1
|
Boulund U, Thorsen J, Trivedi U, Tranæs K, Jiang J, Shah SA, Stokholm J. The role of the early-life gut microbiome in childhood asthma. Gut Microbes 2025; 17:2457489. [PMID: 39882630 PMCID: PMC11784655 DOI: 10.1080/19490976.2025.2457489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/05/2024] [Accepted: 01/17/2025] [Indexed: 01/31/2025] Open
Abstract
Asthma is a chronic disease affecting millions of children worldwide, and in severe cases requires hospitalization. The etiology of asthma is multifactorial, caused by both genetic and environmental factors. In recent years, the role of the early-life gut microbiome in relation to asthma has become apparent, supported by an increasing number of population studies, in vivo research, and intervention trials. Numerous early-life factors, which for decades have been associated with the risk of developing childhood asthma, are now being linked to the disease through alterations of the gut microbiome. These factors include cesarean birth, antibiotic use, breastfeeding, and having siblings or pets, among others. Association studies have highlighted several specific microbes that are altered in children developing asthma, but these can vary between studies and disease phenotype. This demonstrates the importance of the gut microbial ecosystem in asthma, and the necessity of well-designed studies to validate the underlying mechanisms and guide future clinical applications. In this review, we examine the current literature on the role of the gut microbiome in childhood asthma and identify research gaps to allow for future microbial-focused therapeutic applications in asthma.
Collapse
Affiliation(s)
- Ulrika Boulund
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
| | - Jonathan Thorsen
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
| | - Urvish Trivedi
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
- Section of Microbiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kaare Tranæs
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
- Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| | - Jie Jiang
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
- Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| | - Shiraz A. Shah
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
| | - Jakob Stokholm
- Copenhagen Prospective Studies on Asthma in Childhood, Copenhagen University Hospital, Herlev-Gentofte, Gentofte, Denmark
- Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
Mauclin M, Guillien A, Niespodziana K, Boudier A, Schlederer T, Bajic M, Errhalt P, Borochova K, Pin I, Gormand F, Vernet R, Bousquet J, Bouzigon E, Valenta R, Siroux V. Association between asthma and IgG levels specific for rhinovirus and respiratory syncytial virus antigens in children and adults. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2025; 4:100342. [PMID: 39507925 PMCID: PMC11536052 DOI: 10.1016/j.jacig.2024.100342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/27/2024] [Accepted: 07/16/2024] [Indexed: 11/08/2024]
Abstract
Background Viral infections in childhood, especially to rhinovirus (RV) and respiratory syncytial virus (RSV), are associated with asthma inception and exacerbation. However, little is known about the role of RV- and RSV-specific antibodies in childhood versus adult asthma. Objective We sought to investigate associations between RV- and RSV-specific IgG levels and asthma phenotypes in children and adults. Methods The analysis included 1771 samples from participants of the Epidemiological Study on the Genetics and Environment of Asthma (530 children; age [mean ± SD], 11.1 ± 2.8, and 1241 adults; age [mean ± SD], 43.4 ± 16.7, among whom 274 and 498 had ever asthma, respectively). RSV- and RV-specific IgG levels were determined using microarrayed virus-derived antigens and peptides. Cross-sectional associations between standardized RSV- and RV-specific IgG levels and asthma phenotypes were estimated by multiple regression models. Results In children, ever asthma was associated with higher IgG levels specific to RV, especially to RV-A and RV-C, and to RSV (adjusted odds ratios [95% CI] for a 1 - SD increase in IgG levels were 1.52 [1.16-1.99], 1.42 [1.10-1.83], and 1.24 [0.99-1.54], respectively). These associations were stronger for moderate to severe asthma than for mild asthma. Conversely in adults, ever asthma was associated with lower RV-A, RV-B, and RV-C IgG levels (adjusted odds ratios [95% CI] were 0.86 [0.74-0.99], 0.83 [0.73-0.95], and 0.85 [0.73-0.99], respectively). Conclusions Our results suggest that the association between respiratory virus-specific antibody levels and asthma varies during life, with asthma associated with higher levels of IgG to RSV, RV-A, and RV-C in children and lower levels of IgG responses to RV-A/B/C in adults.
Collapse
Affiliation(s)
- Marion Mauclin
- Université Grenoble Alpes, INSERM U 1209, CNRS UMR 5309, Team of Environmental Epidemiology Applied to the Development and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
| | - Alicia Guillien
- Université Grenoble Alpes, INSERM U 1209, CNRS UMR 5309, Team of Environmental Epidemiology Applied to the Development and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
| | - Katarzyna Niespodziana
- the Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna
| | - Anne Boudier
- Université Grenoble Alpes, INSERM U 1209, CNRS UMR 5309, Team of Environmental Epidemiology Applied to the Development and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
- CHU Grenoble-Alpes, Grenoble, France
| | - Thomas Schlederer
- the Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna
| | - Maja Bajic
- the Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna
- the Department of Pneumology, University Hospital Krems and Karl Landsteiner University of Health Sciences, Krems
| | - Peter Errhalt
- the Department of Pneumology, University Hospital Krems and Karl Landsteiner University of Health Sciences, Krems
| | - Kristina Borochova
- the Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna
| | - Isabelle Pin
- Université Grenoble Alpes, INSERM U 1209, CNRS UMR 5309, Team of Environmental Epidemiology Applied to the Development and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
| | | | - Raphaël Vernet
- Université Paris Cité, INSERM UMR 1124, Group of Genomic Epidemiology of Multifactorial Diseases, Paris
| | - Jean Bousquet
- Université Paris-Saclay, UVSQ, Université Paris-Sud, INSERM, Equipe d’Epidémiologie Respiratoire Intégrative, CESP, Villejuif
| | - Emmanuelle Bouzigon
- Université Paris Cité, INSERM UMR 1124, Group of Genomic Epidemiology of Multifactorial Diseases, Paris
| | - Rudolf Valenta
- the Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna
- Karl Landsteiner University, Krems
| | - Valérie Siroux
- Université Grenoble Alpes, INSERM U 1209, CNRS UMR 5309, Team of Environmental Epidemiology Applied to the Development and Respiratory Health, Institute for Advanced Biosciences, Grenoble, France
| |
Collapse
|
3
|
Chen D, Wu W, Li J, Huang X, Chen S, Zheng T, Huang G, Ouyang S. Targeting mitochondrial function as a potential therapeutic approach for allergic asthma. Inflamm Res 2025; 74:1. [PMID: 39762562 DOI: 10.1007/s00011-024-01972-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 11/14/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Allergic asthma is a chronic complex airway disease characterized by airway hyperresponsiveness, eosinophilic inflammation, excessive mucus secretion, and airway remodeling, with increasing mortality and incidence globally. The pathogenesis of allergic asthma is influenced by various factors including genetics, environment, and immune responses, making it complex and diverse. Recent studies have found that various cellular functions of mitochondria such as calcium regulation, adenosine triphosphate production, changes in redox potential, and free radical scavenging, are involved in regulating the pathogenesis of asthma. This review explores the involvement of mitochondrial functional changes in the pathogenesis of asthma, and investigate the potential of targeting cellular mitochondria as a therapeutic approach for asthma. Those insights can provide a novel theoretical foundations and treatment strategies for understanding and preventing asthma.
Collapse
Affiliation(s)
- Daichi Chen
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, College of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Wanhua Wu
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, College of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Jianing Li
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, College of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Xueqin Huang
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, College of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Su Chen
- Liaobu Hospital of Dongguan City, Dongguan, 523430, China
| | - TingTing Zheng
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, College of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Gonghua Huang
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, College of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Suidong Ouyang
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, College of Medical Technology, Guangdong Medical University, Dongguan, 523808, China.
- Liaobu Hospital of Dongguan City, Dongguan, 523430, China.
| |
Collapse
|
4
|
Pividori M, Sadeeq S, Krishnan A, Stranger BE, Gignoux CR. Uncovering hidden gene-trait patterns through biclustering analysis of the UK Biobank. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.08.622657. [PMID: 39605717 PMCID: PMC11601405 DOI: 10.1101/2024.11.08.622657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The growing availability of genome-wide association studies (GWAS) and large-scale biobanks provides an unprecedented opportunity to explore the genetic basis of complex traits and diseases. However, with this vast amount of data comes the challenge of interpreting numerous associations across thousands of traits, especially given the high polygenicity and pleiotropy underlying complex phenotypes. Traditional clustering methods, which identify global patterns in data, lack the resolution to capture overlapping associations relevant to subsets of traits or genes. Consequently, there is a critical need for innovative analytic approaches capable of revealing local, biologically meaningful patterns that could advance our understanding of trait comorbidities and gene-trait interactions. Here, we applied BiBit, a biclustering algorithm, to transcriptome-wide association study (TWAS) results from PhenomeXcan, a large resource of gene-trait associations derived from the UK Biobank. BiBit allows simultaneous grouping of traits and genes, identifying biclusters that represent local, overlapping associations. Our analyses uncovered biologically interpretable patterns, including asthma-related biclusters enriched for immune-related gene sets, connections between eye traits and blood pressure, and associations between dietary traits, high cholesterol, and specific loci on chromosome 19. These biclusters highlight gene-trait relationships and patterns of trait co-occurrence that may otherwise be obscured by traditional methods. Our findings demonstrate that biclustering can provide a nuanced view of the genetic architecture of complex traits, offering insights into pleiotropy and disease mechanisms. By enabling the exploration of complex, overlapping patterns within biobank-scale datasets, this approach provides a valuable framework for advancing research on genetic associations, comorbidities, and polygenic traits.
Collapse
Affiliation(s)
- Milton Pividori
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045, USA; Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Suraju Sadeeq
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Arjun Krishnan
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045, USA; Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Barbara E. Stranger
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Christopher R. Gignoux
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045, USA; Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
5
|
Panganiban RA, Nadeau KC, Lu Q. Pyroptosis, gasdermins and allergic diseases. Allergy 2024; 79:2380-2395. [PMID: 39003568 PMCID: PMC11368650 DOI: 10.1111/all.16236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/15/2024]
Abstract
Pyroptosis is an inflammatory form of programmed cell death that is distinct from necrosis and apoptosis. Pyroptosis is primarily mediated by the gasdermin family of proteins (GSDMA-E and PVJK), which, when activated by proteolytic cleavage, form pores in the plasma membrane, leading to cell death. While much of the past research on pyroptosis has focused on its role in cancer, metabolic disorders, and infectious diseases, recent experimental and observational studies have begun to implicate pyroptosis in allergic diseases. These studies suggest that gasdermin-mediated pyroptosis contributes to the development of allergic conditions and could offer novel targets for therapy. Here, we review our current understanding of pyroptosis with an emphasis on the role of gasdermins as executioners of pyroptosis and potential mediators to allergic disease. We highlight new discoveries that establish a mechanistic link between the biochemical actions of gasdermins and the onset of allergic diseases. Additionally, we discuss how pyroptosis and gasdermins might contribute to the dysfunction of epithelial barrier, a key factor believed to initiate the progression of various allergic diseases.
