1
|
Wang Y, Tzeng JY, Huang Y, Maguire R, Hoyo C, Allen TK. Duration of exposure to epidural anesthesia at delivery, DNA methylation in umbilical cord blood and their association with offspring asthma in Non-Hispanic Black women. ENVIRONMENTAL EPIGENETICS 2022; 9:dvac026. [PMID: 36694712 PMCID: PMC9854336 DOI: 10.1093/eep/dvac026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/16/2022] [Accepted: 12/12/2022] [Indexed: 06/17/2023]
Abstract
Epidural anesthesia is an effective pain relief modality, widely used for labor analgesia. Childhood asthma is one of the commonest chronic medical illnesses in the USA which places a significant burden on the health-care system. We recently demonstrated a negative association between the duration of epidural anesthesia and the development of childhood asthma; however, the underlying molecular mechanisms still remain unclear. In this study of 127 mother-child pairs comprised of 75 Non-Hispanic Black (NHB) and 52 Non-Hispanic White (NHW) from the Newborn Epigenetic Study, we tested the hypothesis that umbilical cord blood DNA methylation mediates the association between the duration of exposure to epidural anesthesia at delivery and the development of childhood asthma and whether this differed by race/ethnicity. In the mother-child pairs of NHB ancestry, the duration of exposure to epidural anesthesia was associated with a marginally lower risk of asthma (odds ratio = 0.88, 95% confidence interval = 0.76-1.01) for each 1-h increase in exposure to epidural anesthesia. Of the 20 CpGs in the NHB population showing the strongest mediation effect, 50% demonstrated an average mediation proportion of 52%, with directional consistency of direct and indirect effects. These top 20 CpGs mapped to 21 genes enriched for pathways engaged in antigen processing, antigen presentation, protein ubiquitination and regulatory networks related to the Major Histocompatibility Complex (MHC) class I complex and Nuclear Factor Kappa-B (NFkB) complex. Our findings suggest that DNA methylation in immune-related pathways contributes to the effects of the duration of exposure to epidural anesthesia on childhood asthma risk in NHB offspring.
Collapse
Affiliation(s)
- Yaxu Wang
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27607, USA
| | - Jung-Ying Tzeng
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27607, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695-7633, USA
- Department of Statistics, North Carolina State University, Raleigh, NC 27607, USA
| | - Yueyang Huang
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27607, USA
| | - Rachel Maguire
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695-7633, USA
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Cathrine Hoyo
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Terrence K Allen
- Department of Anesthesiology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
2
|
Wjst M. Exome variants associated with asthma and allergy. Sci Rep 2022; 12:21028. [PMID: 36470944 PMCID: PMC9722654 DOI: 10.1038/s41598-022-24960-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
The mutational spectrum of asthma and allergy associated genes is not known although recent biobank based exome sequencing studies included these traits. We therefore conducted a secondary analysis of exome data from 281,104 UK Biobank samples for association of mostly rare variants with asthma, allergic rhinitis and atopic dermatitis. Variants of interest (VOI) were tabulated, shared genes annotated and compared to earlier genome-wide SNP association studies (GWAS), whole genome sequencing, exome and bisulfit sequencing studies. 354 VOI were significantly associated with asthma, allergic rhinitis and atopic dermatitis. They cluster mainly in two large regions on chromosome 6 and 17. After exclusion of the variants associated with atopic dermatitis and redundant variants, 321 unique VOI remain in 122 unique genes. 30 genes are shared among the 87 genes with increased and the 65 genes with decreased risk for allergic disease. 85% of genes identified earlier by common GWAS SNPs are not replicated here. Most identified genes are located in interferon ɣ and IL33 signaling pathway. These genes include already known but also new pharmacological targets, including the IL33 receptor ST2/IL1RL1, as well as TLR1, ALOX15, GSDMA, BTNL2, IL13 and IKZF3. Future pharmacological studies will need to included these VOI for stratification of the study population paving the way to individualized treatment.
Collapse
Affiliation(s)
- Matthias Wjst
- Institute of Lung Health and Immunity (LHI), Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstädter Landstr. 1, 85764, Neuherberg, München, Germany. .,Institut für KI und Informatik in der Medizin, Lehrstuhl für Medizinische Informatik, Klinikum Rechts der Isar, Grillparzerstr. 18, 81675, München, Germany.
| |
Collapse
|
3
|
Kaur D, Chachi L, Gomez E, Sylvius N, Singh SR, Ramsheh MY, Saunders R, Brightling CE. ST2 expression and release by the bronchial epithelium is downregulated in asthma. Allergy 2020; 75:3184-3194. [PMID: 32516479 DOI: 10.1111/all.14436] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 05/01/2020] [Accepted: 05/04/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND The airway epithelium plays an important role in wound repair, host defense and is involved in the immunopathogenesis of asthma. Genome wide association studies have described associations between ST2/Interleukin (IL)-33 genes in asthma, but its role in bronchial epithelium is unclear. METHODS ST2 expression was examined in subjects with asthma and healthy controls in bronchial epithelium from biopsies (n = 27 versus n = 9) and brushings (n = 34 versus n = 20) by immunohistochemistry and RNA-Seq. In human primary bronchial epithelial cells ST2 mRNA and protein expression were assessed by qPCR, flow cytometry, Western blotting, and immunofluorescence. IL-33 function in epithelial cells was examined by intracellular calcium measurements, wound healing assays, and synthetic activation by gene array and ELISA. RESULTS Bronchial epithelial ST2 protein expression was significantly decreased in biopsies in subjects with asthma compared to healthy controls (P = .039). IL1RL1 gene expression in bronchial brushes was not different between health and disease. In vitro primary bronchial epithelial cells expressed ST2 and IL-33 stimulation led to an increase in intracellular calcium, altered gene expression, but had no effect upon wound repair. Epithelial cells released sST2 spontaneously, which was reduced following stimulation with TNFα or poly-IC. Stimulation by TNFα or poly-IC did not affect the total ST2 expression by epithelial cell whereas surface ST2 decreased in response to TNFα, but not poly-IC. CONCLUSION In asthma, bronchial epithelium protein expression of ST2 is decreased. Our in vitro findings suggest that this decrease might be a consequence of the pro-inflammatory environment in asthma or in response to viral infection.
Collapse
Affiliation(s)
- Davinder Kaur
- Institute for Lung Health Department of Respiratory Sciences University of Leicester Leicester UK
| | - Latifa Chachi
- Institute for Lung Health Department of Respiratory Sciences University of Leicester Leicester UK
| | - Edith Gomez
- Institute for Lung Health Department of Respiratory Sciences University of Leicester Leicester UK
| | - Nicolas Sylvius
- Genomic Core Facility Department of Genetics University of Leicester Leicester UK
| | - Shailendra R. Singh
- Institute for Lung Health Department of Respiratory Sciences University of Leicester Leicester UK
| | - Mohammadali Y. Ramsheh
- Institute for Lung Health Department of Respiratory Sciences University of Leicester Leicester UK
| | - Ruth Saunders
- Institute for Lung Health Department of Respiratory Sciences University of Leicester Leicester UK
| | | |
Collapse
|
4
|
Reese SE, Xu CJ, den Dekker HT, Lee MK, Sikdar S, Ruiz-Arenas C, Merid SK, Rezwan FI, Page CM, Ullemar V, Melton PE, Oh SS, Yang IV, Burrows K, Söderhäll C, Jima DD, Gao L, Arathimos R, Küpers LK, Wielscher M, Rzehak P, Lahti J, Laprise C, Madore AM, Ward J, Bennett BD, Wang T, Bell DA, Vonk JM, Håberg SE, Zhao S, Karlsson R, Hollams E, Hu D, Richards AJ, Bergström A, Sharp GC, Felix JF, Bustamante M, Gruzieva O, Maguire RL, Gilliland F, Baïz N, Nohr EA, Corpeleijn E, Sebert S, Karmaus W, Grote V, Kajantie E, Magnus MC, Örtqvist AK, Eng C, Liu AH, Kull I, Jaddoe VWV, Sunyer J, Kere J, Hoyo C, Annesi-Maesano I, Arshad SH, Koletzko B, Brunekreef B, Binder EB, Räikkönen K, Reischl E, Holloway JW, Jarvelin MR, Snieder H, Kazmi N, Breton CV, Murphy SK, Pershagen G, Anto JM, Relton CL, Schwartz DA, Burchard EG, Huang RC, Nystad W, Almqvist C, Henderson AJ, Melén E, Duijts L, Koppelman GH, London SJ. Epigenome-wide meta-analysis of DNA methylation and childhood asthma. J Allergy Clin Immunol 2019; 143:2062-2074. [PMID: 30579849 PMCID: PMC6556405 DOI: 10.1016/j.jaci.2018.11.043] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 10/01/2018] [Accepted: 11/16/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Epigenetic mechanisms, including methylation, can contribute to childhood asthma. Identifying DNA methylation profiles in asthmatic patients can inform disease pathogenesis. OBJECTIVE We sought to identify differential DNA methylation in newborns and children related to childhood asthma. METHODS Within the Pregnancy And Childhood Epigenetics consortium, we performed epigenome-wide meta-analyses of school-age asthma in relation to CpG methylation (Illumina450K) in blood measured either in newborns, in prospective analyses, or cross-sectionally in school-aged children. We also identified differentially methylated regions. RESULTS In newborns (8 cohorts, 668 cases), 9 CpGs (and 35 regions) were differentially methylated (epigenome-wide significance, false discovery rate < 0.05) in relation to asthma development. In a cross-sectional meta-analysis of asthma and methylation in children (9 cohorts, 631 cases), we identified 179 CpGs (false discovery rate < 0.05) and 36 differentially methylated regions. In replication studies of methylation in other tissues, most of the 179 CpGs discovered in blood replicated, despite smaller sample sizes, in studies of nasal respiratory epithelium or eosinophils. Pathway analyses highlighted enrichment for asthma-relevant immune processes and overlap in pathways enriched both in newborns and children. Gene expression correlated with methylation at most loci. Functional annotation supports a regulatory effect on gene expression at many asthma-associated CpGs. Several implicated genes are targets for approved or experimental drugs, including IL5RA and KCNH2. CONCLUSION Novel loci differentially methylated in newborns represent potential biomarkers of risk of asthma by school age. Cross-sectional associations in children can reflect both risk for and effects of disease. Asthma-related differential methylation in blood in children was substantially replicated in eosinophils and respiratory epithelium.
