accepted: 05/17/2024] [Indexed: 10/01/2024]
Abstract
Although many susceptibility loci for IgA nephropathy (IgAN) have been identified, they only account for 11.0% of the overall IgAN variance. We performed a large genome-wide meta-analysis of IgAN in Han Chinese with 3616 cases and 10 417 controls to identify additional genetic loci of IgAN. Considering that inflammatory bowel disease (IBD) and asthma might share an etiology of dysregulated mucosal immunity with IgAN, we performed cross-trait integrative analysis by leveraging functional annotations of relevant cell type and the pleiotropic information from IBD and asthma. Among 8 669 456 imputed variants, we identified a novel locus at 4p14 containing the long noncoding RNA LOC101060498. Cell type enrichment analysis based on annotations suggested that PMA-I-stimulated CD4+CD25-IL17+ Th17 cell was the most relevant cell type for IgAN, which highlights the essential role of Th17 pathway in the pathogenesis of IgAN. Furthermore, we identified six more novel loci associated with IgAN, which included three loci showing pleiotropic effects with IBD or asthma (2q35/PNKD, 6q25.2/SCAF8, and 22q11.21/UBE2L3) and three loci specific to IgAN (14q32.32/TRAF3, 16q22.2/TXNL4B, and 21q21.3/LINC00113) in the pleiotropic analysis. Our findings support the involvement of mucosal immunity, especially T cell immune response and IL-17 signal pathway, in the development of IgAN and shed light on further investigation of IgAN.
Collapse
Affiliation(s)
- Ming Li
- Department of Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangzhou, 510080, China
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dianchun Shi
- Department of Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangzhou, 510080, China
| | - Shanshan Cheng
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhong Zhong
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- NHC Key Laboratory of Nephrology (Sun Yat-sen University), and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Lu Cai
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- NHC Key Laboratory of Nephrology (Sun Yat-sen University), and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, 510080, China
| | - Minghui Jiang
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lin Ding
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lanbo Ding
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chaolong Wang
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Xueqing Yu
- Department of Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
- Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangzhou, 510080, China.
| |
Collapse
|
29
|
Rumker L, Sakaue S, Reshef Y, Kang JB, Yazar S, Alquicira-Hernandez J, Valencia C, Lagattuta KA, Mah-Som A, Nathan A, Powell JE, Loh PR, Raychaudhuri S. Identifying genetic variants that influence the abundance of cell states in single-cell data. Nat Genet 2024; 56:2068-2077. [PMID: 39327486 DOI: 10.1038/s41588-024-01909-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 08/14/2024] [Indexed: 09/28/2024]
Abstract
Disease risk alleles influence the composition of cells present in the body, but modeling genetic effects on the cell states revealed by single-cell profiling is difficult because variant-associated states may reflect diverse combinations of the profiled cell features that are challenging to predefine. We introduce Genotype-Neighborhood Associations (GeNA), a statistical tool to identify cell-state abundance quantitative trait loci (csaQTLs) in high-dimensional single-cell datasets. Instead of testing associations to predefined cell states, GeNA flexibly identifies the cell states whose abundance is most associated with genetic variants. In a genome-wide survey of single-cell RNA sequencing peripheral blood profiling from 969 individuals, GeNA identifies five independent loci associated with shifts in the relative abundance of immune cell states. For example, rs3003-T (P = 1.96 × 10-11) associates with increased abundance of natural killer cells expressing tumor necrosis factor response programs. This csaQTL colocalizes with increased risk for psoriasis, an autoimmune disease that responds to anti-tumor necrosis factor treatments. Flexibly characterizing csaQTLs for granular cell states may help illuminate how genetic background alters cellular composition to confer disease risk.
Collapse
Affiliation(s)
- Laurie Rumker
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Saori Sakaue
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yakir Reshef
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joyce B Kang
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Seyhan Yazar
- Translational Genomics, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- UNSW Cellular Genomics Futures Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Jose Alquicira-Hernandez
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Translational Genomics, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Cristian Valencia
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kaitlyn A Lagattuta
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Annelise Mah-Som
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Aparna Nathan
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joseph E Powell
- Translational Genomics, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- UNSW Cellular Genomics Futures Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Po-Ru Loh
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA.
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
30
|
Melén E, Zar HJ, Siroux V, Shaw D, Saglani S, Koppelman GH, Hartert T, Gern JE, Gaston B, Bush A, Zein J. Asthma Inception: Epidemiologic Risk Factors and Natural History Across the Life Course. Am J Respir Crit Care Med 2024; 210:737-754. [PMID: 38981012 PMCID: PMC11418887 DOI: 10.1164/rccm.202312-2249so] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 07/09/2024] [Indexed: 07/11/2024] Open
Abstract
Asthma is a descriptive label for an obstructive inflammatory disease in the lower airways manifesting with symptoms including breathlessness, cough, difficulty in breathing, and wheezing. From a clinician's point of view, asthma symptoms can commence at any age, although most patients with asthma-regardless of their age of onset-seem to have had some form of airway problems during childhood. Asthma inception and related pathophysiologic processes are therefore very likely to occur early in life, further evidenced by recent lung physiologic and mechanistic research. Herein, we present state-of-the-art updates on the role of genetics and epigenetics, early viral and bacterial infections, immune response, and pathophysiology, as well as lifestyle and environmental exposures, in asthma across the life course. We conclude that early environmental insults in genetically vulnerable individuals inducing abnormal, pre-asthmatic airway responses are key events in asthma inception, and we highlight disease heterogeneity across ages and the potential shortsightedness of treating all patients with asthma using the same treatments. Although there are no interventions that, at present, can modify long-term outcomes, a precision-medicine approach should be implemented to optimize treatment and tailor follow-up for all patients with asthma.