Collapse
Affiliation(s)
- Ronald Allan Panganiban
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Division of Allergy and Inflammation, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Quan Lu
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Makrinioti H, Fainardi V, Bonnelykke K, Custovic A, Cicutto L, Coleman C, Eiwegger T, Kuehni C, Moeller A, Pedersen E, Pijnenburg M, Pinnock H, Ranganathan S, Tonia T, Subbarao P, Saglani S. European Respiratory Society statement on preschool wheezing disorders: updated definitions, knowledge gaps and proposed future research directions. Eur Respir J 2024; 64:2400624. [PMID: 38843917 DOI: 10.1183/13993003.00624-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/05/2024] [Indexed: 07/28/2024]
Abstract
Since the publication of the European Respiratory Society (ERS) task force reports on the management of preschool wheezing in 2008 and 2014, a large body of evidence has accumulated suggesting that the clinical phenotypes that were proposed (episodic (viral) wheezing and multiple-trigger wheezing) do not relate to underlying airway pathology and may not help determine response to treatment. Specifically, using clinical phenotypes alone may no longer be appropriate, and new approaches that can be used to inform clinical care are needed for future research. This ERS task force reviewed the literature published after 2008 related to preschool wheezing and has suggested that the criteria used to define wheezing disorders in preschool children should include age of diagnosis (0 to <6 years), confirmation of wheezing on at least one occasion, and more than one episode of wheezing ever. Furthermore, diagnosis and management may be improved by identifying treatable traits, including inflammatory biomarkers (blood eosinophils, aeroallergen sensitisation) associated with type-2 immunity and differential response to inhaled corticosteroids, lung function parameters and airway infection. However, more comprehensive use of biomarkers/treatable traits in predicting the response to treatment requires prospective validation. There is evidence that specific genetic traits may help guide management, but these must be adequately tested. In addition, the task force identified an absence of caregiver-reported outcomes, caregiver/self-management options and features that should prompt specialist referral for this age group. Priorities for future research include a focus on identifying 1) mechanisms driving preschool wheezing; 2) biomarkers of treatable traits and efficacy of interventions in those without allergic sensitisation/eosinophilia; 3) the need to include both objective outcomes and caregiver-reported outcomes in clinical trials; 4) the need for a suitable action plan for children with preschool wheezing; and 5) a definition of severe/difficult-to-treat preschool wheezing.
Collapse
Affiliation(s)
- Heidi Makrinioti
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- H. Makrinioti and V. Fainardi contributed equally to the manuscript
| | - Valentina Fainardi
- Department of Medicine and Surgery, Paediatric Clinic, University of Parma, Parma, Italy
- H. Makrinioti and V. Fainardi contributed equally to the manuscript
| | - Klaus Bonnelykke
- Department of Pediatrics, University of Copenhagen, Copenhagen, Denmark
| | - Adnan Custovic
- National Heart and Lung Institute, Imperial College London, Imperial NIHR Biomedical Research Centre, and Centre for Paediatrics and Child Health, Imperial College London, London, UK
| | - Lisa Cicutto
- Community Research Department, National Jewish Health, University of Colorado, Denver, CO, USA
| | - Courtney Coleman
- Patient Involvement and Engagement, European Lung Foundation, Sheffield, UK
| | - Thomas Eiwegger
- Department of Pediatric and Adolescent Medicine, University Hospital St Pölten, St Pölten, Austria
- Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
- Translational Medicine Program, Research Institute, Hospital for Sick Children, Toronto, ON, Canada
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Claudia Kuehni
- Institute of Social and Preventive Medicine, Bern, Switzerland
| | - Alexander Moeller
- Department of Respiratory Medicine, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Eva Pedersen
- Institute of Social and Preventive Medicine, Bern, Switzerland
| | - Marielle Pijnenburg
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | | | - Thomy Tonia
- Institute of Social and Preventive Medicine, Bern, Switzerland
| | - Padmaja Subbarao
- SickKids Research Institute, Toronto, ON, Canada
- S. Saglani and P. Subbarao contributed equally to the manuscript
| | - Sejal Saglani
- National Heart and Lung Institute, Imperial College London, Imperial NIHR Biomedical Research Centre, and Centre for Paediatrics and Child Health, Imperial College London, London, UK
- S. Saglani and P. Subbarao contributed equally to the manuscript
| |
Collapse
|
7
|
Ahmad S, Single S, Liu Y, Hough KP, Wang Y, Thannickal VJ, Athar M, Goliwas KF, Deshane JS. Heavy Metal Exposure-Mediated Dysregulation of Sphingolipid Metabolism. Antioxidants (Basel) 2024; 13:978. [PMID: 39199224 DOI: 10.3390/antiox13080978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
Exposure to heavy metals (HMs) is often associated with inflammation and cell death, exacerbating respiratory diseases including asthma. Most inhaled particulate HM exposures result in the deposition of HM-bound fine particulate matter, PM2.5, in pulmonary cell populations. While localized high concentrations of HMs may be a causative factor, existing studies have mostly evaluated the effects of systemic or low-dose chronic HM exposures. This report investigates the impact of local high concentrations of specific HMs (NaAsO2, MnCl2, and CdCl2) on sphingolipid homeostasis and oxidative stress, as both play a role in mediating responses to HM exposure and have been implicated in asthma. Utilizing an in vitro model system and three-dimensional ex vivo human tissue models, we evaluated the expression of enzymatic regulators of the salvage, recycling, and de novo synthesis pathways of sphingolipid metabolism, and observed differential modulation in these enzymes between HM exposures. Sphingolipidomic analyses of specific HM-exposed cells showed increased levels of anti-apoptotic sphingolipids and reduced pro-apoptotic sphingolipids, suggesting activation of the salvage and de novo synthesis pathways. Differential sphingolipid regulation was observed within HM-exposed lung tissues, with CdCl2 exposure and NaAsO2 exposure activating the salvage and de novo synthesis pathway, respectively. Additionally, using spatial transcriptomics and quantitative real-time PCR, we identified HM exposure-induced transcriptomic signatures of oxidative stress in epithelial cells and human lung tissues.
Collapse
Affiliation(s)
- Shaheer Ahmad
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| | - Sierra Single
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| | - Yuelong Liu
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| | - Kenneth P Hough
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| | - Yong Wang
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| | - Victor J Thannickal
- John W. Deming Department of Medicine, Tulane University School of Medicine and Southeast Veterans Healthcare System, New Orleans, LA 70119-6535, USA
| | - Mohammad Athar
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kayla F Goliwas
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| | - Jessy S Deshane
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| |
Collapse
|
8
|
Stikker B, Trap L, Sedaghati-Khayat B, de Bruijn MJW, van Ijcken WFJ, de Roos E, Ikram A, Hendriks RW, Brusselle G, van Rooij J, Stadhouders R. Epigenomic partitioning of a polygenic risk score for asthma reveals distinct genetically driven disease pathways. Eur Respir J 2024; 64:2302059. [PMID: 38901884 PMCID: PMC11358516 DOI: 10.1183/13993003.02059-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 05/28/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Individual differences in susceptibility to developing asthma, a heterogeneous chronic inflammatory lung disease, are poorly understood. Whether genetics can predict asthma risk and how genetic variants modulate the complex pathophysiology of asthma are still debated. AIM To build polygenic risk scores for asthma risk prediction and epigenomically link predictive genetic variants to pathophysiological mechanisms. METHODS Restricted polygenic risk scores were constructed using single nucleotide variants derived from genome-wide association studies and validated using data generated in the Rotterdam Study, a Dutch prospective cohort of 14 926 individuals. Outcomes used were asthma, childhood-onset asthma, adulthood-onset asthma, eosinophilic asthma and asthma exacerbations. Genome-wide chromatin analysis data from 19 disease-relevant cell types were used for epigenomic polygenic risk score partitioning. RESULTS The polygenic risk scores obtained predicted asthma and related outcomes, with the strongest associations observed for childhood-onset asthma (2.55 odds ratios per polygenic risk score standard deviation, area under the curve of 0.760). Polygenic risk scores allowed for the classification of individuals into high-risk and low-risk groups. Polygenic risk score partitioning using epigenomic profiles identified five clusters of variants within putative gene regulatory regions linked to specific asthma-relevant cells, genes and biological pathways. CONCLUSIONS Polygenic risk scores were associated with asthma(-related traits) in a Dutch prospective cohort, with substantially higher predictive power observed for childhood-onset than adult-onset asthma. Importantly, polygenic risk score variants could be epigenomically partitioned into clusters of regulatory variants with different pathophysiological association patterns and effect estimates, which likely represent distinct genetically driven disease pathways. Our findings have potential implications for personalised risk mitigation and treatment strategies.