Collapse
Affiliation(s)
- Sarah E Reese
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC
| | - Cheng-Jian Xu
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Herman T den Dekker
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands; Department of Pediatrics, Division of Respiratory Medicine and Allergology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands; Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Mi Kyeong Lee
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC
| | - Sinjini Sikdar
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC
| | - Carlos Ruiz-Arenas
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain; ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Simon K Merid
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Faisal I Rezwan
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Christian M Page
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway; Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| | - Vilhelmina Ullemar
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Phillip E Melton
- Curtin/UWA Centre for Genetic Origins of Health and Disease, Faculty of Health and Medical Sciences, University of Western Australia, Crawley, Australia; School of Pharmacy and Biomedical Sciences, Curtin University, Bentley, Australia
| | - Sam S Oh
- Department of Medicine, University of California San Francisco, San Francisco, Calif
| | - Ivana V Yang
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Kimberley Burrows
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom; Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - Cilla Söderhäll
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden; Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Dereje D Jima
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC
| | - Lu Gao
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Ryan Arathimos
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom; School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Leanne K Küpers
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom; Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom; Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Matthias Wielscher
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment & Health, School of Public Health, Imperial College London, London, United Kingdom
| | - Peter Rzehak
- Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, Ludwig-Maximilians Universität München (LMU), Munich, Germany
| | - Jari Lahti
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland; Helsinki Collegium for Advanced Studies, University of Helsinki, Helsinki, Finland
| | - Catherine Laprise
- Centre intégré universitaire de santé et de services sociaux du Saguenay, Saguenay, Quebec, Canada; Département des sciences fondamentales, Université du Québec à Chicoutimi, Saguenay, Quebec, Canada
| | - Anne-Marie Madore
- Département des sciences fondamentales, Université du Québec à Chicoutimi, Saguenay, Quebec, Canada
| | - James Ward
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC
| | - Brian D Bennett
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC
| | - Tianyuan Wang
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC
| | - Douglas A Bell
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC
| | - Judith M Vonk
- GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Siri E Håberg
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Shanshan Zhao
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC
| | - Robert Karlsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Elysia Hollams
- Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Donglei Hu
- Department of Medicine, University of California San Francisco, San Francisco, Calif
| | - Adam J Richards
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Anna Bergström
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Centre for Occupational and Environmental Medicine, Stockholm County Council, Stockholm, Sweden
| | - Gemma C Sharp
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom; Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom; Bristol Dental School, University of Bristol, Bristol, United Kingdom
| | - Janine F Felix
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands; Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands; Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Mariona Bustamante
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain; ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; Center for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Olena Gruzieva
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Centre for Occupational and Environmental Medicine, Stockholm County Council, Stockholm, Sweden
| | - Rachel L Maguire
- Department of Biological Sciences, North Carolina State University, Raleigh, NC; Department of Community and Family Medicine, Duke University Medical Center, Durham, NC
| | - Frank Gilliland
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Nour Baïz
- Epidemiology of Allergic and Respiratory Diseases Department, IPLESP, INSERM and UPMC Sorbonne Université, Paris, France
| | - Ellen A Nohr
- Research Unit for Gynaecology and Obstetrics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Eva Corpeleijn
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sylvain Sebert
- Biocenter Oulu, University of Oulu, Oulu, Finland; Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland; Department of Genomics of Complex Diseases, School of Public Health, Imperial College London, London, United Kingdom
| | - Wilfried Karmaus
- Division of Epidemiology, Biostatistics and Environmental Health, School of Public Health, University of Memphis, Memphis, Tenn
| | - Veit Grote
- Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, Ludwig-Maximilians Universität München (LMU), Munich, Germany
| | - Eero Kajantie
- Chronic Disease Prevention Unit, National Institute for Health and Welfare, Helsinki, Finland; Department of Obstetrics and Gynaecology, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Hospital for Children and Adolescents, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | - Maria C Magnus
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom; Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom; Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Anne K Örtqvist
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Celeste Eng
- Department of Medicine, University of California San Francisco, San Francisco, Calif
| | | | - Inger Kull
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden; Sachs' Children's Hospital, Södersjukhuset, Stockholm, Sweden
| | - Vincent W V Jaddoe
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands; Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands; Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jordi Sunyer
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain; ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden; Department of Medical and Molecular Genetics, King's College London, London, United Kingdom
| | - Cathrine Hoyo
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC; Department of Biological Sciences, North Carolina State University, Raleigh, NC
| | - Isabella Annesi-Maesano
- Epidemiology of Allergic and Respiratory Diseases Department, IPLESP, INSERM and UPMC Sorbonne Université, Paris, France
| | - Syed Hasan Arshad
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom; David Hide Asthma and Allergy Research Centre, Isle of Wight, United Kingdom
| | - Berthold Koletzko
- Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, Ludwig-Maximilians Universität München (LMU), Munich, Germany
| | - Bert Brunekreef
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands; Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Elisabeth B Binder
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, Ga; Max-Planck-Institute of Psychiatry, Munich, Germany
| | - Katri Räikkönen
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| | - Eva Reischl
- Research Unit of Molecular Epidemiology, Institute of Epidemiology II, Helmholtz Zentrum Muenchen, Munich, Germany
| | - John W Holloway
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom; Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Marjo-Riitta Jarvelin
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment & Health, School of Public Health, Imperial College London, London, United Kingdom; Biocenter Oulu, University of Oulu, Oulu, Finland; Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Nabila Kazmi
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom; School of Social and Community Medicine, University of Bristol, Bristol, United Kingdom
| | - Carrie V Breton
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, Calif
| | - Susan K Murphy
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC; Nicholas School of the Environment, Duke University, Durham, NC
| | - Göran Pershagen
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Centre for Occupational and Environmental Medicine, Stockholm County Council, Stockholm, Sweden
| | - Josep Maria Anto
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain; ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Caroline L Relton
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom; Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - David A Schwartz
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colo
| | - Esteban G Burchard
- Department of Medicine, University of California San Francisco, San Francisco, Calif; Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, Calif
| | - Rae-Chi Huang
- Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Wenche Nystad
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Catarina Almqvist
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Pediatric Allergy and Pulmonology Unit at Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - A John Henderson
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Erik Melén
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Sachs' Children's Hospital, Södersjukhuset, Stockholm, Sweden
| | - Liesbeth Duijts
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands; Department of Pediatrics, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Gerard H Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands; GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Stephanie J London
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC.
| |
Collapse
|
5
|
Sturek J, Noth I. Wandering out of the GWAS wilderness: a new pathway paradigm for complex disease genetics. Thorax 2019; 74:215-216. [PMID: 30661023 DOI: 10.1136/thoraxjnl-2018-212511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2019] [Indexed: 11/04/2022]
Affiliation(s)
- Jeffrey Sturek
- Division of Pulmonary and Critical Care, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Imre Noth
- Division of Pulmonary and Critical Care, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
6
|
Dizier MH, Margaritte-Jeannin P, Pain L, Sarnowski C, Brossard M, Mohamdi H, Lavielle N, Babron MCC, Just J, Lathrop M, Laprise C, Bouzigon E, Demenais F, Nadif R. Interactive effect between ATPase-related genes and early-life tobacco smoke exposure on bronchial hyper-responsiveness detected in asthma-ascertained families. Thorax 2018; 74:254-260. [PMID: 30282721 DOI: 10.1136/thoraxjnl-2018-211797] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 06/21/2018] [Accepted: 08/20/2018] [Indexed: 01/29/2023]
Abstract
BACKGROUND A positional cloning study of bronchial hyper-responsiveness (BHR) at the 17p11 locus in the French Epidemiological study on the Genetics and Environment of Asthma (EGEA) families showed significant interaction between early-life environmental tobacco smoke (ETS) exposure and genetic variants located in DNAH9. This gene encodes the heavy chain subunit of axonemal dynein, which is involved with ATP in the motile cilia function.Our goal was to identify genetic variants at other genes interacting with ETS in BHR by investigating all genes belonging to the 'ATP-binding' and 'ATPase activity' pathways which include DNAH9, are targets of cigarette smoke and play a crucial role in the airway inflammation. METHODS Family-based interaction tests between ETS-exposed and unexposed BHR siblings were conducted in 388 EGEA families. Twenty single-nucleotide polymorphisms (SNP) showing interaction signals (p≤5.10-3) were tested in the 253 Saguenay-Lac-Saint-Jean (SLSJ) families. RESULTS One of these SNPs was significantly replicated for interaction with ETS in SLSJ families (p=0.003). Another SNP reached the significance threshold after correction for multiple testing in the combined analysis of the two samples (p=10-5). Results were confirmed using both a robust log-linear test and a gene-based interaction test. CONCLUSION The SNPs showing interaction with ETS belong to the ATP8A1 and ABCA1 genes, which play a role in the maintenance of asymmetry and homeostasis of lung membrane lipids.