Collapse
Affiliation(s)
- Erik Melén
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Heather J. Zar
- Department of Paediatrics and Child Health and South African Medical Research Council Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Valerie Siroux
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Team of Environmental Epidemiology Applied to Development and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
| | - Dominic Shaw
- Department of Respiratory Sciences, University of Leicester, Leicester, United Kingdom
| | - Sejal Saglani
- National Heart and Lung Institute, Centre for Paediatrics and Child Health, Imperial College London, London, United Kingdom
| | - Gerard H. Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology, Groningen Research Institute for Asthma and COPD, University of Groningen, University Medical Center Groningen, Beatrix Children’s Hospital, Groningen, the Netherlands
| | - Tina Hartert
- Department of Medicine and Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - James E. Gern
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin
| | | | - Andrew Bush
- National Heart and Lung Institute, Centre for Paediatrics and Child Health, Imperial College London, London, United Kingdom
| | | |
Collapse
|
31
|
Stikker B, Trap L, Sedaghati-Khayat B, de Bruijn MJW, van Ijcken WFJ, de Roos E, Ikram A, Hendriks RW, Brusselle G, van Rooij J, Stadhouders R. Epigenomic partitioning of a polygenic risk score for asthma reveals distinct genetically driven disease pathways. Eur Respir J 2024; 64:2302059. [PMID: 38901884 PMCID: PMC11358516 DOI: 10.1183/13993003.02059-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 05/28/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Individual differences in susceptibility to developing asthma, a heterogeneous chronic inflammatory lung disease, are poorly understood. Whether genetics can predict asthma risk and how genetic variants modulate the complex pathophysiology of asthma are still debated. AIM To build polygenic risk scores for asthma risk prediction and epigenomically link predictive genetic variants to pathophysiological mechanisms. METHODS Restricted polygenic risk scores were constructed using single nucleotide variants derived from genome-wide association studies and validated using data generated in the Rotterdam Study, a Dutch prospective cohort of 14 926 individuals. Outcomes used were asthma, childhood-onset asthma, adulthood-onset asthma, eosinophilic asthma and asthma exacerbations. Genome-wide chromatin analysis data from 19 disease-relevant cell types were used for epigenomic polygenic risk score partitioning. RESULTS The polygenic risk scores obtained predicted asthma and related outcomes, with the strongest associations observed for childhood-onset asthma (2.55 odds ratios per polygenic risk score standard deviation, area under the curve of 0.760). Polygenic risk scores allowed for the classification of individuals into high-risk and low-risk groups. Polygenic risk score partitioning using epigenomic profiles identified five clusters of variants within putative gene regulatory regions linked to specific asthma-relevant cells, genes and biological pathways. CONCLUSIONS Polygenic risk scores were associated with asthma(-related traits) in a Dutch prospective cohort, with substantially higher predictive power observed for childhood-onset than adult-onset asthma. Importantly, polygenic risk score variants could be epigenomically partitioned into clusters of regulatory variants with different pathophysiological association patterns and effect estimates, which likely represent distinct genetically driven disease pathways. Our findings have potential implications for personalised risk mitigation and treatment strategies.
Collapse
Affiliation(s)
- Bernard Stikker
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Lianne Trap
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- L. Trap and B. Sedaghati-Khayat made an equal contribution to this study
| | - Bahar Sedaghati-Khayat
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- L. Trap and B. Sedaghati-Khayat made an equal contribution to this study
| | - Marjolein J W de Bruijn
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Wilfred F J van Ijcken
- Center for Biomics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Emmely de Roos
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Arfan Ikram
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Guy Brusselle
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jeroen van Rooij
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- J. van Rooij and R. Stadhouders contributed equally to this article as lead authors and supervised the work
| | - Ralph Stadhouders
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- J. van Rooij and R. Stadhouders contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
32
|
Carey CE, Shafee R, Wedow R, Elliott A, Palmer DS, Compitello J, Kanai M, Abbott L, Schultz P, Karczewski KJ, Bryant SC, Cusick CM, Churchhouse C, Howrigan DP, King D, Davey Smith G, Neale BM, Walters RK, Robinson EB. Principled distillation of UK Biobank phenotype data reveals underlying structure in human variation. Nat Hum Behav 2024; 8:1599-1615. [PMID: 38965376 PMCID: PMC11343713 DOI: 10.1038/s41562-024-01909-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 05/14/2024] [Indexed: 07/06/2024]
Abstract
Data within biobanks capture broad yet detailed indices of human variation, but biobank-wide insights can be difficult to extract due to complexity and scale. Here, using large-scale factor analysis, we distill hundreds of variables (diagnoses, assessments and survey items) into 35 latent constructs, using data from unrelated individuals with predominantly estimated European genetic ancestry in UK Biobank. These factors recapitulate known disease classifications, disentangle elements of socioeconomic status, highlight the relevance of psychiatric constructs to health and improve measurement of pro-health behaviours. We go on to demonstrate the power of this approach to clarify genetic signal, enhance discovery and identify associations between underlying phenotypic structure and health outcomes. In building a deeper understanding of ways in which constructs such as socioeconomic status, trauma, or physical activity are structured in the dataset, we emphasize the importance of considering the interwoven nature of the human phenome when evaluating public health patterns.
Collapse
Affiliation(s)
- Caitlin E Carey
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA.
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
| | - Rebecca Shafee
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Section on Developmental Neurogenomics, National Institute of Mental Health, Bethesda, MD, USA
| | - Robbee Wedow
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Sociology, Purdue University, West Lafayette, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
- AnalytiXIN, Indianapolis, IN, USA
- Center on Aging and the Life Course, Purdue University, West Lafayette, IN, USA
- Department of Statistics, Purdue University, West Lafayette, IN, USA
| | - Amanda Elliott
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Duncan S Palmer
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Nuffield Department of Population Health, Medical Sciences Division University of Oxford, Oxford, UK
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - John Compitello
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Masahiro Kanai
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Liam Abbott
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Patrick Schultz
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Konrad J Karczewski
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Samuel C Bryant
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Caroline M Cusick
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Claire Churchhouse
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Daniel P Howrigan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Daniel King
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - George Davey Smith
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- MRC Integrative Epidemiology Unit, University of Bristol, Oakfield House, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Benjamin M Neale
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Raymond K Walters
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | - Elise B Robinson
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
33
|
Stefansson OA, Sigurpalsdottir BD, Rognvaldsson S, Halldorsson GH, Juliusson K, Sveinbjornsson G, Gunnarsson B, Beyter D, Jonsson H, Gudjonsson SA, Olafsdottir TA, Saevarsdottir S, Magnusson MK, Lund SH, Tragante V, Oddsson A, Hardarson MT, Eggertsson HP, Gudmundsson RL, Sverrisson S, Frigge ML, Zink F, Holm H, Stefansson H, Rafnar T, Jonsdottir I, Sulem P, Helgason A, Gudbjartsson DF, Halldorsson BV, Thorsteinsdottir U, Stefansson K. The correlation between CpG methylation and gene expression is driven by sequence variants. Nat Genet 2024; 56:1624-1631. [PMID: 39048797 PMCID: PMC11319203 DOI: 10.1038/s41588-024-01851-2] [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: 04/13/2022] [Accepted: 06/27/2024] [Indexed: 07/27/2024]
Abstract
Gene promoter and enhancer sequences are bound by transcription factors and are depleted of methylated CpG sites (cytosines preceding guanines in DNA). The absence of methylated CpGs in these sequences typically correlates with increased gene expression, indicating a regulatory role for methylation. We used nanopore sequencing to determine haplotype-specific methylation rates of 15.3 million CpG units in 7,179 whole-blood genomes. We identified 189,178 methylation depleted sequences where three or more proximal CpGs were unmethylated on at least one haplotype. A total of 77,789 methylation depleted sequences (~41%) associated with 80,503 cis-acting sequence variants, which we termed allele-specific methylation quantitative trait loci (ASM-QTLs). RNA sequencing of 896 samples from the same blood draws used to perform nanopore sequencing showed that the ASM-QTL, that is, DNA sequence variability, drives most of the correlation found between gene expression and CpG methylation. ASM-QTLs were enriched 40.2-fold (95% confidence interval 32.2, 49.9) among sequence variants associating with hematological traits, demonstrating that ASM-QTLs are important functional units in the noncoding genome.