Collapse
Affiliation(s)
- Bernard Stikker
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Lianne Trap
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- L. Trap and B. Sedaghati-Khayat made an equal contribution to this study
| | - Bahar Sedaghati-Khayat
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- L. Trap and B. Sedaghati-Khayat made an equal contribution to this study
| | - Marjolein J W de Bruijn
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Wilfred F J van Ijcken
- Center for Biomics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Emmely de Roos
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Arfan Ikram
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Guy Brusselle
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jeroen van Rooij
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- J. van Rooij and R. Stadhouders contributed equally to this article as lead authors and supervised the work
| | - Ralph Stadhouders
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- J. van Rooij and R. Stadhouders contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
9
|
Singh S, Kulshrestha MR, Pathak AK, Srivastava S, Singh A, Tiwari V. Transfluthrin is Associated with High Susceptibility to Asthma in Children with Promoter Variants of Beta Chain of High-Affinity Receptor IgE and Tumour Necrosis Factors-α Genes. Biochem Genet 2024; 62:2553-2570. [PMID: 37980703 DOI: 10.1007/s10528-023-10555-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 10/20/2023] [Indexed: 11/21/2023]
Abstract
This study investigates the genetic variations in FcεR1β-109 C/T (rs512555) and TNF-α-308 G/A (rs1800629) genes and examines whether the mosquito repellent transfluthrin (TFT) modifies the risk for asthmatic children. A case-control study was conducted involving 130 asthmatic children and 123 age-sex matched controls. Differential leukocyte counts, IgE, and hs-CRP levels were estimated using a five-part haematology analyzer and Beckman Coulter (AU480), respectively. Genetic variations in FcεR1β-109 and TNF-α-308 were analysed using restriction fragment length polymorphism. Serum TFT levels were measured using gas chromatography-tandem mass spectrometry. Asthmatic children had significantly increased total leukocyte, neutrophil, lymphocyte, eosinophil, and basophil counts (p < 0.0001), while their monocyte counts were lower compared to controls (p < 0.0001). TFT levels were higher in asthmatic children (1.38 ± 0.91 vs. control 0.69 ± 0.41µg/L, p < 0.0001), which predominantly induced wheezing. Elevated TFT levels were associated with an increased risk of childhood asthma (OR: 3.08, p < 0.0001). Children with the FcεRIβ TT (OR: 2.39, p < 0.017) and TNF-α GG genotypes (OR: 7.17, p < 0.0001) were more susceptible to asthma. TFT synergistically enhanced the risk of asthma in both FcεRIβ-109 TT (OR: 5.3, p = 0.001) and TNF-α-308 GG (OR: 17.18, p < 0.0001) genotypes. TFT levels were correlated with IgE (r = 0.363; p = 0.006), hs-CRP (r = 0.324; p = 0.049) and eosinophil (r = 0.300; p = 0.038), respectively. IgE and eosinophils were correlated (r = 0.599, p = 0.001) in the FcεRIβ TT genotype-carrying asthmatic children. Similarly, neutrophils and hs-CRP were correlated (r = 0.768, p < 0.0001) in asthmatic children with TNF-α GG genotype. The risk of asthma is inherently higher in children with FcεRIβ TT and TNF-α GG variants. TFT exposure amplifies the risk of asthma in children among all the subgenotypes of both genes. TFT influences IgE and eosinophil in FcεRIβ TT genotype while it influences neutrophils and hs-CRP in TNF-α GG genotypes.
Collapse
Affiliation(s)
- Shivani Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, Uttar Pradesh, 226028, India
- Department of Biochemistry, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, 226010, India
| | - Manish Raj Kulshrestha
- Department of Biochemistry, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, 226010, India
| | - Anumesh K Pathak
- Department of Biochemistry, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, 226010, India
| | - Shetanshu Srivastava
- Department of Pediatrics, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, 226010, India
| | - Aditi Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow Campus, Lucknow, Uttar Pradesh, 226028, India
| | - Vandana Tiwari
- Department of Biochemistry, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, 226010, India.
| |
Collapse
|
10
|
Chatziparasidis G, Chatziparasidi MR, Kantar A, Bush A. Time-dependent gene-environment interactions are essential drivers of asthma initiation and persistence. Pediatr Pulmonol 2024; 59:1143-1152. [PMID: 38380964 DOI: 10.1002/ppul.26935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/27/2024] [Accepted: 02/12/2024] [Indexed: 02/22/2024]
Abstract
Asthma is a clinical syndrome caused by heterogeneous underlying mechanisms with some of them having a strong genetic component. It is known that up to 82% of atopic asthma has a genetic background with the rest being influenced by environmental factors that cause epigenetic modification(s) of gene expression. The interaction between the gene(s) and the environment has long been regarded as the most likely explanation of asthma initiation and persistence. Lately, much attention has been given to the time frame the interaction occurs since the host response (immune or biological) to environmental triggers, differs at different developmental ages. The integration of the time variant into asthma pathogenesis is appearing to be equally important as the gene(s)-environment interaction. It seems that, all three factors should be present to trigger the asthma initiation and persistence cascade. Herein, we introduce the importance of the time variant in asthma pathogenesis and emphasize the long-term clinical significance of the time-dependent gene-environment interactions in childhood.
Collapse
Affiliation(s)
- Grigorios Chatziparasidis
- Faculty of Nursing, University of Thessaly, Volos, Greece
- School of Physical Education, Sport Science & Dietetics, University of Thessaly, Volos, Greece
| | | | - Ahmad Kantar
- Pediatric Asthma and Cough Centre, Instituti Ospedalieri Bergamashi, Bergamo, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Andrew Bush
- Departments of Paediatrics and Paediatric Respiratory Medicine, Royal Brompton Harefield NHS Foundation Trust and Imperial College, London, UK
| |
Collapse
|
11
|
Li X, He T, Duan S, Liang J, Feng G, Li F, Shen Z, Ye W, Liu B, Jiang B, Chen Y, Liu N, Szeto IMY, Cai L. Infant Formulas With Partially or Extensively Hydrolyzed Milk Proteins for the Prevention of Allergic Diseases: A Systematic Review and Meta-Analysis of Clinical Trials. Adv Nutr 2024; 15:100217. [PMID: 38579971 PMCID: PMC11063603 DOI: 10.1016/j.advnut.2024.100217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 03/21/2024] [Accepted: 03/29/2024] [Indexed: 04/07/2024] Open
Abstract
Despite the widely recommended usage of partially hydrolyzed formula (PHF) or extensively hydrolyzed formula (EHF) of milk protein for preventing allergic diseases (ADs), clinical studies have been inconclusive regarding their efficacy compared with that of cow's milk formula (CMF) or breast milk (BM). We aimed to systematically evaluate the effects of PHF or EHF compared with those of CMF or BM on risk of ADs (cow's milk allergy, allergic rhinitis, eczema, asthma, wheeze, food allergy, and sensitization) in children. We searched PubMed, Embase, Cochrane Library, and Web of Science for clinical trials published from inception to 21 October, 2022. We used the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach to grade the strength of evidence. Overall, 24 trials (10,950 infants) were included, 17 of which specifically included high-risk infants. GRADE was low for the evidence that, compared with CMF, infants early fed with EHF had lower risk of cow's milk allergy at age 0-2 y [relative risk (RR): 0.62; 95% CI: 0.39, 0.99]. Moderate evidence supported that PHF and EHF reduced risk of eczema in children aged younger or older than 2 y, respectively (RR: 0.71; 95% CI: 0.52, 0.96; and RR: 0.79; 95% CI: 0.67, 0.94, respectively). We also identified moderate systematic evidence indicating that PHF reduced risk of wheeze at age 0-2 y compared with CMF (RR: 0.50; 95% CI: 0.29, 0.85), but PHF and EHF increased the risk compared with BM (RR: 1.61; 95% CI: 1.11, 2.31; and RR: 1.64; 95% CI: 1.26, 2.14). Neither PHF nor EHF had significant effects on other ADs in children of any age. In conclusion, compared with CMF, PHF, or EHF had different preventive effect on cow's milk allergy, eczema, and wheeze. Compared with BM, both PHF and EHF may increase risk of wheeze but not other ADs. Given that most trials included only high-risk infants, more research on non-high-risk infants is warranted before any generalization is attempted. This protocol was registered at PROSPERO as CRD42022320787.
Collapse
Affiliation(s)
- Xiaoxu Li
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Tingchao He
- Inner Mongolia Dairy Technology Research Institute, Hohhot, China; Inner Mongolia Yili Industrial Group, Yili Maternal and Infant Nutrition Institute (YMINI), Beijing, China
| | - Sufang Duan
- Inner Mongolia Dairy Technology Research Institute, Hohhot, China; Inner Mongolia Yili Industrial Group, Yili Maternal and Infant Nutrition Institute (YMINI), Beijing, China
| | - Jinghong Liang
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Gang Feng
- Inner Mongolia Yili Industrial Group, Yili Maternal and Infant Nutrition Institute (YMINI), Beijing, China; National Center of Technology Innovation for Dairy, Hohhot, China
| | - Fang Li
- Inner Mongolia Dairy Technology Research Institute, Hohhot, China; Inner Mongolia Yili Industrial Group, Yili Maternal and Infant Nutrition Institute (YMINI), Beijing, China
| | - Zhenyu Shen
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Wenhui Ye
- Inner Mongolia Yili Industrial Group, Hohhot, China
| | - Biao Liu
- Inner Mongolia Yili Industrial Group, Hohhot, China
| | - Bibo Jiang
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yujing Chen
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Nan Liu
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Ignatius Man-Yau Szeto
- Inner Mongolia Yili Industrial Group, Yili Maternal and Infant Nutrition Institute (YMINI), Beijing, China; National Center of Technology Innovation for Dairy, Hohhot, China.
| | - Li Cai
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong Province, China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
12
|
Witonsky JI, Elhawary JR, Eng C, Oh SS, Salazar S, Contreras MG, Medina V, Secor EA, Zhang P, Everman JL, Fairbanks-Mahnke A, Pruesse E, Sajuthi SP, Chang CH, Guerrero TR, Fuentes KC, Lopez N, Montanez-Lopez CA, Otero RA, Rivera RC, Rodriguez L, Vazquez G, Hu D, Huntsman S, Jackson ND, Li Y, Morin A, Nieves NA, Rios C, Serrano G, Williams BJM, Ziv E, Moore CM, Sheppard D, Burchard EG, Seibold MA, Rodriguez Santana JR. The Puerto Rican Infant Metagenomic and Epidemiologic Study of Respiratory Outcomes (PRIMERO): Design and Baseline Characteristics for a Birth Cohort Study of Early-life Viral Respiratory Illnesses and Airway Dysfunction in Puerto Rican Children. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.15.24305359. [PMID: 38699325 PMCID: PMC11065009 DOI: 10.1101/2024.04.15.24305359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Epidemiologic studies demonstrate an association between early-life respiratory illnesses (RIs) and the development of childhood asthma. However, it remains uncertain whether these children are predisposed to both conditions or if early-life RIs induce alterations in airway function, immune responses, or other human biology that contribute to the development of asthma. Puerto Rican children experience a disproportionate burden of early-life RIs and asthma, making them an important population for investigating this complex interplay. PRIMERO, the Puerto Rican Infant Metagenomics and Epidemiologic Study of Respiratory Outcomes , recruited pregnant women and their newborns to investigate how the airways develop in early life among infants exposed to different viral RIs, and will thus provide a critical understanding of childhood asthma development. As the first asthma birth cohort in Puerto Rico, PRIMERO will prospectively follow 2,100 term healthy infants. Collected samples include post-term maternal peripheral blood, infant cord blood, the child's peripheral blood at the year two visit, and the child's nasal airway epithelium, collected using minimally invasive nasal swabs, at birth, during RIs over the first two years of life, and at annual healthy visits until age five. Herein, we describe the study's design, population, recruitment strategy, study visits and procedures, and primary outcomes.