Collapse
Affiliation(s)
- Marie-Hélène Dizier
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Patricia Margaritte-Jeannin
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Lucile Pain
- Département des Sciences Fondamentales, Université du Québec, Chicoutimi, Quebec, Canada
| | - Chloé Sarnowski
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Myriam Brossard
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Hamida Mohamdi
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Nolwenn Lavielle
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Marie-Claude C Babron
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Jocelyne Just
- Service d'Allergologie Pédiatrique, Centre de l'Asthme et des Allergies, Hôpital d'Enfants Armand-Trousseau (APHP), UPMC Paris 06, Paris, France
| | - Mark Lathrop
- Department of Human Genetics, McGill University and Genome Quebec's Innovation Centre, Montréal, Québec, Canada
| | - Catherine Laprise
- Département des Sciences Fondamentales, Université du Québec, Chicoutimi, Quebec, Canada
| | - Emmanuelle Bouzigon
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Florence Demenais
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Université Paris Diderot, Paris, France
| | - Rachel Nadif
- Aging and Chronic Diseases-Epidemiological and Public Health Approaches (VIMA), Inserm, U1168, Villejuif, France.,UMR-S 1168, Université de Versailles Saint-Quentin-en-Yvelines, Paris, France
| |
Collapse
|
7
|
Abstract
The extracellular forms of the IL-1 cytokines are active through binding to specific receptors on the surface of target cells. IL-1 ligands bind to the extracellular portion of their ligand-binding receptor chain. For signaling to take place, a non-binding accessory chain is recruited into a heterotrimeric complex. The intracellular approximation of the Toll-IL-1-receptor (TIR) domains of the 2 receptor chains is the event that initiates signaling. The family of IL-1 receptors (IL-1R) includes 10 structurally related members, and the distantly related soluble protein IL-18BP that acts as inhibitor of the cytokine IL-18. Over the years the receptors of the IL-1 family have been known with many different names, with significant confusion. Thus, we will use here a recently proposed unifying nomenclature. The family includes several ligand-binding chains (IL-1R1, IL-1R2, IL-1R4, IL-1R5, and IL-1R6), 2 types of accessory chains (IL-1R3, IL-1R7), molecules that act as inhibitors of signaling (IL-1R2, IL-1R8, IL-18BP), and 2 orphan receptors (IL-1R9, IL-1R10). In this review, we will examine how the receptors of the IL-1 family regulate the inflammatory and anti-inflammatory functions of the IL-1 cytokines and are, more at large, involved in modulating defensive and pathological innate immunity and inflammation. Regulation of the IL-1/IL-1R system in the brain will be also described, as an example of the peculiarities of organ-specific modulation of inflammation.
Collapse
Affiliation(s)
- Diana Boraschi
- Institute of Protein Biochemistry, National Research Council, Naples, Italy
| | - Paola Italiani
- Institute of Protein Biochemistry, National Research Council, Naples, Italy
| | - Sabrina Weil
- Immunology FB08, Justus-Liebig-Universitat Giessen, Giessen, Germany
| | - Michael U Martin
- Immunology FB08, Justus-Liebig-Universitat Giessen, Giessen, Germany
| |
Collapse
|
8
|
Cayrol C, Girard JP. Interleukin-33 (IL-33): A nuclear cytokine from the IL-1 family. Immunol Rev 2017; 281:154-168. [DOI: 10.1111/imr.12619] [Citation(s) in RCA: 601] [Impact Index Per Article: 75.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Corinne Cayrol
- Institut de Pharmacologie et de Biologie Structurale; IPBS; Université de Toulouse; CNRS; UPS; Toulouse France
| | - Jean-Philippe Girard
- Institut de Pharmacologie et de Biologie Structurale; IPBS; Université de Toulouse; CNRS; UPS; Toulouse France
| |
Collapse
|
9
|
Lodge CJ, Bråbäck L, Lowe AJ, Dharmage SC, Olsson D, Forsberg B. Grandmaternal smoking increases asthma risk in grandchildren: A nationwide Swedish cohort. Clin Exp Allergy 2017; 48:167-174. [PMID: 28925522 DOI: 10.1111/cea.13031] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 07/31/2017] [Accepted: 08/28/2017] [Indexed: 11/28/2022]
Abstract
BACKGROUND There is growing interest in exposures prior to conception as possible risk factors for offspring asthma. Although partially supported by evidence from limited human studies, current evidence is inconsistent and based on recall of exposure status. OBJECTIVE We aimed to investigate grandmaternal smoking during pregnancy and the risk of asthma in grandchildren using prospectively collected population-based data. METHODS Information on grandmaternal and maternal smoking during pregnancy and grandchild use of asthma medications was collected from national Swedish registries. Associations between grandmaternal smoking during pregnancy (10-12 weeks) and asthma medication use in grandchildren were investigated using generalized estimating equations. Ages at which asthma medications were prescribed classified childhood asthma into never, early transient (0-3 years), late onset (3-6 years) and early persistent (0-3 and 3-6 years) phenotypes. RESULTS From 1982 to 1986, 44 583 grandmothers gave birth to 46 197 mothers, who gave birth to 66 271 grandchildren (born 1996-2010). Children aged 1-6 years had an increased asthma risk if their grandmothers had smoked during pregnancy, with a higher risk for more exposure (10+ cigs/d; adjusted OR 1.23; 1.17, 1.30). Maternal smoking did not modify this relationship. CONCLUSIONS & CLINICAL RELEVANCE Children had an increased risk of asthma in the first 6 years of life if their grandmothers smoked during early pregnancy, independent of maternal smoking. Importantly, this exhibited a dose-response relationship and was associated with a persistent childhood asthma phenotype. These findings support possible epigenetic transmission of risk from environmental exposures in previous generations.
Collapse
Affiliation(s)
- C J Lodge
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, School of Population and Global Health, University of Melbourne, Melbourne, Vic., Australia.,Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Melbourne, Vic., Australia.,Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - L Bråbäck
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - A J Lowe
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, School of Population and Global Health, University of Melbourne, Melbourne, Vic., Australia.,Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Melbourne, Vic., Australia.,Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - S C Dharmage
- Allergy and Lung Health Unit, Centre for Epidemiology and Biostatistics, School of Population and Global Health, University of Melbourne, Melbourne, Vic., Australia.,Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Melbourne, Vic., Australia
| | - D Olsson
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - B Forsberg
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| |
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW Food allergies (FAs) are a growing epidemic in western countries with poorly defined etiology. Defined as an adverse immune response to common food allergens, FAs present heterogeneously as a single- or multi-organ response that ranges in severity from localized hives and angioedema to systemic anaphylaxis. RECENT FINDINGS Current research focusing on epithelial-derived cytokines contends that temporal regulation by these factors impact initial sensitization and persistence of FA responses upon repeated food allergen exposure. Mechanistic understanding of FA draws insight from a myriad of atopic conditions studied in humans and modeled in mice. In this review, we will highlight how epithelial-derived cytokines initiate and then potentiate FAs. We will also review existing evidence of the contribution of other atopic diseases to FA pathogenesis and whether FA symptoms overlap with other atopic diseases.
Collapse
|
11
|
Belgrave D, Henderson J, Simpson A, Buchan I, Bishop C, Custovic A. Disaggregating asthma: Big investigation versus big data. J Allergy Clin Immunol 2017; 139:400-407. [PMID: 27871876 PMCID: PMC5292995 DOI: 10.1016/j.jaci.2016.11.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/07/2016] [Accepted: 11/09/2016] [Indexed: 12/21/2022]
Abstract
We are facing a major challenge in bridging the gap between identifying subtypes of asthma to understand causal mechanisms and translating this knowledge into personalized prevention and management strategies. In recent years, "big data" has been sold as a panacea for generating hypotheses and driving new frontiers of health care; the idea that the data must and will speak for themselves is fast becoming a new dogma. One of the dangers of ready accessibility of health care data and computational tools for data analysis is that the process of data mining can become uncoupled from the scientific process of clinical interpretation, understanding the provenance of the data, and external validation. Although advances in computational methods can be valuable for using unexpected structure in data to generate hypotheses, there remains a need for testing hypotheses and interpreting results with scientific rigor. We argue for combining data- and hypothesis-driven methods in a careful synergy, and the importance of carefully characterized birth and patient cohorts with genetic, phenotypic, biological, and molecular data in this process cannot be overemphasized. The main challenge on the road ahead is to harness bigger health care data in ways that produce meaningful clinical interpretation and to translate this into better diagnoses and properly personalized prevention and treatment plans. There is a pressing need for cross-disciplinary research with an integrative approach to data science, whereby basic scientists, clinicians, data analysts, and epidemiologists work together to understand the heterogeneity of asthma.