Collapse
Affiliation(s)
| | - Brynja Dogg Sigurpalsdottir
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- School of Technology, Reykjavik University, Reykjavik, Iceland
| | | | - Gisli Hreinn Halldorsson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | | | | | | | | | - Thorunn Asta Olafsdottir
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Saedis Saevarsdottir
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Magnus Karl Magnusson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Sigrun Helga Lund
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | | | | | - Marteinn Thor Hardarson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- School of Technology, Reykjavik University, Reykjavik, Iceland
| | | | | | | | | | | | - Hilma Holm
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
| | | | | | - Ingileif Jonsdottir
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | | | - Agnar Helgason
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Department of Anthropology, University of Iceland, Reykjavik, Iceland
| | - Daniel F Gudbjartsson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | - Bjarni V Halldorsson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- School of Technology, Reykjavik University, Reykjavik, Iceland
| | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Kari Stefansson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland.
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.
| |
Collapse
|
34
|
Sayers I, John C, Chen J, Hall IP. Genetics of chronic respiratory disease. Nat Rev Genet 2024; 25:534-547. [PMID: 38448562 DOI: 10.1038/s41576-024-00695-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2024] [Indexed: 03/08/2024]
Abstract
Chronic respiratory diseases, such as chronic obstructive pulmonary disease (COPD), asthma and interstitial lung diseases are frequently occurring disorders with a polygenic basis that account for a large global burden of morbidity and mortality. Recent large-scale genetic epidemiology studies have identified associations between genetic variation and individual respiratory diseases and linked specific genetic variants to quantitative traits related to lung function. These associations have improved our understanding of the genetic basis and mechanisms underlying common lung diseases. Moreover, examining the overlap between genetic associations of different respiratory conditions, along with evidence for gene-environment interactions, has yielded additional biological insights into affected molecular pathways. This genetic information could inform the assessment of respiratory disease risk and contribute to stratified treatment approaches.
Collapse
Affiliation(s)
- Ian Sayers
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, University Park, Nottingham, UK
- Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, UK
| | - Catherine John
- University of Leicester, Leicester, UK
- University Hospitals of Leicester, Leicester, UK
| | - Jing Chen
- University of Leicester, Leicester, UK
| | - Ian P Hall
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, University Park, Nottingham, UK.
- Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham, UK.
| |
Collapse
|
35
|
Jeong A. Systems biology approaches to utilise polygenic risk scores for chronic diseases. Eur Respir J 2024; 64:2401133. [PMID: 39209466 DOI: 10.1183/13993003.01133-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 09/04/2024]
Affiliation(s)
- Ayoung Jeong
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| |
Collapse
|
36
|
Mack TM, Raddatz MA, Pershad Y, Nachun DC, Taylor KD, Guo X, Shuldiner AR, O'Connell JR, Kenny EE, Loos RJF, Redline S, Cade BE, Psaty BM, Bis JC, Brody JA, Silverman EK, Yun JH, Cho MH, DeMeo DL, Levy D, Johnson AD, Mathias RA, Yanek LR, Heckbert SR, Smith NL, Wiggins KL, Raffield LM, Carson AP, Rotter JI, Rich SS, Manichaikul AW, Gu CC, Chen YDI, Lee WJ, Shoemaker MB, Roden DM, Kooperberg C, Auer PL, Desai P, Blackwell TW, Smith AV, Reiner AP, Jaiswal S, Weinstock JS, Bick AG. Epigenetic and proteomic signatures associate with clonal hematopoiesis expansion rate. NATURE AGING 2024; 4:1043-1052. [PMID: 38834882 PMCID: PMC11832052 DOI: 10.1038/s43587-024-00647-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 05/08/2024] [Indexed: 06/06/2024]
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP), whereby somatic mutations in hematopoietic stem cells confer a selective advantage and drive clonal expansion, not only correlates with age but also confers increased risk of morbidity and mortality. Here, we leverage genetically predicted traits to identify factors that determine CHIP clonal expansion rate. We used the passenger-approximated clonal expansion rate method to quantify the clonal expansion rate for 4,370 individuals in the National Heart, Lung, and Blood Institute (NHLBI) Trans-Omics for Precision Medicine (TOPMed) cohort and calculated polygenic risk scores for DNA methylation aging, inflammation-related measures and circulating protein levels. Clonal expansion rate was significantly associated with both genetically predicted and measured epigenetic clocks. No associations were identified with inflammation-related lab values or diseases and CHIP expansion rate overall. A proteome-wide search identified predicted circulating levels of myeloid zinc finger 1 and anti-Müllerian hormone as associated with an increased CHIP clonal expansion rate and tissue inhibitor of metalloproteinase 1 and glycine N-methyltransferase as associated with decreased CHIP clonal expansion rate. Together, our findings identify epigenetic and proteomic patterns associated with the rate of hematopoietic clonal expansion.
Collapse
Affiliation(s)
- Taralynn M Mack
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Michael A Raddatz
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yash Pershad
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Daniel C Nachun
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Alan R Shuldiner
- Department of Medicine, University of Maryland, Baltimore, Baltimore, MD, USA
| | - Jeffrey R O'Connell
- Department of Medicine, University of Maryland, Baltimore, Baltimore, MD, USA
| | - Eimear E Kenny
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ruth J F Loos
- The Charles Bronfman Institute of Personalized Medicine, Mount Sinai Hospital, New York City, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Susan Redline
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Brian E Cade
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Jeong H Yun
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Michael H Cho
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Dawn L DeMeo
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Daniel Levy
- National Heart, Lung and Blood Institute, Population Sciences Branch, Framingham, MA, USA
| | - Andrew D Johnson
- National Heart, Lung and Blood Institute, Population Sciences Branch, Framingham, MA, USA
| | - Rasika A Mathias
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lisa R Yanek
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Susan R Heckbert
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA, USA
| | - Nicholas L Smith
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA, USA
- Seattle Epidemiologic Research and Information Center, Department of Veterans Affairs Office of Research and Development, Seattle, WA, USA
| | - Kerri L Wiggins
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - April P Carson
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Ani W Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - C Charles Gu
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Yii-Der Ida Chen
- Medical Genetics Translational Genomics and Population Sciences (TGPS), Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Wen-Jane Lee
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City, Taiwan
| | - M Benjamin Shoemaker
- Division of Cardiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dan M Roden
- Departments of Medicine, Pharmacology, and Biomedical Informatics, Vanderbilt University, Nashville, TN, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Paul L Auer
- Division of Biostatistics, Institute for Health and Equity, and Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Pinkal Desai
- Division of Hematology and Oncology, Weill Cornell Medicine, New York, NY, USA
- Englander Institute of Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Thomas W Blackwell
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Albert V Smith
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Alexander P Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Joshua S Weinstock
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Alexander G Bick
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
37
|
Risemberg EL, Smeekens JM, Cruz Cisneros MC, Hampton BK, Hock P, Linnertz CL, Miller DR, Orgel K, Shaw GD, de Villena FPM, Burks AW, Valdar W, Kulis MD, Ferris MT. A mutation in Themis contributes to anaphylaxis severity following oral peanut challenge in CC027 mice. J Allergy Clin Immunol 2024; 154:387-397. [PMID: 38670234 PMCID: PMC11323216 DOI: 10.1016/j.jaci.2024.03.027] [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: 09/07/2023] [Revised: 03/12/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND The development of peanut allergy is due to a combination of genetic and environmental factors, although specific genes have proven difficult to identify. Previously, we reported that peanut-sensitized Collaborative Cross strain CC027/GeniUnc (CC027) mice develop anaphylaxis upon oral challenge to peanut, in contrast to C3H/HeJ (C3H) mice. OBJECTIVE This study aimed to determine the genetic basis of orally induced anaphylaxis to peanut in CC027 mice. METHODS A genetic mapping population between CC027 and C3H mice was designed to identify the genetic factors that drive oral anaphylaxis. A total of 356 CC027xC3H backcrossed mice were generated, sensitized to peanut, then challenged to peanut by oral gavage. Anaphylaxis and peanut-specific IgE were quantified for all mice. T-cell phenotyping was conducted on CC027 mice and 5 additional Collaborative Cross strains. RESULTS Anaphylaxis to peanut was absent in 77% of backcrossed mice, with 19% showing moderate anaphylaxis and 4% having severe anaphylaxis. There were 8 genetic loci associated with variation in response to peanut challenge-6 associated with anaphylaxis (temperature decrease) and 2 associated with peanut-specific IgE levels. There were 2 major loci that impacted multiple aspects of the severity of acute anaphylaxis, at which the CC027 allele was associated with worse outcome. At one of these loci, CC027 has a private genetic variant in the Themis gene. Consistent with described functions of Themis, we found that CC027 mice have more immature T cells with fewer CD8+, CD4+, and CD4+CD25+CD127- regulatory T cells. CONCLUSIONS Our results demonstrate a key role for Themis in the orally reactive CC027 mouse model of peanut allergy.