Collapse
|
13
|
Jakwerth CA, Weckmann M, Illi S, Charles H, Zissler UM, Oelsner M, Guerth F, Omony J, Nemani SSP, Grychtol R, Dittrich AM, Skevaki C, Foth S, Weber S, Alejandre Alcazar MA, van Koningsbruggen-Rietschel S, Brock R, Blau S, Hansen G, Bahmer T, Rabe KF, Brinkmann F, Kopp MV, Chaker AM, Schaub B, von Mutius E, Schmidt-Weber CB. 17q21 Variants Disturb Mucosal Host Defense in Childhood Asthma. Am J Respir Crit Care Med 2024; 209:947-959. [PMID: 38064241 PMCID: PMC11531215 DOI: 10.1164/rccm.202305-0934oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 12/07/2023] [Indexed: 03/13/2024] Open
Abstract
Rationale: The strongest genetic risk factor for childhood-onset asthma, the 17q21 locus, is associated with increased viral susceptibility and disease-promoting processes.Objectives: To identify biological targets underlying the escalated viral susceptibility associated with the clinical phenotype mediated by the 17q21 locus.Methods: Genome-wide transcriptome analysis of nasal brush samples from 261 children (78 healthy, 79 with wheezing at preschool age, 104 asthmatic) within the ALLIANCE (All-Age-Asthma) cohort, with a median age of 10.0 (range, 1.0-20.0) years, was conducted to explore the impact of their 17q21 genotype (SNP rs72163891). Concurrently, nasal secretions from the same patients and visits were collected, and high-sensitivity mesoscale technology was employed to measure IFN protein levels.Measurements and Main Results: This study revealed that the 17q21 risk allele induces a genotype- and asthma/wheeze phenotype-dependent enhancement of mucosal GSDMB expression as the only relevant 17q21-encoded gene in children with preschool wheeze. Increased GSDMB expression correlated with the activation of a type-1 proinflammatory, cell-lytic immune, and natural killer signature, encompassing key genes linked to an IFN type-2-signature (IFNG, CXCL9, CXCL10, KLRC1, CD8A, GZMA). Conversely, there was a reduction in IFN type 1 and type 3 expression signatures at the mRNA and protein levels.Conclusions: This study demonstrates a novel disease-driving mechanism induced by the 17q21 risk allele. Increased mucosal GSDMB expression is associated with a cell-lytic immune response coupled with compromised airway immunocompetence. These findings suggest that GSDMB-related airway cell death and perturbations in the mucosal IFN signature account for the increased vulnerability of 17q21 risk allele carriers to respiratory viral infections during early life, opening new options for future biological interventions.The All-Age-Asthma (ALLIANCE) cohort is registered at www.clinicaltrials.gov (pediatric arm, NCT02496468).
Collapse
Affiliation(s)
- Constanze A. Jakwerth
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
- Member of the German Center for Lung Research (DZL), Germany
| | - Markus Weckmann
- Member of the German Center for Lung Research (DZL), Germany
- Division of Epigenetics in Chronic Lung Disease, Priority Area Chronic Lung Diseases, Research Center Borstel-Leibniz Lung Center, Borstel, Germany
- Department of Pediatric Pneumology and Allergology, University Medical Center Schleswig-Holstein, Lübeck, Germany
- Airway Research Center North, Borstel, Lübeck, Kiel, Grosshansdorf, Germany
| | - Sabina Illi
- Member of the German Center for Lung Research (DZL), Germany
- Institute for Asthma and Allergy Prevention, Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany
- Comprehensive Pneumology Center–Munich, Munich, Germany
| | - Helen Charles
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
- Member of the German Center for Lung Research (DZL), Germany
| | - Ulrich M. Zissler
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
- Member of the German Center for Lung Research (DZL), Germany
| | - Madlen Oelsner
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
- Member of the German Center for Lung Research (DZL), Germany
| | - Ferdinand Guerth
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
- Member of the German Center for Lung Research (DZL), Germany
| | - Jimmy Omony
- Member of the German Center for Lung Research (DZL), Germany
- Institute for Asthma and Allergy Prevention, Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany
- Comprehensive Pneumology Center–Munich, Munich, Germany
| | - Sai Sneha Priya Nemani
- Member of the German Center for Lung Research (DZL), Germany
- Department of Pediatric Pneumology and Allergology, University Medical Center Schleswig-Holstein, Lübeck, Germany
- Airway Research Center North, Borstel, Lübeck, Kiel, Grosshansdorf, Germany
| | - Ruth Grychtol
- Member of the German Center for Lung Research (DZL), Germany
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Hanover, Germany
| | - Anna-Maria Dittrich
- Member of the German Center for Lung Research (DZL), Germany
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Hanover, Germany
| | - Chrysanthi Skevaki
- Member of the German Center for Lung Research (DZL), Germany
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics and
| | - Svenja Foth
- Member of the German Center for Lung Research (DZL), Germany
- Universities of Giessen and Marburg Lung Center, Philipps University Marburg and University Children’s Hospital Marburg, University of Marburg, Marburg, Germany
| | - Stefanie Weber
- Member of the German Center for Lung Research (DZL), Germany
- Universities of Giessen and Marburg Lung Center, Philipps University Marburg and University Children’s Hospital Marburg, University of Marburg, Marburg, Germany
| | - Miguel A. Alejandre Alcazar
- Member of the German Center for Lung Research (DZL), Germany
- Institute for Lung Health and Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Giessen, Germany
- Translational Experimental Pediatrics, Experimental Pulmonology, Department of Pediatrics
- Center for Molecular Medicine Cologne and Cologne Excellence Cluster on Stress Responses in Aging-associated Diseases, and
- Pediatric Pulmonology and Allergology, Department of Pediatrics, Faculty of Medicine, University of Cologne and University Hospital Cologne, Cologne, Germany; and
| | - Silke van Koningsbruggen-Rietschel
- Member of the German Center for Lung Research (DZL), Germany
- Pediatric Pulmonology and Allergology, Department of Pediatrics, Faculty of Medicine, University of Cologne and University Hospital Cologne, Cologne, Germany; and
| | - Robert Brock
- Member of the German Center for Lung Research (DZL), Germany
- Pediatric Pulmonology and Allergology, Department of Pediatrics, Faculty of Medicine, University of Cologne and University Hospital Cologne, Cologne, Germany; and
| | - Samira Blau
- Member of the German Center for Lung Research (DZL), Germany
- Pediatric Pulmonology and Allergology, Department of Pediatrics, Faculty of Medicine, University of Cologne and University Hospital Cologne, Cologne, Germany; and
| | - Gesine Hansen
- Member of the German Center for Lung Research (DZL), Germany
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Hanover, Germany
- Cluster of Excellence 2115 (RESIST), Hannover Medical School, Hanover, Germany
| | - Thomas Bahmer
- Member of the German Center for Lung Research (DZL), Germany
- Airway Research Center North, Borstel, Lübeck, Kiel, Grosshansdorf, Germany
- Internal Medicine Department I, University Hospital Schleswig-Holstein–Campus Kiel, Kiel, Germany
| | - Klaus F. Rabe
- Member of the German Center for Lung Research (DZL), Germany
- Airway Research Center North, Borstel, Lübeck, Kiel, Grosshansdorf, Germany
- LungenClinic Grosshansdorf GmbH and Medical Clinics, Christian Albrechts University, Kiel, Germany
| | - Folke Brinkmann
- Member of the German Center for Lung Research (DZL), Germany
- Division of Epigenetics in Chronic Lung Disease, Priority Area Chronic Lung Diseases, Research Center Borstel-Leibniz Lung Center, Borstel, Germany
- Department of Pediatric Pneumology and Allergology, University Medical Center Schleswig-Holstein, Lübeck, Germany
- Airway Research Center North, Borstel, Lübeck, Kiel, Grosshansdorf, Germany
| | - Matthias Volkmar Kopp
- Department of Pediatric Pneumology and Allergology, University Medical Center Schleswig-Holstein, Lübeck, Germany
- Pediatric Respiratory Medicine, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Airway Research Center North, Borstel, Lübeck, Kiel, Grosshansdorf, Germany
| | - Adam M. Chaker
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
- Department of Otorhinolaryngology and Head and Neck Surgery, Medical School, Technical University of Munich, Munich, Germany
| | - Bianca Schaub
- Member of the German Center for Lung Research (DZL), Germany
- Dr. von Hauner Children’s Hospital, Ludwig Maximilians University, Munich, Germany
- Comprehensive Pneumology Center–Munich, Munich, Germany
| | - Erika von Mutius
- Member of the German Center for Lung Research (DZL), Germany
- Institute for Asthma and Allergy Prevention, Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany
- Dr. von Hauner Children’s Hospital, Ludwig Maximilians University, Munich, Germany
| | - Carsten B. Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
- Member of the German Center for Lung Research (DZL), Germany
| | - the ALLIANCE Study Group as part of the German Center for Lung Research
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
- Member of the German Center for Lung Research (DZL), Germany
- Institute for Asthma and Allergy Prevention, Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany
- Dr. von Hauner Children’s Hospital, Ludwig Maximilians University, Munich, Germany
- Comprehensive Pneumology Center–Munich, Munich, Germany
- Division of Epigenetics in Chronic Lung Disease, Priority Area Chronic Lung Diseases, Research Center Borstel-Leibniz Lung Center, Borstel, Germany
- Department of Pediatric Pneumology and Allergology, University Medical Center Schleswig-Holstein, Lübeck, Germany
- Department of Otorhinolaryngology and Head and Neck Surgery, Medical School, Technical University of Munich, Munich, Germany
- Pediatric Respiratory Medicine, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Airway Research Center North, Borstel, Lübeck, Kiel, Grosshansdorf, Germany
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Hanover, Germany
- Internal Medicine Department I, University Hospital Schleswig-Holstein–Campus Kiel, Kiel, Germany
- LungenClinic Grosshansdorf GmbH and Medical Clinics, Christian Albrechts University, Kiel, Germany
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics and
- Institute for Lung Health and Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Giessen, Germany
- Universities of Giessen and Marburg Lung Center, Philipps University Marburg and University Children’s Hospital Marburg, University of Marburg, Marburg, Germany
- Translational Experimental Pediatrics, Experimental Pulmonology, Department of Pediatrics
- Center for Molecular Medicine Cologne and Cologne Excellence Cluster on Stress Responses in Aging-associated Diseases, and
- Pediatric Pulmonology and Allergology, Department of Pediatrics, Faculty of Medicine, University of Cologne and University Hospital Cologne, Cologne, Germany; and
- Cluster of Excellence 2115 (RESIST), Hannover Medical School, Hanover, Germany
| |
Collapse
|
14
|
Kilic A, Halu A, De Marzio M, Maiorino E, Duvall MG, Bruggemann TR, Rojas Quintero JJ, Chase R, Mirzakhani H, Sungur AÖ, Koepke J, Nakano T, Peh HY, Krishnamoorthy N, Abdulnour RE, Georgopoulos K, Litonjua AA, Demay M, Renz H, Levy BD, Weiss ST. Vitamin D constrains inflammation by modulating the expression of key genes on Chr17q12-21.1. eLife 2024; 12:RP89270. [PMID: 38567749 PMCID: PMC10990493 DOI: 10.7554/elife.89270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024] Open
Abstract
Vitamin D possesses immunomodulatory functions and vitamin D deficiency has been associated with the rise in chronic inflammatory diseases, including asthma (Litonjua and Weiss, 2007). Vitamin D supplementation studies do not provide insight into the molecular genetic mechanisms of vitamin D-mediated immunoregulation. Here, we provide evidence for vitamin D regulation of two human chromosomal loci, Chr17q12-21.1 and Chr17q21.2, reliably associated with autoimmune and chronic inflammatory diseases. We demonstrate increased vitamin D receptor (Vdr) expression in mouse lung CD4+ Th2 cells, differential expression of Chr17q12-21.1 and Chr17q21.2 genes in Th2 cells based on vitamin D status and identify the IL-2/Stat5 pathway as a target of vitamin D signaling. Vitamin D deficiency caused severe lung inflammation after allergen challenge in mice that was prevented by long-term prenatal vitamin D supplementation. Mechanistically, vitamin D induced the expression of the Ikzf3-encoded protein Aiolos to suppress IL-2 signaling and ameliorate cytokine production in Th2 cells. These translational findings demonstrate mechanisms for the immune protective effect of vitamin D in allergic lung inflammation with a strong molecular genetic link to the regulation of both Chr17q12-21.1 and Chr17q21.2 genes and suggest further functional studies and interventional strategies for long-term prevention of asthma and other autoimmune disorders.