Collapse
Affiliation(s)
| | - John Henderson
- School of Social and Community Medicine, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Angela Simpson
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Iain Buchan
- Health Informatics, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | | | - Adnan Custovic
- Department of Paediatrics, Imperial College, London, United Kingdom.
| |
Collapse
|
12
|
Affiliation(s)
- Surinder K Jindal
- Jindal Clinics, SCO 21, Dakshin Marg, Sector 20D, Near Guru Ravi Das Bhawan, Chandigarh 160 020, India
| |
Collapse
|
13
|
Moheimani F, Hsu ACY, Reid AT, Williams T, Kicic A, Stick SM, Hansbro PM, Wark PAB, Knight DA. The genetic and epigenetic landscapes of the epithelium in asthma. Respir Res 2016; 17:119. [PMID: 27658857 PMCID: PMC5034566 DOI: 10.1186/s12931-016-0434-4] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 09/17/2016] [Indexed: 12/24/2022] Open
Abstract
Asthma is a global health problem with increasing prevalence. The airway epithelium is the initial barrier against inhaled noxious agents or aeroallergens. In asthma, the airway epithelium suffers from structural and functional abnormalities and as such, is more susceptible to normally innocuous environmental stimuli. The epithelial structural and functional impairments are now recognised as a significant contributing factor to asthma pathogenesis. Both genetic and environmental risk factors play important roles in the development of asthma with an increasing number of genes associated with asthma susceptibility being expressed in airway epithelium. Epigenetic factors that regulate airway epithelial structure and function are also an attractive area for assessment of susceptibility to asthma. In this review we provide a comprehensive discussion on genetic factors; from using linkage designs and candidate gene association studies to genome-wide association studies and whole genome sequencing, and epigenetic factors; DNA methylation, histone modifications, and non-coding RNAs (especially microRNAs), in airway epithelial cells that are functionally associated with asthma pathogenesis. Our aims were to introduce potential predictors or therapeutic targets for asthma in airway epithelium. Overall, we found very small overlap in asthma susceptibility genes identified with different technologies. Some potential biomarkers are IRAKM, PCDH1, ORMDL3/GSDMB, IL-33, CDHR3 and CST1 in airway epithelial cells. Recent studies on epigenetic regulatory factors have further provided novel insights to the field, particularly their effect on regulation of some of the asthma susceptibility genes (e.g. methylation of ADAM33). Among the epigenetic regulatory mechanisms, microRNA networks have been shown to regulate a major portion of post-transcriptional gene regulation. Particularly, miR-19a may have some therapeutic potential.
Collapse
Affiliation(s)
- Fatemeh Moheimani
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, HMRI building, The University of Newcastle, Callaghan, NSW, 2308, Australia. .,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, New South Wales, Australia.
| | - Alan C-Y Hsu
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, HMRI building, The University of Newcastle, Callaghan, NSW, 2308, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, New South Wales, Australia
| | - Andrew T Reid
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, HMRI building, The University of Newcastle, Callaghan, NSW, 2308, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, New South Wales, Australia
| | - Teresa Williams
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, HMRI building, The University of Newcastle, Callaghan, NSW, 2308, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, New South Wales, Australia.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, Canada
| | - Anthony Kicic
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, 6009, Western Australia, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, 6001, Western Australia, Australia.,School of Paediatrics and Child Health, The University of Western Australia, Nedlands, 6009, Western Australia, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, 6009, Western Australia, Australia
| | - Stephen M Stick
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, 6009, Western Australia, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, 6001, Western Australia, Australia.,School of Paediatrics and Child Health, The University of Western Australia, Nedlands, 6009, Western Australia, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, 6009, Western Australia, Australia
| | - Philip M Hansbro
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, HMRI building, The University of Newcastle, Callaghan, NSW, 2308, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, New South Wales, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, New South Wales, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, New South Wales, Australia
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, HMRI building, The University of Newcastle, Callaghan, NSW, 2308, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, New South Wales, Australia.,Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, Canada
| |
Collapse
|
14
|
Cho SH, Min JY, Kim DY, Oh SS, Torgerson DR, Pino-Yanes M, Hu D, Sen S, Huntsman S, Eng C, Farber HJ, Rodriguez-Cintron W, Rodriguez-Santana JR, Serebrisky D, Thyne SM, Borrell LN, Williams LK, DuPont W, Seibold MA, Burchard EG, Avila PC, Kumar R. Association of a PAI-1 Gene Polymorphism and Early Life Infections with Asthma Risk, Exacerbations, and Reduced Lung Function. PLoS One 2016; 11:e0157848. [PMID: 27556405 PMCID: PMC4996454 DOI: 10.1371/journal.pone.0157848] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 06/06/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Plasminogen activator inhibitor-1 (PAI-1) is induced in airways by virus and may mediate asthmatic airway remodeling. We sought to evaluate if genetic variants and early life lower respiratory infections jointly affect asthma risk. METHODS We included Latino children, adolescents, and young adults aged 8-21 years (1736 subjects with physician-diagnosed asthma and 1747 healthy controls) from five U.S. centers and Puerto Rico after excluding subjects with incomplete clinical or genetic data. We evaluated the independent and joint effects of a PAI-1 gain of function polymorphism and bronchiolitis / Respiratory Syncytial Virus (RSV) or other lower respiratory infections (LRI) within the first 2 years of life on asthma risk, asthma exacerbations and lung function. RESULTS RSV infection (OR 9.9, 95%CI 4.9-20.2) and other LRI (OR 9.1, 95%CI 7.2-11.5) were independently associated with asthma, but PAI-1 genotype was not. There were joint effects on asthma risk for both genotype-RSV (OR 17.7, 95% CI 6.3-50.2) and genotype-LRI (OR 11.7, 95% CI 8.8-16.4). A joint effect of genotype-RSV resulted in a 3.1-fold increased risk for recurrent asthma hospitalizations. In genotype-respiratory infection joint effect analysis, FEV1% predicted and FEV1/FVC % predicted were further reduced in the genotype-LRI group (β -2.1, 95% CI -4.0 to -0.2; β -2.0, 95% CI -3.1 to -0.8 respectively). Similarly, lower FEV1% predicted was noted in genotype-RSV group (β -3.1, 95% CI -6.1 to -0.2) with a trend for lower FEV1/FVC % predicted. CONCLUSIONS A genetic variant of PAI-1 together with early life LRI such as RSV bronchiolitis is associated with an increased risk of asthma, morbidity, and reduced lung function in this Latino population.
Collapse
Affiliation(s)
- Seong H. Cho
- Division of Allergy-Immunology, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
- Division of Allergy-Immunology, Department of Internal Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Jin-Young Min
- Department of Otolaryngology, Northwestern University, Chicago, Illinois, United States of America
| | - Dong Young Kim
- Division of Allergy-Immunology, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Sam S. Oh
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Dara R. Torgerson
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Maria Pino-Yanes
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Donglei Hu
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Saunak Sen
- Division of Biostatistics, Department of Preventive Medicine, UTHSC, Memphis, Tennessee, United States of America
| | - Scott Huntsman
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Celeste Eng
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Harold J. Farber
- Department of Pediatrics, Section of Pulmonology, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, United States of America
| | | | | | - Denise Serebrisky
- Pediatric Pulmonary Division, Jacobi Medical Center, Bronx, New York, United States of America
| | - Shannon M. Thyne
- Department of Pediatrics, University of California, San Francisco, California, United States of America
| | - Luisa N. Borrell
- Department of Health Sciences, Lehman College, CUNY, New York, New York, United States of America
| | - L. Keoki Williams
- Department of Internal Medicine, Henry Ford Health System, Detroit, Michigan, United States of America
- Center for Health Policy and Health Services Research, Henry Ford Health System, Detroit, Michigan, United States of America
| | - William DuPont
- Department of Biostatistics, Vanderbilt University Medical School, Nashville, Tennessee, United States of America
| | - Max A. Seibold
- Center for Genes, Environment and Health, National Jewish Health, Denver, Colorado, United States of America
| | - Esteban G. Burchard
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Pedro C. Avila
- Division of Allergy-Immunology, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Rajesh Kumar
- Division of Allergy-Immunology, Department of Pediatrics, Northwestern University, Chicago, Illinois, United States of America
- The Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
15
|
Murk W, DeWan AT. Genome-wide search identifies a gene-gene interaction between 20p13 and 2q14 in asthma. BMC Genet 2016; 17:102. [PMID: 27387956 PMCID: PMC4936310 DOI: 10.1186/s12863-016-0376-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/20/2016] [Indexed: 12/11/2022] Open
Abstract
Background Many studies have attempted to identify gene-gene interactions affecting asthma susceptibility. However, these studies have typically used candidate gene approaches in limiting the genetic search space, and there have been few searches for gene-gene interactions on a genome-wide scale. We aimed to conduct a genome-wide gene-gene interaction study for asthma, using data from the GABRIEL Consortium. Results A two-stage study design was used, including a screening analysis (N = 1625 subjects) and a follow-up analysis (N = 5264 subjects). In the screening analysis, all pairwise interactions among 301,547 SNPs were evaluated, encompassing a total of 4.55 × 1010 interactions. Those with a screening interaction p-value < 10−5 were evaluated in the follow-up analysis. No interaction selected from the screening analysis met strict statistical significance in the follow-up (p-value < 1.45 × 10−7). However, the top-ranked interaction (rs910652 [20p13] × rs11684871 [2q14]) in the follow-up (p-value = 1.58 × 10−6) was significant in one component of a replication analysis. This interaction was notable in that rs910652 is located within 78 kilobases of ADAM33, which is one of the most well studied asthma susceptibility genes. In addition, rs11684871 is located in or near GLI2, which may have biologically relevant roles in asthma. Conclusions Using a genome-wide approach, we identified and found suggestive evidence of replication for a gene-gene interaction in asthma involving loci that are potentially highly relevant in asthma pathogenesis. Electronic supplementary material The online version of this article (doi:10.1186/s12863-016-0376-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- William Murk
- Department of Chronic Disease Epidemiology, Yale School of Public Health, 60 College St., New Haven, CT, 06510, USA
| | - Andrew T DeWan
- Department of Chronic Disease Epidemiology, Yale School of Public Health, 60 College St., New Haven, CT, 06510, USA.