Collapse
Affiliation(s)
- Ellen L Risemberg
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Johanna M Smeekens
- Division of Allergy and Immunology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Marta C Cruz Cisneros
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC; Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Brea K Hampton
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC; Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Pablo Hock
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Colton L Linnertz
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Darla R Miller
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Kelly Orgel
- Division of Allergy and Immunology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Ginger D Shaw
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Fernando Pardo Manuel de Villena
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - A Wesley Burks
- Division of Allergy and Immunology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - William Valdar
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC.
| | - Michael D Kulis
- Division of Allergy and Immunology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC.
| | - Martin T Ferris
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC.
| |
Collapse
|
38
|
Liu T, Woodruff PG, Zhou X. Advances in non-type 2 severe asthma: from molecular insights to novel treatment strategies. Eur Respir J 2024; 64:2300826. [PMID: 38697650 PMCID: PMC11325267 DOI: 10.1183/13993003.00826-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 04/18/2024] [Indexed: 05/05/2024]
Abstract
Asthma is a prevalent pulmonary disease that affects more than 300 million people worldwide and imposes a substantial economic burden. While medication can effectively control symptoms in some patients, severe asthma attacks, driven by airway inflammation induced by environmental and infectious exposures, continue to be a major cause of asthma-related mortality. Heterogeneous phenotypes of asthma include type 2 (T2) and non-T2 asthma. Non-T2 asthma is often observed in patients with severe and/or steroid-resistant asthma. This review covers the molecular mechanisms, clinical phenotypes, causes and promising treatments of non-T2 severe asthma. Specifically, we discuss the signalling pathways for non-T2 asthma including the activation of inflammasomes, interferon responses and interleukin-17 pathways, and their contributions to the subtypes, progression and severity of non-T2 asthma. Understanding the molecular mechanisms and genetic determinants underlying non-T2 asthma could form the basis for precision medicine in severe asthma treatment.
Collapse
Affiliation(s)
- Tao Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Nanjing, China
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Prescott G Woodruff
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine and Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
39
|
Oguchi A, Suzuki A, Komatsu S, Yoshitomi H, Bhagat S, Son R, Bonnal RJP, Kojima S, Koido M, Takeuchi K, Myouzen K, Inoue G, Hirai T, Sano H, Takegami Y, Kanemaru A, Yamaguchi I, Ishikawa Y, Tanaka N, Hirabayashi S, Konishi R, Sekito S, Inoue T, Kere J, Takeda S, Takaori-Kondo A, Endo I, Kawaoka S, Kawaji H, Ishigaki K, Ueno H, Hayashizaki Y, Pagani M, Carninci P, Yanagita M, Parrish N, Terao C, Yamamoto K, Murakawa Y. An atlas of transcribed enhancers across helper T cell diversity for decoding human diseases. Science 2024; 385:eadd8394. [PMID: 38963856 DOI: 10.1126/science.add8394] [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: 07/17/2022] [Accepted: 05/01/2024] [Indexed: 07/06/2024]
Abstract
Transcribed enhancer maps can reveal nuclear interactions underpinning each cell type and connect specific cell types to diseases. Using a 5' single-cell RNA sequencing approach, we defined transcription start sites of enhancer RNAs and other classes of coding and noncoding RNAs in human CD4+ T cells, revealing cellular heterogeneity and differentiation trajectories. Integration of these datasets with single-cell chromatin profiles showed that active enhancers with bidirectional RNA transcription are highly cell type-specific and that disease heritability is strongly enriched in these enhancers. The resulting cell type-resolved multimodal atlas of bidirectionally transcribed enhancers, which we linked with promoters using fine-scale chromatin contact maps, enabled us to systematically interpret genetic variants associated with a range of immune-mediated diseases.