Collapse
Affiliation(s)
- Ayse Kilic
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Arda Halu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Margherita De Marzio
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical SchoolBostonUnited States
- Department of Environmental Health, Harvard TH Chan School of Public HealthBostonUnited States
| | - Enrico Maiorino
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Melody G Duvall
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical SchoolBostonUnited States
| | - Thayse Regina Bruggemann
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical SchoolBostonUnited States
| | - Joselyn J Rojas Quintero
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical SchoolBostonUnited States
| | - Robert Chase
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Hooman Mirzakhani
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical SchoolBostonUnited States
| | - Ayse Özge Sungur
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps-UniversityMarburgGermany
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Justus Liebig University GiessenGiessenGermany
| | - Janine Koepke
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Justus Liebig University GiessenGiessenGermany
| | - Taiji Nakano
- Department of Pediatrics, Graduate School of Medicine, Chiba UniversityChibaJapan
| | - Hong Yong Peh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical SchoolBostonUnited States
| | - Nandini Krishnamoorthy
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical SchoolBostonUnited States
| | - Raja-Elie Abdulnour
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical SchoolBostonUnited States
| | - Katia Georgopoulos
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical SchoolBostonUnited States
| | - Augusto A Litonjua
- Division of Pediatric Pulmonary Medicine, Golisano Children’s Hospital at Strong, University of Rochester Medical CenterRochesterUnited States
| | - Marie Demay
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical SchoolBostonUnited States
| | - Harald Renz
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University of Marburg and German Center for Lung Research (DZL)MarburgGermany
- Department of Clinical Immunology and Allergology, Laboratory of Immunopathology Sechenov UniversityMoscowRussian Federation
| | - Bruce D Levy
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical SchoolBostonUnited States
| | - Scott T Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical SchoolBostonUnited States
| |
Collapse
|
15
|
Hizawa N. Common Pathogeneses Underlying Asthma and Chronic Obstructive Pulmonary Disease -Insights from Genetic Studies. Int J Chron Obstruct Pulmon Dis 2024; 19:633-642. [PMID: 38464563 PMCID: PMC10922945 DOI: 10.2147/copd.s441992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/21/2024] [Indexed: 03/12/2024] Open
Abstract
Neither asthma nor chronic obstructive pulmonary disease (COPD) is a single disease consisting of a uniform pathogenesis; rather, they are both syndromes that result from a variety of basic distinct pathogeneses. Many of the basic pathogeneses overlap between the two diseases, and multiple basic pathogeneses are simultaneously involved at varying proportions in individual patients. The specific combination of different basic pathogeneses in each patient determines the phenotype of the patient, and it varies widely from patient to patient. For example, type 2 airway inflammation and neutrophilic airway inflammation may coexist in the same patient, and quite a few patients have clinical characteristics of both asthma and COPD. Even in the same patient, the contribution of each pathogenesis is expected to differ at different life stages (eg, childhood, adolescence, middle age, and older), during different seasons (eg, high seasons for hay fever and rhinovirus infection), and depending on the nature of treatments. This review describes several basic pathogeneses commonly involved in both asthma and COPD, including chronic non-type 2 inflammation, type 2 inflammation, viral infections, and lung development. Understanding of the basic molecular pathogeneses in individual patients, rather than the use of clinical diagnosis, such as asthma, COPD, or even asthma COPD overlap, will enable us to better deal with the diversity seen in disease states, and lead to optimal treatment practices tailored for each patient with less disease burden, such as drug-induced side effects, and improved prognosis. Furthermore, we can expect to focus on these molecular pathways as new drug discovery targets.
Collapse
Affiliation(s)
- Nobuyuki Hizawa
- Department of Pulmonary Medicine, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
16
|
Chen Y, Checa A, Zhang P, Huang M, Kelly RS, Kim M, Chen YCS, Lee-Sarwar KA, Prince N, Mendez KM, Begum S, Kachroo P, Chu SH, Stokholm J, Bønnelykke K, Litonjua AA, Bisgaard H, Weiss ST, Chawes BL, Wheelock CE, Lasky-Su JA. Sphingolipid classes and the interrelationship with pediatric asthma and asthma risk factors. Allergy 2024; 79:404-418. [PMID: 38014461 PMCID: PMC11175620 DOI: 10.1111/all.15942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 10/10/2023] [Accepted: 10/15/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND While dysregulated sphingolipid metabolism has been associated with risk of childhood asthma, the specific sphingolipid classes and/or mechanisms driving this relationship remain unclear. We aimed to understand the multifaceted role between sphingolipids and other established asthma risk factors that complicate this relationship. METHODS We performed targeted LC-MS/MS-based quantification of 77 sphingolipids in plasma from 997 children aged 6 years from two independent cohorts (VDAART and COPSAC2010 ). We examined associations of circulatory sphingolipids with childhood asthma, lung function, and three asthma risk factors: functional SNPs in ORMDL3, low vitamin D levels, and reduced gut microbial maturity. Given racial differences between these cohorts, association analyses were performed separately and then meta-analyzed together. RESULTS We observed elevations in circulatory sphingolipids with asthma phenotypes and risk factors; however, there were differential associations of sphingolipid classes with clinical outcomes and/or risk factors. While elevations from metabolites involved in ceramide recycling and catabolic pathways were associated with asthma and worse lung function [meta p-value range: 1.863E-04 to 2.24E-3], increased ceramide levels were associated with asthma risk factors [meta p-value range: 7.75E-5 to .013], but not asthma. Further investigation identified that some ceramides acted as mediators while some interacted with risk factors in the associations with asthma outcomes. CONCLUSION This study demonstrates the differential role that sphingolipid subclasses may play in asthma and its risk factors. While overall elevations in sphingolipids appeared to be deleterious overall; elevations in ceramides were uniquely associated with increases in asthma risk factors only; while elevations in asthma phenotypes were associated with recycling sphingolipids. Modification of asthma risk factors may play an important role in regulating sphingolipid homeostasis via ceramides to affect asthma. Further function work may validate the observed associations.
Collapse
Affiliation(s)
- Yulu Chen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Antonio Checa
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institute, 171 77, Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, 171 76, Stockholm, Sweden
| | - Pei Zhang
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institute, 171 77, Stockholm, Sweden
- Gunma University Initiative for Advanced Research (GIAR), Gunma University, Maebashi, Gunma, 371-8511, Japan
| | - Mengna Huang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Rachel S. Kelly
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Min Kim
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, 2820 Gentofte, Denmark
| | - Yih-Chieh S. Chen
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Kathleen A. Lee-Sarwar
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Nicole Prince
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Kevin M. Mendez
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Sofina Begum
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Priyadarshini Kachroo
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Su H. Chu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Jakob Stokholm
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, 2820 Gentofte, Denmark
| | - Klaus Bønnelykke
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, 2820 Gentofte, Denmark
| | - Augusto A. Litonjua
- Division of Pediatric Pulmonary Medicine, Department of Pediatrics, Golisano Children’s Hospital and University of Rochester Medical Center, Rochester, NY, USA
| | - Hans Bisgaard
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, 2820 Gentofte, Denmark
| | - Scott T. Weiss
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Bo L. Chawes
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, 2820 Gentofte, Denmark
| | - Craig E. Wheelock
- Unit of Integrative Metabolomics, Institute of Environmental Medicine, Karolinska Institute, 171 77, Stockholm, Sweden
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, 171 76, Stockholm, Sweden
- Gunma University Initiative for Advanced Research (GIAR), Gunma University, Maebashi, Gunma, 371-8511, Japan
| | - Jessica A. Lasky-Su
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Wolters AAB, Kersten ETG, Koppelman GH. Genetics of preschool wheeze and its progression to childhood asthma. Pediatr Allergy Immunol 2024; 35:e14067. [PMID: 38284918 DOI: 10.1111/pai.14067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 01/30/2024]
Abstract
Wheezing is a common and heterogeneous condition in preschool children. In some countries, the prevalence can be as high as 30% and up to 50% of all children experience wheezing before the age of 6. Asthma often starts with preschool wheeze, but not all wheezing children will develop asthma at school age. At this moment, it is not possible to accurately predict which wheezing children will develop asthma. Recently, studying the genetics of wheeze and the childhood-onset of asthma have grown in interest. Childhood-onset asthma has a stronger heritability in comparison with adult-onset asthma. In early childhood asthma exacerbations, CDHR3, which encodes the receptor for Rhinovirus C, was identified, as well as IL33, and the 17q locus that includes GSDMB and ORMDL3 genes. The 17q locus is the strongest wheeze and childhood-onset asthma locus, and was shown to interact with many environmental factors, including smoking and infections. Finally, ANXA1 was recently associated with early-onset, persistent wheeze. ANXA1 may help resolve eosinophilic inflammation. Overall, despite its complexities, genetic approaches to unravel the early-onset of wheeze and asthma are promising, since these shed more light on mechanisms of childhood asthma-onset. Implicated genes point toward airway epithelium and its response to external factors, such as viral infections. However, the heterogeneity of wheeze phenotypes complicates genetic studies. It is therefore important to define accurate wheezing phenotypes and forge larger international collaborations to gain a better understanding of the pathways underlying early-onset asthma.