| |
Collapse
|
16
|
Dizier MH, Nadif R, Margaritte-Jeannin P, Barton SJ, Sarnowski C, Gagné-Ouellet V, Brossard M, Lavielle N, Just J, Lathrop M, Holloway JW, Laprise C, Bouzigon E, Demenais F. Interaction between the DNAH9 gene and early smoke exposure in bronchial hyperresponsiveness. Eur Respir J 2016; 47:1072-81. [PMID: 26797031 DOI: 10.1183/13993003.00849-2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 11/18/2015] [Indexed: 12/21/2022]
Abstract
A previous genome-wide linkage scan of bronchial hyperresponsiveness (BHR) in the French Epidemiological study on the Genetics and Environment of Asthma (EGEA) families, performed in the presence of a gene×early-life environmental tobacco smoke (ETS) exposure interaction, showed the strongest interaction in the 17p11 region where linkage was detected only among unexposed siblings. Our goal was to conduct fine-scale mapping of 17p11 to identify single nucleotide polymorphisms (SNPs) interacting with ETS that influence BHR.Analyses were performed in 388 French EGEA asthmatic families, using a two-step strategy: 1) selection of SNPs displaying family-based association test (FBAT) association signals (p≤0.01) with BHR in unexposed siblings, and 2) a FBAT homogeneity test between exposed and unexposed siblings plus a robust log-linear interaction test.A single SNP reached the threshold (p≤3×10(-3)) for significant interaction with ETS using both interaction tests, after accounting for multiple testing. Results were replicated in 253 French-Canadian families, but not in 341 UK families, probably due in part to differences in phenotypic features between datasets.The SNP showing significant interaction with ETS belongs toDNAH9(dynein, axonemal, heavy chain 9), a promising candidate gene involved in respiratory cilia mobility and associated with primary ciliary dyskinesia, a disease associated with abnormalities of pulmonary function.
Collapse
Affiliation(s)
- Marie-Hélène Dizier
- INSERM, UMR 946, Genetic Variation and Human Diseases Unit, Paris, France Université Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
| | - Rachel Nadif
- INSERM, U1168, Aging and Chronic Diseases, Epidemiological and Public Health Approaches (VIMA), Villejuif, France Université Versailles Saint-Quentin-en-Yvelines, UMR_S 1168, Paris, France These authors contributed equally to this work
| | - Patricia Margaritte-Jeannin
- INSERM, UMR 946, Genetic Variation and Human Diseases Unit, Paris, France Université Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France These authors contributed equally to this work
| | - Sheila J Barton
- MRC Lifecourse Epidemiology Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Chloé Sarnowski
- INSERM, UMR 946, Genetic Variation and Human Diseases Unit, Paris, France Université Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France Université Paris-Sud, UMR_S 1018, Villejuif, France
| | | | - Myriam Brossard
- INSERM, UMR 946, Genetic Variation and Human Diseases Unit, Paris, France Université Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France Université Paris-Sud, UMR_S 1018, Villejuif, France
| | - Nolwenn Lavielle
- INSERM, UMR 946, Genetic Variation and Human Diseases Unit, Paris, France Université Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France Université Paris-Sud, UMR_S 1018, Villejuif, France
| | - Jocelyne Just
- Service d'Allergologie Pédiatrique, Centre de l'Asthme et des Allergies, Hôpital d'Enfants Armand-Trousseau (APHP) - Sorbonne Universités, UPMC Université Paris 06, UMR_S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Equipe EPAR, Paris, France
| | - Mark Lathrop
- McGill University and Genome Quebec's Innovation Centre, Montréal, Canada
| | - John W Holloway
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Catherine Laprise
- Université du Québec, Chicoutimi, Canada These authors contributed equally to this work
| | - Emmanuelle Bouzigon
- INSERM, UMR 946, Genetic Variation and Human Diseases Unit, Paris, France Université Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France These authors contributed equally to this work
| | - Florence Demenais
- INSERM, UMR 946, Genetic Variation and Human Diseases Unit, Paris, France Université Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
| |
Collapse
|
17
|
Lasso-Pirot A, Delgado-Villalta S, Spanier AJ. Early childhood wheezers: identifying asthma in later life. J Asthma Allergy 2015; 8:63-73. [PMID: 26203265 PMCID: PMC4508083 DOI: 10.2147/jaa.s70066] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Wheeze in young children is common, and asthma is the most common noncommunicable disease in children. Prevalence studies of recurrent asthma-like symptoms in children under the age of 5 years have reported that one third of children in the US and Europe are affected, and rates and severity appear to be higher in developing countries. Over the last few decades, significant research efforts have focused on identification of risk factors and predictors of wheeze and on tools to identify which children who wheeze will progress to develop asthma. We reviewed the phenotypes of childhood wheezing, genetic risk factors, environmental factors, testing/predictive indices, and primary prevention. While it is generally agreed that a complex interaction of environmental exposure and genetic susceptibility contributes to the development of asthma, limitations in predictive tools and tests restrict our ability to provide families with guidance as to whether their child with wheeze will ultimately develop asthma. Additional research is needed to clarify childhood wheeze phenotypes, to develop tools to determine which children will develop asthma, and to determine how and when to intervene. If these areas can be addressed, it would help reduce this large burden on children, families, and society.
Collapse
Affiliation(s)
- Anayansi Lasso-Pirot
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Adam J Spanier
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
18
|
Polonikov AV, Ivanov VP, Bogomazov AD, Solodilova MA. [Genetic and biochemical mechanisms of involvement of antioxidant defense enzymes in the development of bronchial asthma]. BIOMEDITSINSKAIA KHIMIIA 2015; 61:427-439. [PMID: 26350733 DOI: 10.18097/pbmc20156104427] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In the present review we have analyzed and summarized recent literature data on genetic and biochemical mechanisms responsible for involvement of antioxidant defense enzymes in the etiology and pathogenesis of bronchial asthma. It has been shown that the mechanisms of asthma development are linked with genetically determined abnormalities in the functioning of antioxidant defense enzymes. These alterations are accompanied by a systemic imbalance between oxidative and anti-oxidative reactions with the shift of the redox state toward increased free radical production and oxidative stress, a key element in the pathogenesis of bronchial asthma.
Collapse
Affiliation(s)
| | - V P Ivanov
- Kursk State Medical University, Kursk, Russia
| | | | | |
Collapse
|
19
|
Yang IV, Pedersen BS, Liu A, O'Connor GT, Teach SJ, Kattan M, Misiak RT, Gruchalla R, Steinbach SF, Szefler SJ, Gill MA, Calatroni A, David G, Hennessy CE, Davidson EJ, Zhang W, Gergen P, Togias A, Busse WW, Schwartz DA. DNA methylation and childhood asthma in the inner city. J Allergy Clin Immunol 2015; 136:69-80. [PMID: 25769910 PMCID: PMC4494877 DOI: 10.1016/j.jaci.2015.01.025] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 01/21/2015] [Accepted: 01/23/2015] [Indexed: 01/15/2023]
Abstract
BACKGROUND Epigenetic marks are heritable, influenced by the environment, direct the maturation of T lymphocytes, and in mice enhance the development of allergic airway disease. Thus it is important to define epigenetic alterations in asthmatic populations. OBJECTIVE We hypothesize that epigenetic alterations in circulating PBMCs are associated with allergic asthma. METHODS We compared DNA methylation patterns and gene expression in inner-city children with persistent atopic asthma versus healthy control subjects by using DNA and RNA from PBMCs. Results were validated in an independent population of asthmatic patients. RESULTS Comparing asthmatic patients (n = 97) with control subjects (n = 97), we identified 81 regions that were differentially methylated. Several immune genes were hypomethylated in asthma, including IL13, RUNX3, and specific genes relevant to T lymphocytes (TIGIT). Among asthmatic patients, 11 differentially methylated regions were associated with higher serum IgE concentrations, and 16 were associated with percent predicted FEV1. Hypomethylated and hypermethylated regions were associated with increased and decreased gene expression, respectively (P < 6 × 10(-12) for asthma and P < .01 for IgE). We further explored the relationship between DNA methylation and gene expression using an integrative analysis and identified additional candidates relevant to asthma (IL4 and ST2). Methylation marks involved in T-cell maturation (RUNX3), TH2 immunity (IL4), and oxidative stress (catalase) were validated in an independent asthmatic cohort of children living in the inner city. CONCLUSIONS Our results demonstrate that DNA methylation marks in specific gene loci are associated with asthma and suggest that epigenetic changes might play a role in establishing the immune phenotype associated with asthma.