Collapse
Affiliation(s)
- Akiko Oguchi
- RIKEN-IFOM Joint Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akari Suzuki
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Shuichiro Komatsu
- RIKEN-IFOM Joint Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- IFOM ETS - the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Hiroyuki Yoshitomi
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
- Department of Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shruti Bhagat
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
| | - Raku Son
- RIKEN-IFOM Joint Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Shohei Kojima
- Genome Immunobiology RIKEN Hakubi Research Team, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Masaru Koido
- Division of Molecular Pathology, Department of Cancer Biology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kazuhiro Takeuchi
- RIKEN-IFOM Joint Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
- Department of Medical Systems Genomics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keiko Myouzen
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Gyo Inoue
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Tomoya Hirai
- RIKEN-IFOM Joint Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Hiromi Sano
- RIKEN-IFOM Joint Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | | | | | | | - Yuki Ishikawa
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Nao Tanaka
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Shigeki Hirabayashi
- RIKEN-IFOM Joint Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Division of Precision Medicine, Kyushu University Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Riyo Konishi
- Inter-Organ Communication Research Team, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Sho Sekito
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Mie University, Tsu, Japan
| | - Takahiro Inoue
- Department of Nephro-Urologic Surgery and Andrology, Mie University Graduate School of Medicine, Mie University, Tsu, Japan
| | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
- Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
- Folkhalsan Research Center, Helsinki, Finland
| | - Shunichi Takeda
- Department of Radiation Genetics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Akifumi Takaori-Kondo
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Shinpei Kawaoka
- Inter-Organ Communication Research Team, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Department of Integrative Bioanalytics, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Hideya Kawaji
- Research Center for Genome & Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
- Preventive Medicine and Applied Genomics Unit, RIKEN Center for Integrative Medical Science, Yokohama, Japan
- RIKEN Preventive Medicine and Diagnosis Innovation Program, Wako, Japan
| | - Kazuyoshi Ishigaki
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Hideki Ueno
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
- Department of Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshihide Hayashizaki
- K.K. DNAFORM, Yokohama, Japan
- RIKEN Preventive Medicine and Diagnosis Innovation Program, Wako, Japan
| | - Massimiliano Pagani
- IFOM ETS - the AIRC Institute of Molecular Oncology, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi, Milan, Italy
| | - Piero Carninci
- Laboratory for Transcriptome Technology, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Human Technopole, Milan, Italy
| | - Motoko Yanagita
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Nicholas Parrish
- Genome Immunobiology RIKEN Hakubi Research Team, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan
- Department of Applied Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Kazuhiko Yamamoto
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yasuhiro Murakawa
- RIKEN-IFOM Joint Laboratory for Cancer Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
- IFOM ETS - the AIRC Institute of Molecular Oncology, Milan, Italy
- Department of Medical Systems Genomics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
40
|
Xiu Z, Sun L, Liu K, Cao H, Qu HQ, Glessner JT, Ding Z, Zheng G, Wang N, Xia Q, Li J, Li MJ, Hakonarson H, Liu W, Li J. Shared molecular mechanisms and transdiagnostic potential of neurodevelopmental disorders and immune disorders. Brain Behav Immun 2024; 119:767-780. [PMID: 38677625 DOI: 10.1016/j.bbi.2024.04.026] [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: 10/21/2023] [Revised: 02/27/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024] Open
Abstract
The co-occurrence and familial clustering of neurodevelopmental disorders and immune disorders suggest shared genetic risk factors. Based on genome-wide association summary statistics from five neurodevelopmental disorders and four immune disorders, we conducted genome-wide, local genetic correlation and polygenic overlap analysis. We further performed a cross-trait GWAS meta-analysis. Pleotropic loci shared between the two categories of diseases were mapped to candidate genes using multiple algorithms and approaches. Significant genetic correlations were observed between neurodevelopmental disorders and immune disorders, including both positive and negative correlations. Neurodevelopmental disorders exhibited higher polygenicity compared to immune disorders. Around 50%-90% of genetic variants of the immune disorders were shared with neurodevelopmental disorders. The cross-trait meta-analysis revealed 154 genome-wide significant loci, including 8 novel pleiotropic loci. Significant associations were observed for 30 loci with both types of diseases. Pathway analysis on the candidate genes at these loci revealed common pathways shared by the two types of diseases, including neural signaling, inflammatory response, and PI3K-Akt signaling pathway. In addition, 26 of the 30 lead SNPs were associated with blood cell traits. Neurodevelopmental disorders exhibit complex polygenic architecture, with a subset of individuals being at a heightened genetic risk for both neurodevelopmental and immune disorders. The identification of pleiotropic loci has important implications for exploring opportunities for drug repurposing, enabling more accurate patient stratification, and advancing genomics-informed precision in the medical field of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Zhanjie Xiu
- Department of Cell Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Medical Epigenetics, Tianjin Institute of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China; Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ling Sun
- Department of Child and Adolescent Psychology, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Kunlun Liu
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Haiyan Cao
- Department of Child and Adolescent Psychology, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, Tianjin, China
| | - Hui-Qi Qu
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Joseph T Glessner
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States; Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, United States; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Zhiyong Ding
- Mills Institute for Personalized Cancer Care, Fynn Biotechnologies Ltd., Jinan, China
| | - Gang Zheng
- National Supercomputer Center in Tianjin (NSCC-TJ), Tianjin, China
| | - Nan Wang
- Mills Institute for Personalized Cancer Care, Fynn Biotechnologies Ltd., Jinan, China
| | - Qianghua Xia
- Department of Cell Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Medical Epigenetics, Tianjin Institute of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China; Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jie Li
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Tianjin Medical University, Tianjin, China
| | - Mulin Jun Li
- Department of Bioinformatics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, United States; Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, United States; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| | - Wei Liu
- Tianjin Children's Hospital (Tianjin University Children's Hospital), Tianjin, China; Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin, China.
| | - Jin Li
- Department of Cell Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Medical Epigenetics, Tianjin Institute of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China; Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
41
|
Szczesny B, Boorgula MP, Chavan S, Campbell M, Johnson RK, Kammers K, Thompson EE, Cox MS, Shankar G, Cox C, Morin A, Lorizio W, Daya M, Kelada SNP, Beaty TH, Doumatey AP, Cruz AA, Watson H, Naureckas ET, Giles BL, Arinola GA, Sogaolu O, Falade AG, Hansel NN, Yang IV, Olopade CO, Rotimi CN, Landis RC, Figueiredo CA, Altman MC, Kenny E, Ruczinski I, Liu AH, Ober C, Taub MA, Barnes KC, Mathias RA. Multi-omics in nasal epithelium reveals three axes of dysregulation for asthma risk in the African Diaspora populations. Nat Commun 2024; 15:4546. [PMID: 38806494 PMCID: PMC11133339 DOI: 10.1038/s41467-024-48507-7] [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: 05/18/2023] [Accepted: 05/02/2024] [Indexed: 05/30/2024] Open
Abstract
Asthma has striking disparities across ancestral groups, but the molecular underpinning of these differences is poorly understood and minimally studied. A goal of the Consortium on Asthma among African-ancestry Populations in the Americas (CAAPA) is to understand multi-omic signatures of asthma focusing on populations of African ancestry. RNASeq and DNA methylation data are generated from nasal epithelium including cases (current asthma, N = 253) and controls (never-asthma, N = 283) from 7 different geographic sites to identify differentially expressed genes (DEGs) and gene networks. We identify 389 DEGs; the top DEG, FN1, was downregulated in cases (q = 3.26 × 10-9) and encodes fibronectin which plays a role in wound healing. The top three gene expression modules implicate networks related to immune response (CEACAM5; p = 9.62 × 10-16 and CPA3; p = 2.39 × 10-14) and wound healing (FN1; p = 7.63 × 10-9). Multi-omic analysis identifies FKBP5, a co-chaperone of glucocorticoid receptor signaling known to be involved in drug response in asthma, where the association between nasal epithelium gene expression is likely regulated by methylation and is associated with increased use of inhaled corticosteroids. This work reveals molecular dysregulation on three axes - increased Th2 inflammation, decreased capacity for wound healing, and impaired drug response - that may play a critical role in asthma within the African Diaspora.