Collapse
Affiliation(s)
- Alba A B Wolters
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Elin T G Kersten
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gerard H Koppelman
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
18
|
Almacioglu M, Keskin O, Ozkars MY, Balci SO, Kucukosmanoglu E, Pehlivan S, Keskin M. Association of childhood asthma with Gasdermin B (GSDMB) and Oromucoid-like 3 (ORMDL3) genes. North Clin Istanb 2023; 10:769-777. [PMID: 38328715 PMCID: PMC10846573 DOI: 10.14744/nci.2023.22120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/19/2022] [Accepted: 01/29/2023] [Indexed: 02/17/2023] Open
Abstract
OBJECTIVE Genome-length association studies have shown that Gasdermin B (GSDMB) and Orosomucoid-like 3 (ORMDL3) genes located on the long arm of chromosome 17 are associated with asthma. In this study, it was aimed to determine the possible relationship between asthma control test (ACT), exercise provocation test (ECT), and fractional nitric oxide (FENO) levels and GSDMB and ORMDL3 gene expressions. METHODS 59 asthmatic and 38 non-asthmatic children were included in the study. We divided the patient group into two subgroups as mild persistent asthma (29 patients) and moderate persistent asthma (30 patients). ORMDL3, GSDMB gene expression levels, ECT, total IgE levels, and eosinophil counts were measured in all cases. In addition, ACT and FeNO levels were measured in children with asthma. Afterward, the relationship of ORMDL3 and GSDMB gene expression coefficient changes with ECT, ACT, and FeNO was examined. RESULTS When patients with ACT ≤15 were compared with patients with ACT ≥20, ORMDL3 and GSDMB gene expressions were increased 6.74 and 11.74 times, respectively. Comparing patients with ACT ≥20 and ACT ≤15 in terms of coefficient changes (ΔCq), higher change values were observed for ΔCq ORMDL3 in patients with ACT ≤15 (p=0.015). Similarly, when patients with FENO ≤25 ppb were compared with patients with FENO >25 ppb, ORMDL3 and GSDMB gene expressions were increased by 2.93 and 3.56 times, respectively. When the coefficient changes were compared, no significant difference was found between FENO≤25 and FENO >25 patients. There was a slight negative correlation between ΔCq values and ACT score (p=0.003, r=-0.418 for ORMDL3, and p=0.016, r=-0.345 for GSDMB). In addition, we observed a statistically significant positive correlation between ORMDL3 and GSDMB gene expressions (r=0.80, p<0.001). CONCLUSION We showed that increased ORMDL3 and GSDMB gene expression levels may be associated with ACT scores, FeNO and ECT in asthma. These findings may encourage future studies with larger numbers of subjects that can use gene expression levels in various asthma phenotypes for prognostic prediction.
Collapse
Affiliation(s)
- Mehmet Almacioglu
- Department of Pediatrics, SANKO University Faculty of Medicine, Gaziantep, Turkiye
| | - Ozlem Keskin
- Department of Pediatric Allergy and Immunology, Gaziantep University Faculty of Medicine, Gaziantep, Turkiye
| | - Mehmet Yasar Ozkars
- Department of Pediatric Allergy and Immunology, Gaziantep University Faculty of Medicine, Gaziantep, Turkiye
| | - Sibel Oguzkan Balci
- Department of Biology, Gaziantep University Faculty of Medicine, Gaziantep, Turkiye
| | - Ercan Kucukosmanoglu
- Department of Pediatric Allergy and Immunology, Gaziantep University Faculty of Medicine, Gaziantep, Turkiye
| | - Sacide Pehlivan
- Department of Biology, Istanbul University Faculty of Medicine, Istanbul, Turkiye
| | - Mehmet Keskin
- Department of Pediatrics, Gaziantep University Faculty of Medicine, Gaziantep, Turkiye
| |
Collapse
|
19
|
Margaritte-Jeannin P, Vernet R, Budu-Aggrey A, Ege M, Madore AM, Linhard C, Mohamdi H, von Mutius E, Granell R, Demenais F, Laprise C, Bouzigon E, Dizier MH. TNS1 and NRXN1 Genes Interacting With Early-Life Smoking Exposure in Asthma-Plus-Eczema Susceptibility. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2023; 15:779-794. [PMID: 37957795 PMCID: PMC10643854 DOI: 10.4168/aair.2023.15.6.779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 05/15/2023] [Accepted: 06/13/2023] [Indexed: 11/15/2023]
Abstract
PURPOSE Numerous genes have been associated with allergic diseases (asthma, allergic rhinitis, and eczema), but they explain only part of their heritability. This is partly because most previous studies ignored complex mechanisms such as gene-environment (G-E) interactions and complex phenotypes such as co-morbidity. However, it was recently evidenced that the co-morbidity of asthma-plus-eczema appears as a sub-entity depending on specific genetic factors. Besides, evidence also suggest that gene-by-early life environmental tobacco smoke (ETS) exposure interactions play a role in asthma, but were never investigated for asthma-plus-eczema. To identify genetic variants interacting with ETS exposure that influence asthma-plus-eczema susceptibility. METHODS To conduct a genome-wide interaction study (GWIS) of asthma-plus-eczema according to ETS exposure, we applied a 2-stage strategy with a first selection of single nucleotide polymorphisms (SNPs) from genome-wide association meta-analysis to be tested at a second stage by interaction meta-analysis. All meta-analyses were conducted across 4 studies including a total of 5,516 European-ancestry individuals, of whom 1,164 had both asthma and eczema. RESULTS Two SNPs showed significant interactions with ETS exposure. They were located in 2 genes, NRXN1 (2p16) and TNS1 (2q35), never reported associated and/or interacting with ETS exposure for asthma, eczema or more generally for allergic diseases. TNS1 is a promising candidate gene because of its link to lung and skin diseases with possible interactive effect with tobacco smoke exposure. CONCLUSIONS This first GWIS of asthma-plus-eczema with ETS exposure underlines the importance of studying sub-phenotypes such as co-morbidities as well as G-E interactions to detect new susceptibility genes.
Collapse
Affiliation(s)
- Patricia Margaritte-Jeannin
- Université Paris Cité, UMRS 1124, INSERM, Genomic Epidemiology and Multifactorial Diseases Group, Paris, France
| | - Raphaël Vernet
- Université Paris Cité, UMRS 1124, INSERM, Genomic Epidemiology and Multifactorial Diseases Group, Paris, France
| | - Ashley Budu-Aggrey
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Markus Ege
- Dr von Hauner Children's Hospital, Ludwig Maximilian University; Institute of Asthma and Allergy prevention, Helmholtz Centre Munich; Comprehensive Pneumology Center Munich (CPC-M), German Center for Lung Research, Munich, Germany
| | - Anne-Marie Madore
- Département des sciences fondamentales, Centre intersectoriel en santé durable (CISD), Université du Québec à Chicoutimi, Saguenay, QC, Canada
| | - Christophe Linhard
- Université Paris Cité, UMRS 1124, INSERM, Genomic Epidemiology and Multifactorial Diseases Group, Paris, France
| | - Hamida Mohamdi
- Université Paris Cité, UMRS 1124, INSERM, Genomic Epidemiology and Multifactorial Diseases Group, Paris, France
| | - Erika von Mutius
- Dr von Hauner Children's Hospital, Ludwig Maximilian University; Institute of Asthma and Allergy prevention, Helmholtz Centre Munich; Comprehensive Pneumology Center Munich (CPC-M), German Center for Lung Research, Munich, Germany
| | - Raquell Granell
- Medical Research Council (MRC) Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Florence Demenais
- Université Paris Cité, UMRS 1124, INSERM, Genomic Epidemiology and Multifactorial Diseases Group, Paris, France
| | - Cathrine Laprise
- Département des sciences fondamentales, Centre intersectoriel en santé durable (CISD), Université du Québec à Chicoutimi, Saguenay, QC, Canada
| | - Emmanuelle Bouzigon
- Université Paris Cité, UMRS 1124, INSERM, Genomic Epidemiology and Multifactorial Diseases Group, Paris, France
| | - Marie-Hélène Dizier
- Université Paris Cité, UMRS 1124, INSERM, Genomic Epidemiology and Multifactorial Diseases Group, Paris, France.
| |
Collapse
|
20
|
Chatziparasidis G, Bush A, Chatziparasidi MR, Kantar A. Airway epithelial development and function: A key player in asthma pathogenesis? Paediatr Respir Rev 2023; 47:51-61. [PMID: 37330410 DOI: 10.1016/j.prrv.2023.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 04/07/2023] [Accepted: 04/25/2023] [Indexed: 06/19/2023]
Abstract
Though asthma is a common and relatively easy to diagnose disease, attempts at primary or secondary prevention, and cure, have been disappointing. The widespread use of inhaled steroids has dramatically improved asthma control but has offered nothing in terms of altering long-term outcomes or reversing airway remodeling and impairment in lung function. The inability to cure asthma is unsurprising given our limited understanding of the factors that contribute to disease initiation and persistence. New data have focused on the airway epithelium as a potentially key factor orchestrating the different stages of asthma. In this review we summarize for the clinician the current evidence on the central role of the airway epithelium in asthma pathogenesis and the factors that may alter epithelial integrity and functionality.
Collapse
Affiliation(s)
- Grigorios Chatziparasidis
- Paediatric Respiratory Unit, IASO Hospital, Larissa, Thessaly, Greece; Faculty of Nursing, Thessaly University, Greece.
| | - Andrew Bush
- National Heart and Lung Institute, Royal Brompton & Harefield NHS Foundation Trust, London, UK
| | | | - Ahmad Kantar
- Pediatric Asthma and Cough Centre, Instituti Ospedalieri Bergamaschi, University and Research Hospitals, Bergamo, Italy
| |
Collapse
|
21
|
Zhang Y. From gene identifications to therapeutic targets for asthma: Focus on great potentials of TSLP, ORMDL3, and GSDMB. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2023; 1:139-147. [PMID: 39171126 PMCID: PMC11332877 DOI: 10.1016/j.pccm.2023.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Indexed: 08/23/2024]
Abstract
Asthma is a chronic respiratory disease, and clinically, asthma exacerbations remain difficult to treat. The disease is caused by combinations of and interactions between genetic and environmental factors. Genomic and genetic approaches identified many novel genes to treat asthma and brought new insights into the disease. The products of the genes have functional roles in regulating physiological or pathophysiological processes in airway structural cells and immune system cells. Genetic factors also interact with environmental factors such as air pollutants, and bacterial and viral infections to trigger the disease. Thymic stromal lymphopoietin (TSLP), orosomucoid-like 3 (ORMDL3), and gasdermin B (GSDMB) are three genes identified by genetic studies to have a great potential as therapeutic targets of asthma. TSLP is an important driver of type 2 inflammation. ORMDL3 mediates cell stress, sphingolipid synthesis, and viral and bacterial infections. GSDMB regulates cell pyroptosis through its N and C terminals and can bind sulfatides to influence inflammatory response. Investigating inhibitors or modulators for these pathways would bring a new landscape for therapeutics of asthma in future.