Collapse
Affiliation(s)
- Ivana V Yang
- Department of Medicine, University of Colorado, School of Medicine, Aurora, Colo; Departments of Pediatrics and Medicine, National Jewish Health, Denver, Colo
| | - Brent S Pedersen
- Department of Medicine, University of Colorado, School of Medicine, Aurora, Colo
| | - Andrew Liu
- Departments of Pediatrics and Medicine, National Jewish Health, Denver, Colo
| | - George T O'Connor
- Department of Medicine, Boston University School of Medicine, Boston, Mass
| | | | - Meyer Kattan
- Columbia University Medical Center, New York, NY
| | | | | | | | - Stanley J Szefler
- Department of Pediatrics, Children's Hospital Colorado and University of Colorado, School of Medicine, Aurora, Colo
| | - Michelle A Gill
- University of Texas, Southwestern Medical Center, Dallas, Tex
| | | | | | - Corinne E Hennessy
- Department of Medicine, University of Colorado, School of Medicine, Aurora, Colo
| | - Elizabeth J Davidson
- Department of Medicine, University of Colorado, School of Medicine, Aurora, Colo
| | - Weiming Zhang
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado, Aurora, Colo
| | - Peter Gergen
- National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Alkis Togias
- National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - William W Busse
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - David A Schwartz
- Department of Medicine, University of Colorado, School of Medicine, Aurora, Colo; Departments of Pediatrics and Medicine, National Jewish Health, Denver, Colo; Department of Immunology, University of Colorado, Aurora, Colo.
| |
Collapse
|
20
|
Genetic association of key Th1/Th2 pathway candidate genes, IRF2, IL6, IFNGR2, STAT4 and IL4RA, with atopic asthma in the Indian population. J Hum Genet 2015; 60:443-8. [PMID: 25994869 DOI: 10.1038/jhg.2015.45] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 04/06/2015] [Accepted: 04/10/2015] [Indexed: 02/01/2023]
Abstract
Asthma is a complex, multifactorial disease resulting due to dysregulated immune responses. Genetic factors contribute significantly to asthma pathogenesis, and identification of these factors is one of the major goals in understanding the disease. Th1/Th2 helper differentiation has a critical role in modulating the phenotypes associated with atopic asthma. This study was aimed at identifying genetic modifiers of asthma in selected genes involved in T helper differentiation. A total of 354 single-nucleotide polymorphisms (SNPs) in 33 candidate genes were genotyped in a case-control cohort (cases=147, controls=199) and families (n=247) using Illumina's Golden Gate Assay. Five SNPs, rs3733475A/C (IRF2), rs2069832A/G (IL6), rs2012075G/A (IFNGR2) and rs1400656G/A (STAT4) and rs1805011C/A (IL4RA) were found to be associated with asthma in family based as well as in case-control analyses (P=0.002, P=0.001, P=0.004, P=0.003 and P=0.001, respectively). Interestingly, the minor alleles at these loci showed a protective effect. A five loci haplotype, TAACG, in IRF2 gene, was significantly associated with asthma in families (P=1.1 × 10(-6)) and in case-control cohort (P=0.01). In conclusion, our studies led to identification of some key candidate genes, namely IRF2, IL6, IFNGR2, STAT4 and IL4RA that modulate genetic susceptibility to asthma in the Indian population. Also, this is the first report of independent association of IL6 gene polymorphism with atopic asthma.
Collapse
|
21
|
Piacentini S, Polimanti R, Iorio A, Cortesi M, Papa F, Rongioletti M, Liumbruno GM, Manfellotto D, Fuciarelli M. GSTA1*-69C/T and GSTO2*N142D as asthma- and allergy-related risk factors in Italian adult patients. Clin Exp Pharmacol Physiol 2014; 41:180-4. [PMID: 24471578 DOI: 10.1111/1440-1681.12201] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 01/16/2014] [Accepted: 01/18/2014] [Indexed: 12/12/2022]
Abstract
1. Asthma and allergies are characterized by variable and subjective symptoms influenced by many genes, molecular mechanisms and environmental factors. The presence of inflammation and oxidative stress in the airways are important biochemical features of asthma and respiratory allergies. Glutathione S-transferase (GSTs) enzymes play an important role in cellular protection against inflammation, and functional genetic polymorphisms in GST genes show a significant association with asthma and allergy risk. Specifically, our previous study on asthmatic children highlighted GSTA1 and GSTO2 as novel susceptibility loci for asthma. 2. In the present study we focused our attention on GSTA1*-69C/T (rs3957357) and GSTO2*N142D (rs156697) polymorphisms to confirm our previous results in an independent adult study population and to clarify whether GSTA1 and GSTO2 gene polymorphisms are involved in a non-discriminative pathway towards asthma and respiratory allergy. 3. To accomplish this, we recruited 103 patients with respiratory allergies, 199 patients with asthma and 200 healthy controls. Genomic DNA extracted from buccal cells was screened for GSTA1*-69C/T and GSTO2*N142D single nucleotide polymorphisms. 4. The GSTA1*-69T and GSTO2*D142 variants are both associated with a significantly increased risk of asthma, whereas only GSTA1*-69C/T is significantly associated with allergies. These outcomes confirm the involvement of GSTO2 loci in asthma and suggest that GSTA1 is a common risk factor for asthma and allergies.
Collapse
Affiliation(s)
- Sara Piacentini
- Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Polonikov AV, Ivanov VP, Bogomazov AD, Solodilova MA. Genetic and biochemical mechanisms of involvement of antioxidant defense enzymes in the development of bronchial asthma: A review. BIOCHEMISTRY (MOSCOW) SUPPLEMENT SERIES B: BIOMEDICAL CHEMISTRY 2014; 8:273-285. [DOI: 10.1134/s1990750814040076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
|
23
|
Radder JE, Shapiro SD, Berndt A. Personalized medicine for chronic, complex diseases: chronic obstructive pulmonary disease as an example. Per Med 2014; 11:669-679. [PMID: 29764057 DOI: 10.2217/pme.14.51] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Chronic, complex diseases represent the majority of healthcare utilization and spending in the USA today. Despite this, therapeutics that account for the heterogeneity of these diseases are lacking, begging for more personalized approaches. Improving our understanding of disease phenotypes through retrospective trials of electronic health record data will enable us to better categorize patients. Increased usage of next-generation sequencing will further our understanding of the genetic variants involved in chronic disease. Utilization of data warehousing will be necessary in order to securely handle, integrate and analyze the large sets of data produced with these methods. Finally, increased use of clinical decision support will enable the return of clinically actionable results that physicians can use to apply these personalized approaches.
Collapse
Affiliation(s)
- Josiah E Radder
- Division of Pulmonary, Allergy & Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Steven D Shapiro
- Division of Pulmonary, Allergy & Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Annerose Berndt
- Division of Pulmonary, Allergy & Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
24
|
Mathias RA. Introduction to genetics and genomics in asthma: genetics of asthma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 795:125-55. [PMID: 24162907 DOI: 10.1007/978-1-4614-8603-9_9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
While asthma is a heterogeneous disease, a strong genetic basis has been firmly established. Rather than being a single disease entity, asthma consists of related, overlapping syndromes [Barnes (Proc Am Thor Soc 8:143-148, 2011)] including three general domains: variable airway obstruction, airway hyper-responsiveness, and airway inflammation with a considerable proportion, but not all, of asthma being IgE-mediated further adding to its heterogeneity. This chapter reviews the approaches to the elucidation of genetics of asthma from the early evidence of familial clustering to the current state of knowledge with genome-wide approaches. The conclusion is that research efforts have led to a tremendous repository of genetic determinants of asthma, most of which fall into the above phenotypic domains of the syndrome. We now look to future integrative approaches of genetics, genomics (Chap. 10), and epigenetics (Chap. 11) to better understand the causal mechanism through which, these genetic loci act in manifesting asthma.