Collapse
Affiliation(s)
- Brooke Szczesny
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Meher Preethi Boorgula
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Sameer Chavan
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Monica Campbell
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Randi K Johnson
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, USA
- Quantitative Sciences Division, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kai Kammers
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Emma E Thompson
- Division of Allergy and Infectious Diseases, Dept of Medicine, University of Washington, Seattle, WA, USA
| | - Madison S Cox
- Division of Allergy and Infectious Diseases, Dept of Medicine, University of Washington, Seattle, WA, USA
| | - Gautam Shankar
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Corey Cox
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Andréanne Morin
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Wendy Lorizio
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Michelle Daya
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Samir N P Kelada
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Terri H Beaty
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ayo P Doumatey
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alvaro A Cruz
- Fundacao ProAR and Federal University of Bahia, Salvador, Bahia, Brazil
| | - Harold Watson
- Faculty of Medical Sciences, The University of the West Indies, Queen Elizabeth Hospital, St. Michael, Bridgetown, Barbados
| | | | - B Louise Giles
- Departments of Pediatrics, University of Chicago, Chicago, IL, USA
| | - Ganiyu A Arinola
- Department of Immunology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Olumide Sogaolu
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adegoke G Falade
- Department of Pediatrics, University of Ibadan, and University College Hospital, Ibadan, Nigeria
| | - Nadia N Hansel
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Ivana V Yang
- Departments of Biomedical Informatics and Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | | | - Charles N Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - R Clive Landis
- Edmund Cohen Laboratory for Vascular Research, George Alleyne Chronic Disease Research Centre, Caribbean Institute for Health Research, The University of the West Indies, Cave Hill Campus, Wanstead, Barbados
| | - Camila A Figueiredo
- Federal University of Bahia and Funda. Program for Control of Asthma in Bahia (ProAR), Salvador, Brazil
- Instituto de Ciências de Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | - Matthew C Altman
- Systems Immunology Program, Benaroya Research Institute, Seattle, WA, 98101, USA
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA, 98109, USA
| | - Eimear Kenny
- Center for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ingo Ruczinski
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Andrew H Liu
- Department of Pediatrics, Childrens Hospital Colorado and University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Carole Ober
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Margaret A Taub
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kathleen C Barnes
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.
| | - Rasika A Mathias
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
42
|
Moix S, Sadler MC, Kutalik Z, Auwerx C. Breaking down causes, consequences, and mediating effects of telomere length variation on human health. Genome Biol 2024; 25:125. [PMID: 38760657 PMCID: PMC11101352 DOI: 10.1186/s13059-024-03269-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 05/07/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Telomeres form repeated DNA sequences at the ends of chromosomes, which shorten with each cell division. Yet, factors modulating telomere attrition and the health consequences thereof are not fully understood. To address this, we leveraged data from 326,363 unrelated UK Biobank participants of European ancestry. RESULTS Using linear regression and bidirectional univariable and multivariable Mendelian randomization (MR), we elucidate the relationships between leukocyte telomere length (LTL) and 142 complex traits, including diseases, biomarkers, and lifestyle factors. We confirm that telomeres shorten with age and show a stronger decline in males than in females, with these factors contributing to the majority of the 5.4% of LTL variance explained by the phenome. MR reveals 23 traits modulating LTL. Smoking cessation and high educational attainment associate with longer LTL, while weekly alcohol intake, body mass index, urate levels, and female reproductive events, such as childbirth, associate with shorter LTL. We also identify 24 traits affected by LTL, with risk for cardiovascular, pulmonary, and some autoimmune diseases being increased by short LTL, while longer LTL increased risk for other autoimmune conditions and cancers. Through multivariable MR, we show that LTL may partially mediate the impact of educational attainment, body mass index, and female age at childbirth on proxied lifespan. CONCLUSIONS Our study sheds light on the modulators, consequences, and the mediatory role of telomeres, portraying an intricate relationship between LTL, diseases, lifestyle, and socio-economic factors.
Collapse
Affiliation(s)
- Samuel Moix
- Department of Computational Biology, UNIL, Lausanne, 1015, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland.
| | - Marie C Sadler
- Department of Computational Biology, UNIL, Lausanne, 1015, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
- University Center for Primary Care and Public Health, Lausanne, 1015, Switzerland
| | - Zoltán Kutalik
- Department of Computational Biology, UNIL, Lausanne, 1015, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland.
- University Center for Primary Care and Public Health, Lausanne, 1015, Switzerland.
| | - Chiara Auwerx
- Department of Computational Biology, UNIL, Lausanne, 1015, Switzerland.
- Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland.
- University Center for Primary Care and Public Health, Lausanne, 1015, Switzerland.
- Center for Integrative Genetics, UNIL, Lausanne, 1015, Switzerland.
| |
Collapse
|
43
|
Lincoln MR, Connally N, Axisa PP, Gasperi C, Mitrovic M, van Heel D, Wijmenga C, Withoff S, Jonkers IH, Padyukov L, Rich SS, Graham RR, Gaffney PM, Langefeld CD, Vyse TJ, Hafler DA, Chun S, Sunyaev SR, Cotsapas C. Genetic mapping across autoimmune diseases reveals shared associations and mechanisms. Nat Genet 2024; 56:838-845. [PMID: 38741015 DOI: 10.1038/s41588-024-01732-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 03/21/2024] [Indexed: 05/16/2024]
Abstract
Autoimmune and inflammatory diseases are polygenic disorders of the immune system. Many genomic loci harbor risk alleles for several diseases, but the limited resolution of genetic mapping prevents determining whether the same allele is responsible, indicating a shared underlying mechanism. Here, using a collection of 129,058 cases and controls across 6 diseases, we show that ~40% of overlapping associations are due to the same allele. We improve fine-mapping resolution for shared alleles twofold by combining cases and controls across diseases, allowing us to identify more expression quantitative trait loci driven by the shared alleles. The patterns indicate widespread sharing of pathogenic mechanisms but not a single global autoimmune mechanism. Our approach can be applied to any set of traits and is particularly valuable as sample collections become depleted.
Collapse
Affiliation(s)
- Matthew R Lincoln
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Division of Neurology at the Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Noah Connally
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Pierre-Paul Axisa
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | | | - Mitja Mitrovic
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Faculty of Chemistry and Chemical Engineering, University of Maribor, Maribor, Slovenia
| | - David van Heel
- Blizard Institute, Queen Mary University of London, London, UK
| | - Cisca Wijmenga
- Department of Genetics at the University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Sebo Withoff
- Department of Genetics at the University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Iris H Jonkers
- Department of Genetics at the University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Leonid Padyukov
- Division of Rheumatology at the Department of Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Robert R Graham
- Maze Therapeutics, South San Francisco, CA, USA
- Genentech, South San Francisco, CA, USA
| | - Patrick M Gaffney
- Genes and Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Carl D Langefeld
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Timothy J Vyse
- Department of Medical and Molecular Genetics, Kings College London, London, UK
| | - David A Hafler
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Sung Chun
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Shamil R Sunyaev
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Chris Cotsapas
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA.