Collapse
Affiliation(s)
- Youming Zhang
- National Heart and Lung Institute, Imperial College London, Dovehouse Street, London SW3 6LY, UK
| |
Collapse
|
22
|
Wu D, Zhang X, Zimmerly KM, Wang R, Wang C, Hunter R, Wu X, Campen M, Liu M, Yang XO. Unfolded protein response factor ATF6 augments T helper cell responses and promotes mixed granulocytic airway inflammation. Mucosal Immunol 2023; 16:499-512. [PMID: 37209959 PMCID: PMC10530451 DOI: 10.1016/j.mucimm.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/11/2023] [Indexed: 05/22/2023]
Abstract
The unfolded protein response (UPR) is associated with the risk of asthma, including treatment-refractory severe asthma. Recent studies demonstrated a pathogenic role of activating transcription factor 6a (ATF6a or ATF6), an essential UPR sensor, in airway structural cells. However, its role in T helper (TH) cells has not been well examined. In this study, we found that ATF6 was selectively induced by signal transducer and activator of transcription6 (STAT6) and STAT3 in TH2 and TH17 cells, respectively. ATF6 upregulated UPR genes and promoted the differentiation and cytokine secretion of TH2 and TH17 cells. T cell-specific Atf6-deficiency impaired TH2 and TH17 responses in vitro and in vivo and attenuated mixed granulocytic experimental asthma. ATF6 inhibitor Ceapin A7 suppressed the expression of ATF6 downstream genes and TH cell cytokines by both murine and human memory clusters of differentiation 4 (CD4)+ T cells. At the chronic stage of asthma, administration of Ceapin A7 lessened TH2 and TH17 responses, leading to alleviation of both airway neutrophilia and eosinophilia. Thus, our results demonstrate a critical role of ATF6 in TH2 and TH17 cell-driven mixed granulocytic airway disease, suggesting a novel option to combat steroid-resistant mixed and even T2-low endotypes of asthma by targeting ATF6.
Collapse
Affiliation(s)
- Dandan Wu
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, USA
| | - Xing Zhang
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, USA
| | - Kourtney M Zimmerly
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, USA
| | - Ruoning Wang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, USA
| | - Chunqing Wang
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, USA
| | - Russell Hunter
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, USA
| | - Xiang Wu
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, USA; Department of Parasitology, School of Basic Medical Sciences, Xiangya School of Medicine, Central South University, Changsha, China
| | - Matthew Campen
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, USA
| | - Meilian Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, USA.
| | - Xuexian O Yang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, USA.
| |
Collapse
|
23
|
Wu D, Zhang X, Zimmerly KM, Wang R, Livingston A, Iwawaki T, Kumar A, Wu X, Mandell MA, Liu M, Yang XO. Unconventional Activation of IRE1 Enhances TH17 Responses and Promotes Neutrophilic Airway Inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.30.547286. [PMID: 37461622 PMCID: PMC10349957 DOI: 10.1101/2023.06.30.547286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Treatment-refractory severe asthma manifests a neutrophilic phenotype associated with TH17 responses. Heightened unfolded protein responses (UPRs) are associated with the risk of asthma, including severe asthma. However, how UPRs participate in the deregulation of TH17 cells leading to this type of asthma remains elusive. In this study, we investigated the role of the UPR sensor IRE1 in TH17 cell function and neutrophilic airway inflammation. We found that IRE1 is induced in fungal asthma and is highly expressed in TH17 cells relative to naïve CD4+ T cells. Cytokine (e.g. IL-23) signals induce the IRE1-XBP1s axis in a JAK2-dependent manner. This noncanonical activation of the IRE1-XBP1s pathway promotes UPRs and cytokine secretion by TH17 cells. Ern1 (encoding IRE1)-deficiency decreases the expression of ER stress factors and impairs the differentiation and cytokine secretion of TH17 cells. Genetic ablation of Ern1 leads to alleviated TH17 responses and airway neutrophilia in a Candida albicans asthma model. Consistently, IL-23 activates the JAK2-IRE1-XBP1s pathway in vivo and enhances TH17 responses and neutrophilic infiltration into the airway. Taken together, our data indicate that IRE1, noncanonically activated by cytokine signals, promotes neutrophilic airway inflammation through the UPRmediated secretory function of TH17 cells. The findings provide a novel insight into the fundamental understanding of IRE1 in TH17-biased TH2-low asthma.
Collapse
Affiliation(s)
- Dandan Wu
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Xing Zhang
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Kourtney M. Zimmerly
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Ruoning Wang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Amanda Livingston
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Takao Iwawaki
- Division of Cell Medicine, Medical Research Institute, Kanazawa Medical University, Ishikawa 920-0293, Japan
| | - Ashok Kumar
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77204, USA
| | - Xiang Wu
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
- Department of Parasitology, School of Basic Medical Sciences, Xiangya School of Medicine, Central South University, Changsha, China
| | - Michael A. Mandell
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Meilian Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Xuexian O. Yang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| |
Collapse
|
24
|
Boboltz A, Kumar S, Duncan GA. Inhaled drug delivery for the targeted treatment of asthma. Adv Drug Deliv Rev 2023; 198:114858. [PMID: 37178928 PMCID: PMC10330872 DOI: 10.1016/j.addr.2023.114858] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/14/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023]
Abstract
Asthma is a chronic lung disease affecting millions worldwide. While classically acknowledged to result from allergen-driven type 2 inflammatory responses leading to IgE and cytokine production and the influx of immune cells such as mast cells and eosinophils, the wide range in asthmatic pathobiological subtypes lead to highly variable responses to anti-inflammatory therapies. Thus, there is a need to develop patient-specific therapies capable of addressing the full spectrum of asthmatic lung disease. Moreover, delivery of targeted treatments for asthma directly to the lung may help to maximize therapeutic benefit, but challenges remain in design of effective formulations for the inhaled route. In this review, we discuss the current understanding of asthmatic disease progression as well as genetic and epigenetic disease modifiers associated with asthma severity and exacerbation of disease. We also overview the limitations of clinically available treatments for asthma and discuss pre-clinical models of asthma used to evaluate new therapies. Based on the shortcomings of existing treatments, we highlight recent advances and new approaches to treat asthma via inhalation for monoclonal antibody delivery, mucolytic therapy to target airway mucus hypersecretion and gene therapies to address underlying drivers of disease. Finally, we conclude with discussion on the prospects for an inhaled vaccine to prevent asthma.
Collapse
Affiliation(s)
- Allison Boboltz
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, United States
| | - Sahana Kumar
- Biological Sciences Graduate Program, University of Maryland, College Park, MD 20742, United States
| | - Gregg A Duncan
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, United States; Biological Sciences Graduate Program, University of Maryland, College Park, MD 20742, United States.
| |
Collapse
|
25
|
Nørgaard SK, Følsgaard N, Vissing NH, Kyvsgaard JN, Chawes B, Stokholm J, Smilde AK, Bønnelykke K, Bisgaard H, Rasmussen MA. Novel Connections of Common Childhood Illnesses Based on More Than 5 Million Diary Registrations From Birth Until Age 3 Years. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:2162-2171.e6. [PMID: 37146879 DOI: 10.1016/j.jaip.2023.04.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/04/2023] [Accepted: 04/11/2023] [Indexed: 05/07/2023]
Abstract
BACKGROUND All children experience numerous episodes of illness during the first 3 years of life. Most episodes are mild and handled without medical attention but nevertheless burden the families and society. There is a large, and still unexplained, variation in the burden of illness between children. OBJECTIVE To describe and provide a better understanding of the disease burden of common childhood diseases through a data-driven approach investigating the communalities between symptom patterns and predefined variables on predispositions, pregnancy, birth, environment, and child development. METHODS The study is based on the prospectively followed clinical mother-child cohort COpenhagen Prospective Studies on Asthma in Childhood, which includes 700 children with daily symptom registration in the first 3 years of life, including symptoms of cough, breathlessness, wheeze, cold, pneumonia, sore throat, ear infections, gastrointestinal infections, fever, and eczema. First, we described the number of episodes of symptoms. Next, factor analysis models were used to describe the variation in symptom load in the second year of life (both based on n = 556, with >90% complete diary). Then we characterized patterns of similarity between symptoms using a graphical network model (based on n = 403, with a 3-year monthly compliance of >50%). Finally, predispositions and pregnancy, birth, environmental, and developmental factors were added to the network model. RESULTS The children experienced a median of 17 (interquartile range, 12-23) episodes of symptoms during the first 3 years of life, of which most were respiratory tract infections (median, 13; interquartile range, 9-18). The frequency of symptoms was the highest during the second year of life. Eczema symptoms were unrelated to the other symptoms. The strongest association to respiratory symptoms was found for maternal asthma, maternal smoking during the third trimester, prematurity, and CDHR3 genotype. This was in contrast to the lack of associations for the well-established asthma locus at 17q21. CONCLUSIONS Healthy young children are burdened by multiple episodes of symptoms during the first 3 years of life. Prematurity, maternal asthma, and CDHR3 genotype were among the strongest drivers of symptom burden.