Collapse
Affiliation(s)
- Rasika Ann Mathias
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, 5501 Hopkins Bayview Circle, 3B.79, Baltimore, MD, 21224, USA,
| |
Collapse
|
25
|
Abstract
Preschool children (ie, those aged 5 years or younger) with wheeze consume a disproportionately high amount of health-care resources compared with older children and adults with wheeze or asthma, representing a diagnostic challenge. Although several phenotype classifications have been described, none have been validated to identify individuals responding to specific therapeutic approaches. Several risk factors related to genetic, prenatal, and postnatal environment are associated with preschool wheezing. Findings from several cohort studies have shown that preschool children with wheeze have deficits in lung function at 6 years of age that persisted until early and middle adulthood, suggesting increased susceptibility in the first years of life that might lead to persistent sequelae. Daily inhaled corticosteroids seem to be the most effective therapy for recurrent wheezing in trials of children with interim symptoms or atopy; intermittent high-dose inhaled corticosteroids are effective in moderate-to-severe viral-induced wheezing without interim symptoms. The role of leukotriene receptor antagonist is less clear. Interventions to modify the short-term and long-term outcomes of preschool wheeze should be a research priority.
Collapse
Affiliation(s)
- Francine M Ducharme
- Clinical Research and Knowledge Transfer on Childhood Asthma Unit, Research Centre, Sainte-Justine University Health Centre, Montreal, QC, Canada; Department of Paediatrics, University of Montreal, Montreal, QC, Canada; Department of Social and Preventive Medicine, University of Montreal, Montreal, QC, Canada.
| | - Sze M Tse
- Clinical Research and Knowledge Transfer on Childhood Asthma Unit, Research Centre, Sainte-Justine University Health Centre, Montreal, QC, Canada; Department of Paediatrics, University of Montreal, Montreal, QC, Canada
| | - Bhupendrasinh Chauhan
- Clinical Research and Knowledge Transfer on Childhood Asthma Unit, Research Centre, Sainte-Justine University Health Centre, Montreal, QC, Canada
| |
Collapse
|
26
|
Genome-wide association studies in asthma; perhaps, the end of the beginning. Curr Opin Allergy Clin Immunol 2014; 13:463-9. [PMID: 23945178 DOI: 10.1097/aci.0b013e328364ea5f] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE OF REVIEW A large number of genetic loci contribute towards an individual's susceptibility to asthma and other complex diseases. Genome-wide association studies (GWASs) have provided us with a wealth of loci associated with asthma susceptibility, asthma endotypes and responsiveness to asthma medications. The reproducibility of these genetic loci across different studies highlights the interplay of general and population-specific risk alleles in asthma. Although GWASs have been successful in identifying disease-associated loci, there is still large potential for such studies to provide further insights into asthma pathogenesis. RECENT FINDINGS GWASs over the past year have extended study design well beyond the simple case-control and continuous phenotype association formats, for example, including interactions with environmental factors, integrating GWAS data with epigenetic data and GWASs in animal models, incorporating pathway analyses and utilising emerging sequencing technologies. SUMMARY Moving beyond traditional GWAS formats is likely to significantly enhance our understanding of the genetic basis for asthma. This review discusses where we are after half a decade of asthma GWASs, and focuses on advances over the past year that show where the GWAS field is headed in the future.
Collapse
|
27
|
Involvement of lymphocytes in asthma and allergic diseases: a genetic point of view. Curr Opin Allergy Clin Immunol 2014; 13:500-6. [PMID: 23974678 DOI: 10.1097/aci.0b013e328364ea3a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE OF REVIEW The activation and regulation of lymphocytes play a central role in asthmatic inflammation. It is increasingly recognized that diverse panels of lymphocyte lineages and cytokine profiles are involved in the asthmatic phenotypes. In this review, we discuss the advances in the gene variants associated with the regulation of lymphocytes and relevant cytokines underlying asthma and allergic diseases. We also discuss the current evidence about the epigenetic regulation of lymphocyte differentiation and the interaction with environment. RECENT FINDINGS Many genetic variants in asthma are functionally associated with lymphocytes and relevant cytokines. Interleukin (IL)-2RB is important in the homeostasis of T regulatory cells (Tregs) through effects from IL-2. IL-18R1 and ST2/IL-1RL1 drive the T helper 1 and 2 inflammation via the ligands of their encoding receptors. Novel genes, like orosomucoid 1-like 3/gasdermin-like gene and taste receptor type 2 members are being explored for their roles in T-cell activation. T-cell lineages are epigenetically regulated by de novo methyltransferases, histone methylase, CD44 and microRNA. Environmental factors such as second-hand smoke and ambient air pollution modify Tregs differentiation significantly. SUMMARY Plenty of genetic loci of lymphocyte regulation provide us a deeper insight into the asthma pathogenesis. Future challenge is to define genetic drivers in asthma phenotypes to provide therapeutic targets.
Collapse
|
28
|
Heijink IH, Nawijn MC, Hackett TL. Airway epithelial barrier function regulates the pathogenesis of allergic asthma. Clin Exp Allergy 2014; 44:620-30. [DOI: 10.1111/cea.12296] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- I. H. Heijink
- Department of Pathology and Medical Biology; Experimental Pulmonology and Inflammation Research; University Medical Center Groningen; University of Groningen; Groningen the Netherlands
- Department of Pulmonology; University Medical Center Groningen; University of Groningen; Groningen the Netherlands
- GRIAC Research Institute; University Medical Center Groningen; University of Groningen; Groningen the Netherlands
| | - M. C. Nawijn
- Department of Pathology and Medical Biology; Experimental Pulmonology and Inflammation Research; University Medical Center Groningen; University of Groningen; Groningen the Netherlands
- GRIAC Research Institute; University Medical Center Groningen; University of Groningen; Groningen the Netherlands
| | - T.-L. Hackett
- Centre for Heart Lung Innovation; St Paul's Hospital; University of British Columbia; Vancouver BC Canada
| |
Collapse
|
29
|
Laprise C. The Saguenay-Lac-Saint-Jean asthma familial collection: the genetics of asthma in a young founder population. Genes Immun 2014; 15:247-55. [PMID: 24646526 DOI: 10.1038/gene.2014.12] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 02/07/2014] [Accepted: 02/11/2014] [Indexed: 02/06/2023]
Abstract
To study the genetics of asthma, a familial collection was built in the Saguenay-Lac-Saint-Jean (SLSJ) region, which is localized in northeastern Quebec, Canada, and which is characterized by a young founder effect. Recruitment was performed through probands with a specific asthma phenotype. The collection includes 1394 individuals distributed in 271 families. A phenotypic profile including more than 75 phenotypic characteristics was defined for each participant using a standardized questionnaire, respiratory measures and allergic tests. A genome-wide association study on 1214 of these samples for asthma-related phenotypes (asthma, atopy and rhinitis) and for white blood cell types was performed. These analyses allowed associating 10 single nucleotide polymorphisms (SNPs) with P<1 × 10(-5) with asthma-related phenotypes and 11 SNPs with white blood cell counts. Among them, four SNPs are located in or near genes with functions related to asthma or allergy. Moreover, several loci were identified as 5q31.1 that includes 22 SNPs associated with atopy (P<1 × 10(-3)) and located near a cytokine cluster, and 17q21.2 with seven SNPs associated with asthma. This study highlights the value of a familial collection with a fine phenotypic description and characterized by a young founder effect in the search of the genetic determinants involved in complex traits such as asthma.
Collapse
Affiliation(s)
- C Laprise
- Département des sciences fondamentales, Université du Québec à Chicoutimi, Québec, Canada
| |
Collapse
|
30
|
Ko FWS, Lim TK, Hancox RJ, Yang IA. Year in review 2013: Chronic obstructive pulmonary disease, asthma and airway biology. Respirology 2014; 19:438-47. [PMID: 24708033 DOI: 10.1111/resp.12252] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Accepted: 12/27/2013] [Indexed: 12/11/2022]
Affiliation(s)
- Fanny W S Ko
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | | | | | | |
Collapse
|
31
|
KleinJan A, Klein Wolterink RGJ, Levani Y, de Bruijn MJW, Hoogsteden HC, van Nimwegen M, Hendriks RW. Enforced expression of Gata3 in T cells and group 2 innate lymphoid cells increases susceptibility to allergic airway inflammation in mice. THE JOURNAL OF IMMUNOLOGY 2014; 192:1385-94. [PMID: 24415780 DOI: 10.4049/jimmunol.1301888] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Airway inflammation in allergic asthma reflects a threshold response of the innate immune system, including group 2 innate lymphoid cells (ILC2), followed by an adaptive Th2 cell-mediated response. Transcription factor Gata3 is essential for differentiation of both Th2 cells and ILC2. We investigated the effects of enforced Gata3 expression in T cells and ILC2 on the susceptibility of mice to allergic airway inflammation (AAI). We used CD2-Gata3 transgenic (Tg) mice with enforced Gata3 expression driven by the CD2 promoter, which is active both in T cells and during ILC2 development. CD2-Gata3 Tg mice and wild-type (WT) littermates were analyzed in mild models of AAI without adjuvants. Whereas OVA allergen exposure did not induce inflammation in WT controls, CD2-Gata3 Tg mice showed clear AAI and enhanced levels of IL-5 and IL-13 in bronchoalveolar lavage. Likewise, in house dust mite-driven asthma, CD2-Gata3 Tg mice were significantly more susceptible to AAI than WT littermates, whereby both ILC2 and Th2 cells were important cellular sources of IL-5 and IL-13 in bronchoalveolar lavage and lung tissue. Compared with WT littermates, CD2-Gata3 Tg mice contained increased numbers of ILC2, which expressed high levels of IL-33R and contributed significantly to early production of IL-4, IL-5, and IL-13. CD2-Gata3 Tg mice also had a unique population of IL-33-responsive non-B/non-T lymphoid cells expressing IFN-γ. Enforced Gata3 expression is therefore sufficient to enhance Th2 and ILC2 activity, and leads to increased susceptibility to AAI after mild exposure to inhaled harmless Ags that otherwise induce Ag tolerance.