- Vesalius Therapeutics, Cambridge, MA, USA.
| |
Collapse
|
44
|
Sakano Y, Sakano K, Hurrell BP, Helou DG, Shafiei-Jahani P, Kazemi MH, Li X, Shen S, Hilser JR, Hartiala JA, Allayee H, Barbers R, Akbari O. Blocking CD226 regulates type 2 innate lymphoid cell effector function and alleviates airway hyperreactivity. J Allergy Clin Immunol 2024; 153:1406-1422.e6. [PMID: 38244725 DOI: 10.1016/j.jaci.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/19/2023] [Accepted: 01/03/2024] [Indexed: 01/22/2024]
Abstract
BACKGROUND Type 2 innate lymphoid cells (ILC2s) play a pivotal role in type 2 asthma. CD226 is a costimulatory molecule involved in various inflammatory diseases. OBJECTIVE We aimed to investigate CD226 expression and function within human and mouse ILC2s, and to assess the impact of targeting CD226 on ILC2-mediated airway hyperreactivity (AHR). METHODS We administered IL-33 intranasally to wild-type mice, followed by treatment with anti-CD226 antibody or isotype control. Pulmonary ILC2s were sorted for ex vivo analyses through RNA sequencing and flow cytometry. Next, we evaluated the effects of CD226 on AHR and lung inflammation in wild-type and Rag2-/- mice. Additionally, we compared peripheral ILC2s from healthy donors and asthmatic patients to ascertain the role of CD226 in human ILC2s. RESULTS Our findings demonstrated an inducible expression of CD226 in activated ILC2s, enhancing their cytokine secretion and effector functions. Mechanistically, CD226 alters intracellular metabolism and enhances PI3K/AKT and MAPK signal pathways. Blocking CD226 ameliorates ILC2-dependent AHR in IL-33 and Alternaria alternata-induced models. Interestingly, CD226 is expressed and inducible in human ILC2s, and its blocking reduces cytokine production. Finally, we showed that peripheral ILC2s in asthmatic patients exhibited elevated CD226 expression compared to healthy controls. CONCLUSION Our findings underscore the potential of CD226 as a novel therapeutic target in ILC2s, presenting a promising avenue for ameliorating AHR and allergic asthma.
Collapse
Affiliation(s)
- Yoshihiro Sakano
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Kei Sakano
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Benjamin P Hurrell
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Doumet Georges Helou
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Pedram Shafiei-Jahani
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Mohammad H Kazemi
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Xin Li
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Stephen Shen
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - James R Hilser
- Departments of Population & Public Health Sciences and Biochemistry & Molecular Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Jaana A Hartiala
- Departments of Population & Public Health Sciences and Biochemistry & Molecular Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Hooman Allayee
- Departments of Population & Public Health Sciences and Biochemistry & Molecular Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Richard Barbers
- Department of Clinical Medicine, Division of Pulmonary and Critical Care Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, Calif
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, Calif.
| |
Collapse
|
45
|
Zhou X, Sampath V, Nadeau KC. Effect of air pollution on asthma. Ann Allergy Asthma Immunol 2024; 132:426-432. [PMID: 38253122 PMCID: PMC10990824 DOI: 10.1016/j.anai.2024.01.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 01/16/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024]
Abstract
Asthma is a chronic inflammatory airway disease characterized by respiratory symptoms, variable airflow obstruction, bronchial hyperresponsiveness, and airway inflammation. Exposure to air pollution has been linked to an increased risk of asthma development and exacerbation. This review aims to comprehensively summarize recent data on the impact of air pollution on asthma development and exacerbation. Specifically, we reviewed the effects of air pollution on the pathogenic pathways of asthma, including type 2 and non-type 2 inflammatory responses, and airway epithelial barrier dysfunction. Air pollution promotes the release of epithelial cytokines, driving TH2 responses, and induces oxidative stress and the production of proinflammatory cytokines. The enhanced type 2 inflammation, furthered by air pollution-induced dysfunction of the airway epithelial barrier, may be associated with the exacerbation of asthma. Disruption of the TH17/regulatory T cell balance by air pollutants is also related to asthma exacerbation. As the effects of air pollution exposure may accumulate over time, with potentially stronger impacts in the development of asthma during certain sensitive life periods, we also reviewed the effects of air pollution on asthma across the lifespan. Future research is needed to better characterize the sensitive period contributing to the development of air pollution-induced asthma and to map air pollution-associated epigenetic biomarkers contributing to the epigenetic ages onto asthma-related genes.
Collapse
Affiliation(s)
- Xiaoying Zhou
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Vanitha Sampath
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.
| |
Collapse
|
46
|
Chiesa Fuxench ZC, Mitra N, Del Pozo D, Hoffstad O, Shin DB, Margolis DJ. Risk of atopic dermatitis and the atopic march paradigm in children of mothers with atopic illnesses: A birth cohort study from the United Kingdom. J Am Acad Dermatol 2024; 90:561-568. [PMID: 37984723 PMCID: PMC10922528 DOI: 10.1016/j.jaad.2023.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/13/2023] [Accepted: 11/01/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND Atopic dermatitis (AD) is thought to precede the onset of other allergic illness (OAI) in a temporal progression (ie, atopic march), yet the timing and progression has been questioned. It is also unclear how parental allergic illness impacts the development of these illnesses in offspring. OBJECTIVE (1) Explore risk of incident AD and (2) timing of allergic disease onset in children of mothers with AD compared with mothers without AD from the United Kingdom. METHODS We created a birth cohort of mother-child pairs using IQVIA Medical Research Data database and developed Cox proportional models to examine the above associations (hazard ratio, HR [95% confidence interval, CI]). RESULTS Among 1,224,243 child-mother pairs, mean child (standard deviation) follow-up time was 10.8 (8.3) years and 50.1% were males (N = 600,905). Children were 59% (HR = 1.59 [1.57, 1.60]) more likely to have AD if their mothers had AD compared with no AD with mean age of first AD diagnosis at 3.3 (4.8) years. Most children with any diagnosis of AD present with AD first (91.0%); however, in those with asthma, only 67.8% developed AD first. CONCLUSION Children born to mothers with AD are more prone to develop AD and some develop OAI first, suggesting that not all follow the same sequential pathway.
Collapse
Affiliation(s)
- Zelma C Chiesa Fuxench
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nandita Mitra
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Ole Hoffstad
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Daniel B Shin
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David J Margolis
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
47
|
Liu DS, Wang XS, Zhong XH, Cao H, Zhang F. Sexual dimorphism in the gut microbiota and sexual dimorphism in chronic diseases: Association or causation? J Steroid Biochem Mol Biol 2024; 237:106451. [PMID: 38154505 DOI: 10.1016/j.jsbmb.2023.106451] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/31/2023] [Accepted: 12/24/2023] [Indexed: 12/30/2023]
Abstract
Understanding the sexual dimorphism in diseases is essential to investigate the pathogenesis of some chronic diseases (e.g., autoimmune diseases, etc). The gut microbiota has been found to show a notable impact on the pathology of several chronic diseases in recent years. Intriguingly, the composition of the gut microbiota varies between sexes. Here, we reviewed 'facts and fiction' regarding sexual dimorphism in chronic diseases and sexual dimorphism in the gut microbiota respectively. The association and causative relationship between them aiming to elucidate the pathological mechanisms of sexual dimorphism in chronic diseases were further explored. The development of gender-special food products based on the sexual dimorphism in the gut microbiota were recommended, which would be beneficial to facilitating the personalized treatment.
Collapse
Affiliation(s)
- Dong-Song Liu
- Affiliated Hospital of Jiangnan University, Wuxi, China; Nantong University, Nantong, China
| | - Xue-Song Wang
- Affiliated Hospital of Jiangnan University, Wuxi, China; Nantong University, Nantong, China; Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xiao-Hui Zhong
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Hong Cao
- Affiliated Hospital of Jiangnan University, Wuxi, China; Nantong University, Nantong, China; Wuxi School of Medicine, Jiangnan University, Wuxi, China.
| | - Feng Zhang
- Affiliated Hospital of Jiangnan University, Wuxi, China; Wuxi School of Medicine, Jiangnan University, Wuxi, China.
| |
Collapse
|
48
|
Yue M, Tao S, Gaietto K, Chen W. Omics approaches in asthma research: Challenges and opportunities. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:1-9. [PMID: 39170962 PMCID: PMC11332849 DOI: 10.1016/j.pccm.2024.02.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Indexed: 08/23/2024]
Abstract
Asthma, a chronic respiratory disease with a global prevalence of approximately 300 million individuals, presents a significant societal and economic burden. This multifaceted syndrome exhibits diverse clinical phenotypes and pathogenic endotypes influenced by various factors. The advent of omics technologies has revolutionized asthma research by delving into the molecular foundation of the disease to unravel its underlying mechanisms. Omics technologies are employed to systematically screen for potential biomarkers, encompassing genes, transcripts, methylation sites, proteins, and even the microbiome components. This review provides an insightful overview of omics applications in asthma research, with a special emphasis on genetics, transcriptomics, epigenomics, and the microbiome. We explore the cutting-edge methods, discoveries, challenges, and potential future directions in the realm of asthma omics research. By integrating multi-omics and non-omics data through advanced statistical techniques, we aspire to advance precision medicine in asthma, guiding diagnosis, risk assessment, and personalized treatment strategies for this heterogeneous condition.
Collapse
Affiliation(s)
- Molin Yue
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Shiyue Tao
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Kristina Gaietto
- Division of Pediatric Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Wei Chen
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15224, USA
- Division of Pediatric Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15224, USA
| |
Collapse
|
49
|
Smilnak GJ, Lee Y, Chattopadhyay A, Wyss AB, White JD, Sikdar S, Jin J, Grant AJ, Motsinger-Reif AA, Li JL, Lee M, Yu B, London SJ. Plasma protein signatures of adult asthma. Allergy 2024; 79:643-655. [PMID: 38263798 PMCID: PMC10994188 DOI: 10.1111/all.16000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/08/2023] [Accepted: 12/04/2023] [Indexed: 01/25/2024]
Abstract
BACKGROUND Adult asthma is complex and incompletely understood. Plasma proteomics is an evolving technique that can both generate biomarkers and provide insights into disease mechanisms. We aimed to identify plasma proteomic signatures of adult asthma. METHODS Protein abundance in plasma was measured in individuals from the Agricultural Lung Health Study (ALHS) (761 asthma, 1095 non-case) and the Atherosclerosis Risk in Communities study (470 asthma, 10,669 non-case) using the SOMAScan 5K array. Associations with asthma were estimated using covariate adjusted logistic regression and meta-analyzed using inverse-variance weighting. Additionally, in ALHS, we examined phenotypes based on both asthma and seroatopy (asthma with atopy (n = 207), asthma without atopy (n = 554), atopy without asthma (n = 147), compared to neither (n = 948)). RESULTS Meta-analysis of 4860 proteins identified 115 significantly (FDR<0.05) associated with asthma. Multiple signaling pathways related to airway inflammation and pulmonary injury were enriched (FDR<0.05) among these proteins. A proteomic score generated using machine learning provided predictive value for asthma (AUC = 0.77, 95% CI = 0.75-0.79 in training set; AUC = 0.72, 95% CI = 0.69-0.75 in validation set). Twenty proteins are targeted by approved or investigational drugs for asthma or other conditions, suggesting potential drug repurposing. The combined asthma-atopy phenotype showed significant associations with 20 proteins, including five not identified in the overall asthma analysis. CONCLUSION This first large-scale proteomics study identified over 100 plasma proteins associated with current asthma in adults. In addition to validating previous associations, we identified many novel proteins that could inform development of diagnostic biomarkers and therapeutic targets in asthma management.
Collapse
Affiliation(s)
- Gordon J. Smilnak
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Yura Lee
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Abhijnan Chattopadhyay
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Annah B. Wyss
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Julie D. White
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
- GenOmics and Translational Research Center, Biostatistics and Epidemiology Division, RTI International, Research Triangle Park, NC, USA
| | - Sinjini Sikdar
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
- Department of Mathematics and Statistics, Old Dominion University, Norfolk, VA, USA
| | | | - Andrew J. Grant
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Alison A. Motsinger-Reif
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Jian-Liang Li
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Mikyeong Lee
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Stephanie J. London
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| |
Collapse
|
50
|
Kulshreshtha A, Bhatnagar S. Structural effect of the H992D/H418D mutation of angiotensin-converting enzyme in the Indian population: implications for health and disease. J Biomol Struct Dyn 2024:1-18. [PMID: 38411559 DOI: 10.1080/07391102.2024.2321246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 02/14/2024] [Indexed: 02/28/2024]
Abstract
The Non synonymous SNPs (nsSNPs) of the renin-angiotensin-system (RAS) pathway, unique to the Indian population were investigated in view of its importance as an endocrine system. nsSNPs of the RAS pathway genes were mined from the IndiGenome database. Damaging nsSNPs were predicted using SIFT, PredictSNP, SNP and GO, Snap2 and Protein Variation Effect Analyzer. Loss of function was predicted based on protein stability change using I mutant, PremPS and CONSURF. The structural impact of the nsSNPs was predicted using HOPE and Missense3d followed by modeling, refinement, and energy minimization. Molecular Dynamics studies were carried out using Gromacsv2021.1. 23 Indian nsSNPs of the RAS pathway genes were selected for structural analysis and 8 were predicted to be damaging. Further sequence analysis showed that HEMGH zinc binding motif changes to HEMGD in somatic ACE-C domain (sACE-C) H992D and Testis ACE (tACE) H418D resulted in loss of zinc coordination, which is essential for enzymatic activity in this metalloprotease. There was a loss of internal interactions around the zinc coordination residues in the protein structural network. This was also confirmed by Principal Component Analysis, Free Energy Landscape and residue contact maps. Both mutations lead to broadening of the AngI binding cavity. The H992D mutation in sACE-C is likely to be favorable for cardiovascular health, but may lead to renal abnormalities with secondary impact on the heart. H418D in tACE is potentially associated with male infertility.
Collapse
Affiliation(s)
- Akanksha Kulshreshtha
- Computational and Structural Biology Laboratory, Department of Biological Sciences and Engineering, Netaji Subhas University of Technology, Dwarka, New Delhi, India
| | - Sonika Bhatnagar
- Computational and Structural Biology Laboratory, Department of Biological Sciences and Engineering, Netaji Subhas University of Technology, Dwarka, New Delhi, India
| |
Collapse
|