Collapse
Affiliation(s)
- Alex KleinJan
- Department of Pulmonary Medicine, Erasmus University Medical Center Rotterdam, 3000 CA Rotterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
32
|
Chang JC, Kuo HC, Hsu TY, Ou CY, Liu CA, Chuang H, Liang HM, Huang HW, Yang KD. Different genetic associations of the IgE production among fetus, infancy and childhood. PLoS One 2013; 8:e70362. [PMID: 23936416 PMCID: PMC3731352 DOI: 10.1371/journal.pone.0070362] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 06/19/2013] [Indexed: 11/18/2022] Open
Abstract
Elevation of serum IgE levels has long been associated with allergic diseases. Many genes have been linked to IgE production, but few have been linked to the developmental aspects of genetic association with IgE production. To clarify developmental genetic association, we investigated what genes and gene-gene interactions affect IgE levels among fetus, infancy and childhood in Taiwan individuals. A birth cohort of 571 children with completion of IgE measurements from newborn to 1.5, 3, and 6 years of age was subject to genetic association analysis on the 384-customized SNPs of 159 allergy candidate genes. Fifty-three SNPs in 37 genes on innate and adaptive immunity, and stress and response were associated with IgE production. Polymorphisms of the IL13, and the HLA-DPA1 and HLA-DQA1 were, respectively, the most significantly associated with the IgE production at newborn and 6 years of age. Analyses of gene-gene interactions indentified that the combination of NPSR1, rs324981 TT with FGF1, rs2282797 CC had the highest risk (85.7%) of IgE elevation at 1.5 years of age (P = 1.46×10−4). The combination of IL13, CYFIP2 and PDE2A was significantly associated with IgE elevation at 3 years of age (P = 5.98×10−7), and the combination of CLEC2D, COLEC11 and CCL2 was significantly associated with IgE elevation at 6 years of age (P = 6.65×10−7). Our study showed that the genetic association profiles of the IgE production among fetus, infancy and childhood are different. Genetic markers for early prediction and prevention of allergic sensitization may rely on age-based genetic association profiles.
Collapse
Affiliation(s)
- Jen-Chieh Chang
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- Genomic and Proteomic Core Laboratory, Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ho-Chang Kuo
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Te-Yao Hsu
- Department of Obstetrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chia-Yu Ou
- Department of Obstetrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chieh-An Liu
- Department of Pediatrics, Po-Jen Hospital, Kaohsiung, Taiwan
| | - Hau Chuang
- Genomic and Proteomic Core Laboratory, Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hsiu-Mei Liang
- Department of Obstetrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hurng-Wern Huang
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
- * E-mail: (KDY); (HWH)
| | - Kuender D. Yang
- The Department of Medical Research and Development, Show Chwan Memorial Hospital in Chang Bing, Changhua, Taiwan
- Institute of Clinical Medical Sciences, National Yang Ming University, Taipei City, Taiwan
- * E-mail: (KDY); (HWH)
| |
Collapse
|
33
|
Hamzaoui A, Berraies A, Kaabachi W, Haifa M, Ammar J, Kamel H. Induced sputum levels of IL-33 and soluble ST2 in young asthmatic children. J Asthma 2013; 50:803-9. [PMID: 23855553 DOI: 10.3109/02770903.2013.816317] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Interleukin-33 is an IL-1 family cytokine which signals via its T1/ST2 receptor, and acts as a key regulator of inflammation, notably the type-2 response implicated in asthma. This study aims to measure the expression of soluble ST2 (sST2) and IL-33 in asthmatic children, depending on disease activity. METHODS Thirty-seven children with well-defined asthma (20 moderate and 17 mild asthmatics) were studied. IL-33 and sST2 were measured by ELISA in serum and induced sputum (IS) samples, and compared with 22 age- and sex-matched healthy controls. Real-time quantitative PCR was used to determine IL-33 and TNF-α mRNA expression in IS. RESULTS sST2 and IL-33 levels in IS and serum were significantly higher in patients compared with healthy controls (p = 0.0001). The increase in sST2 and IL33 was significantly more important in moderate cases than in mild asthma. A significant correlation was observed between serum and IS IL-33 levels (r = 0.497; p = 0.0018). Higher levels of IL-33 mRNA were detected in IS from asthmatics than those observed in controls. A significant correlation was found between TNF-α and IL-33 mRNA expression in the asthmatic subjects (r = 0.772, p = 0.0001). CONCLUSIONS Values of sST2 and IL-33 observed in IS were found to correlate with disease activity. Elevated IL-33 mRNA expression in IS and its correlation with TNF-α reflected the inflammatory process observed in the lung of young asthmatics.
Collapse
Affiliation(s)
- Agnes Hamzaoui
- Hospital A. Mami, Department of respiratory diseases, Pavillon B
| | | | | | | | | | | |
Collapse
|
34
|
Mullane K, Williams M. Animal models of asthma: reprise or reboot? Biochem Pharmacol 2013; 87:131-9. [PMID: 23831953 DOI: 10.1016/j.bcp.2013.06.026] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 06/25/2013] [Indexed: 10/26/2022]
Abstract
Animal models of disease represent the pinnacle of hierarchical research efforts to validate targets and compounds for therapeutic intervention. Yet models of asthma, particularly in the mouse, which, for practical reasons, has become the sine qua non of asthma research, have been a bone of contention for decades. With barely a nod to their limitations and an extensive history of translational failures, they continue to be used for target identification and to justify the clinical evaluation of new compounds. Recent improvements - including sensitization directly to the airways; use of more relevant allergens; development of a chronic rather than short-term condition; utilization of techniques to measure lung function beyond uninterpretable measures of airway hyperresponsiveness - are laudable but cannot bridge the chasm between the models and the myriad complexities of the human disorder and multiple asthma endophenotypes. While further model developments are necessary, including recognition of key environmental factors beyond allergens, the judicious integration with newer ex vivo and in vitro techniques, including human precision-cut lung slices, reprograming of patient-derived induced pluripotent stem cells and fibroblasts to epithelial and smooth muscle cells, and use of other clinical samples to create a more holistic depiction of activities, might improve their translational success.
Collapse
Affiliation(s)
- Kevin Mullane
- Profectus Pharma Consulting Inc., San Jose, CA, USA.
| | - Michael Williams
- Department of Molecular Pharmacology and Biological Chemistry, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
35
|
Polygenic risk and the development and course of asthma: an analysis of data from a four-decade longitudinal study. THE LANCET RESPIRATORY MEDICINE 2013; 1:453-61. [PMID: 24429243 DOI: 10.1016/s2213-2600(13)70101-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Genome-wide association studies (GWAS) have discovered genetic variants that predispose individuals to asthma. To integrate these new discoveries with emerging models of asthma pathobiology, we aimed to test how genetic discoveries relate to developmental and biological characteristics of asthma. METHODS In this prospective longitudinal study, we investigated a multilocus profile of genetic risk derived from published GWAS of asthma case status. We then tested associations between this genetic risk score and developmental and biological characteristics of asthma in participants enrolled in a population-based long-running birth cohort, the Dunedin Multidisciplinary Health and Development Study (n=1037). We used data on asthma onset, asthma persistence, atopy, airway hyper-responsiveness, incompletely reversible airflow obstruction, and asthma-related school and work absenteeism and hospital admissions obtained during nine prospective assessments spanning the ages of 9 to 38 years. Analyses included cohort members of European descent from whom genetic data had been obtained. FINDINGS Of the 880 cohort members included in our analysis, those at higher genetic risk developed asthma earlier in life than did those with lower genetic risk (hazard ratio [HR] 1·12, 95% CI 1·01-1·26). Of cohort members with childhood-onset asthma, those with higher genetic risk were more likely to develop life-course-persistent asthma than were those with a lower genetic risk (relative risk [RR] 1·36, 95% CI 1·14-1·63). Participants with asthma at higher genetic risk more often had atopy (RR 1·07, 1·01-1·14), airway hyper-responsiveness (RR 1·16, 1·03-1·32), and incompletely reversible airflow obstruction (RR 1·28, 1·04-1·57) than did those with a lower genetic risk. They were also more likely to miss school or work (incident rate ratio 1·38, 1·02-1·86) and be admitted to hospital (HR 1·38, 1·07-1·79) because of asthma. Genotypic information about asthma risk was independent of and additive to information derived from cohort members' family histories of asthma. INTERPRETATION Our findings confirm that GWAS discoveries for asthma are associated with a childhood-onset phenotype. Genetic risk assessments might be able to predict which childhood-onset asthma cases remit and which become life-course-persistent, who might develop impaired lung function, and the burden of asthma in terms of missed school and work and hospital admissions, although these predictions are not sufficiently sensitive or specific to support immediate clinical translation. FUNDING US National Institute on Aging and UK Medical Research Council.
Collapse
|