Collapse
Affiliation(s)
- Sarah Kristine Nørgaard
- COPSAC (COpenhagen Prospective Studies on Asthma in Childhood), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Nilo Følsgaard
- COPSAC (COpenhagen Prospective Studies on Asthma in Childhood), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Nadja Hawwa Vissing
- COPSAC (COpenhagen Prospective Studies on Asthma in Childhood), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Pediatrics and Adolescence Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Julie Nyholm Kyvsgaard
- COPSAC (COpenhagen Prospective Studies on Asthma in Childhood), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Bo Chawes
- COPSAC (COpenhagen Prospective Studies on Asthma in Childhood), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Stokholm
- COPSAC (COpenhagen Prospective Studies on Asthma in Childhood), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Pediatrics, Slagelse Hospital, Slagelse, Denmark; Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| | - Age K Smilde
- Department of Food Science, University of Copenhagen, Copenhagen, Denmark; Biosystems Data Analysis, Swammerdam Institute for Life Sciences, Amsterdam, The Netherlands
| | - Klaus Bønnelykke
- COPSAC (COpenhagen Prospective Studies on Asthma in Childhood), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark.
| | - Hans Bisgaard
- COPSAC (COpenhagen Prospective Studies on Asthma in Childhood), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Morten Arendt Rasmussen
- COPSAC (COpenhagen Prospective Studies on Asthma in Childhood), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Food Science, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
26
|
Shamsi BH, Chen H, Yang X, Liu M, Liu Y. Association between polymorphisms of the GSDMB gene and allergic rhinitis risk in the Chinese population: a case-control study. J Asthma 2023:1-10. [PMID: 36847643 DOI: 10.1080/02770903.2023.2185893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
BACKGROUND Allergic rhinitis (AR) is a great risk factor for developing asthma, and its pathogenesis is affected by various factors, such as gene and environment. GSDMB is related to allergic diseases. Our purpose is to explore the correlation of single nucleotide polymorphisms (SNPs) in GSDMB and AR risk in the Chinese population. METHODS We performed a case-control study including 1005 cases and 1004 controls. Rs2305479, rs4795400, and rs12450091 in GSDMB were geneotyped using Agena MassARRAY. The relationships between GSDMB SNPs and AR risk were assessed by logistic regression analysis in PLINK1.9. RESULTS Our study showed that rs4795400 was a protective factor for AR in overall (TT vs. CC: OR = 0.66, p = 0.009; TT vs. CC/TC: OR = 0.67, p = 0.008; additive: OR = 0.87, p = 0.042 males, people with BMI ≤ 24, and living in wind-blown sand area. Rs2305479 was associated with a reduced AR risk in males (TT vs. CC: OR = 0.47, p = 0.014; TT vs. CC/TC: OR = 0.43, p = 0.004). However, rs12450091 was a risk factor for AR in people living in the loess hilly region (CC: OR = 4.75, p = 0.047). The levels of EO and EO_per in the case group were significantly higher than those in the control group (p < 0.05). CONCLUSION This study indicated that GSDMB polymorphisms (rs4795400, rs2305479, and rs12450091) were associated with AR susceptibility. Further studies are required to confirm our findings and to clarify the functional relationship.
Collapse
Affiliation(s)
- Bilal Haider Shamsi
- Department of Science and Education, Shenmu Hospital, The Affiliated Shenmu Hospital of Northwest University, Shenmu, China
| | - Haiyuan Chen
- Department of Information, Shenmu Hospital, The Affiliated Shenmu Hospital of Northwest University, Shenmu, China
| | - Xiong Yang
- Department of Nephrology, Shenmu Hospital, The Affiliated Shenmu Hospital of Northwest University, Shenmu, China
| | - Mingxia Liu
- Department of Prevention and Protection, Shenmu Hospital, The Affiliated Shenmu Hospital of Northwest University, Shenmu, China
| | - Yonglin Liu
- Department of Science and Education, Shenmu Hospital, The Affiliated Shenmu Hospital of Northwest University, Shenmu, China
| |
Collapse
|
27
|
What Have Mechanistic Studies Taught Us About Childhood Asthma? THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:684-692. [PMID: 36649800 DOI: 10.1016/j.jaip.2023.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023]
Abstract
Childhood asthma is a chronic heterogeneous syndrome consisting of different disease entities or phenotypes. The immunologic and cellular processes that occur during asthma development are still not fully understood but represent distinct endotypes. Mechanistic studies have examined the role of gene expression, protein levels, and cell types in early life development and the manifestation of asthma, many under the influence of environmental stimuli, which can be both protective and risk factors for asthma. Genetic variants can regulate gene expression, controlled partly by different epigenetic mechanisms. In addition, environmental factors, such as living space, nutrition, and smoking, can contribute to these mechanisms. All of these factors produce modifications in gene expression that can alter the development and function of immune and epithelial cells and subsequently different trajectories of childhood asthma. These early changes in a partially immature immune system can have dramatic effects (e.g., causing dysregulation), which in turn contribute to different disease endotypes and may help to explain differential responsiveness to asthma treatment. In this review, we summarize published studies that have aimed to uncover distinct mechanisms in childhood asthma, considering genetics, epigenetics, and environment. Moreover, a discussion of new, powerful tools for single-cell immunologic assays for phenotypic and functional analysis is included, which promise new mechanistic insights into childhood asthma development and therapeutic and preventive strategies.
Collapse
|
28
|
Bisgaard H, Chawes B, Stokholm J, Mikkelsen M, Schoos AMM, Bønnelykke K. 25 Years of translational research in the Copenhagen Prospective Studies on Asthma in Childhood (COPSAC). J Allergy Clin Immunol 2023; 151:619-633. [PMID: 36642652 DOI: 10.1016/j.jaci.2022.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 10/28/2022] [Accepted: 11/04/2022] [Indexed: 01/15/2023]
Abstract
The Copenhagen Prospective Studies on Asthma in Childhood (COPSAC) mother-child cohorts have provided a foundation of 25 years of research on the origins, prevention, and natural history of childhood asthma and related disorders. COPSAC's approach is characterized by clinical translational research with longitudinal deep phenotyping and exposure assessments from pregnancy, in combination with multi-omic data layers and embedded randomized controlled trials. One trial showed that fish oil supplementation during pregnancy prevented childhood asthma and identified pregnant women with the highest benefits from supplementation, thereby creating the potential for personalized prevention. COPSAC revealed that airway colonization with pathogenic bacteria in early life is associated with an increased risk of asthma. Further, airway bacteria were shown to be a trigger of acute asthma-like symptoms, with benefit from antibiotic treatment. COPSAC identified an immature gut microbiome in early life as a risk factor for asthma and allergy and further demonstrated that asthma can be predicted by infant lung function. At a molecular level, COPSAC has identified novel susceptibility genes, early immune deviations, and metabolomic alterations associated with childhood asthma. Thus, the COPSAC research program has enhanced our understanding of the processes causing childhood asthma and has suggested means of personalized prevention and treatment.
Collapse
Affiliation(s)
- Hans Bisgaard
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Bo Chawes
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Stokholm
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Pediatrics, Slagelse Hospital, Slagelse, Denmark
| | - Marianne Mikkelsen
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Ann-Marie Malby Schoos
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Pediatrics, Slagelse Hospital, Slagelse, Denmark
| | - Klaus Bønnelykke
- Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
29
|
Tutino M, Granell R, Curtin JA, Haider S, Fontanella S, Murray CS, Roberts G, Arshad SH, Turner S, Morris AP, Custovic A, Simpson A. Dog ownership in infancy is protective for persistent wheeze in 17q21 asthma-risk carriers. J Allergy Clin Immunol 2023; 151:423-430. [PMID: 36273658 DOI: 10.1016/j.jaci.2022.10.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Asthma-associated single nucleotide polymorphisms from large genome-wide association studies only explain a fraction of genetic heritability. Likely causes of the missing heritability include broad phenotype definitions and gene-environment interactions (GxE). The mechanisms underlying GxE in asthma are poorly understood. Previous GxE studies on pet ownership showed discordant results. OBJECTIVES We sought to study the GxE between the 17q12-21 locus and pet ownership in infancy in relation to wheeze. METHODS Wheezing classes derived from 5 UK-based birth cohorts (latent class analysis) were used to study GxE between the 17q12-21 asthma-risk variant rs2305480 and dog and cat ownership in infancy, using multinomial logistic regression. A total of 9149 children had both pet ownership and genotype data available. Summary statistics from individual analyses were meta-analyzed. RESULTS rs2305480 G allele was associated with increased risk of persistent wheeze (additive model odds ratio, 1.37; 95% CI, 1.25-1.51). There was no evidence of an association between dog or cat ownership and wheeze. We found significant evidence of a GxE interaction between rs2305480 and dog ownership (P = 8.3 × 10-4) on persistent wheeze; among dog owners, the G allele was no longer associated with an increased risk of persistent wheeze (additive model odds ratio, 0.95; 95% CI, 0.73-1.24). For those without pets, G allele was associated with increased risk of persistent wheeze (odds ratio, 1.61; 95% CI, 1.40-1.86). Among cat owners, no such dampening of the genetic effect was observed. CONCLUSIONS Among dog owners, rs2305480 G was no longer associated with an increased risk of persistent wheeze (or asthma). Early-life environmental exposures may therefore attenuate likelihood of asthma in those carrying 17q12-21 risk alleles.
Collapse
Affiliation(s)
- Mauro Tutino
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester Academic Health Science Centre, and Manchester University NHS Foundation Trust, Manchester, United Kingdom.
| | - Raquel Granell
- MRC Integrative Epidemiology Unit, Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - John A Curtin
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester Academic Health Science Centre, and Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Sadia Haider
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Sara Fontanella
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Clare S Murray
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester Academic Health Science Centre, and Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Graham Roberts
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom; NIHR Southampton Biomedical Research Centre, University Hospitals Southampton NHS Foundation Trust, Southampton, United Kingdom; David Hide Asthma and Allergy Research Centre, Isle of Wight, United Kingdom
| | - S Hasan Arshad
- NIHR Southampton Biomedical Research Centre, University Hospitals Southampton NHS Foundation Trust, Southampton, United Kingdom; David Hide Asthma and Allergy Research Centre, Isle of Wight, United Kingdom; Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Stephen Turner
- Child Health, University of Aberdeen, Aberdeen, United Kingdom
| | - Andrew P Morris
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, The University of Manchester, Manchester, United Kingdom
| | - Adnan Custovic
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Angela Simpson
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester Academic Health Science Centre, and Manchester University NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
30
|
Hizawa N. The understanding of asthma pathogenesis in the era of precision medicine. Allergol Int 2023; 72:3-10. [PMID: 36195530 DOI: 10.1016/j.alit.2022.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 08/30/2022] [Indexed: 01/25/2023] Open
Abstract
Asthma is a syndrome with extremely diverse clinical phenotypes in which the onset, severity, and response to treatment are defined by the complex interplay of many genetic and environmental factors. Environmental factors epigenetically affect gene expression, and the disease is driven by a multidimensional dynamic network involving RNA and protein molecules derived from gene expression, as well as various metabolic products. In other words, specific pathophysiological mechanisms or endotypes are dynamic networks that arise in response to individual genotypes and the various environmental factors to which individuals have been exposed since before birth, such as diet, infection, air pollution, smoking, antibiotic use, and the bacterial flora of the intestinal tract, skin, and lungs. A key feature of asthma genome scans is their potential to reveal the molecular pathways that lead to pathogenesis. Endotypes that drive the disease have a significant impact on the phenotypes of asthma patients, including their drug responsiveness. Understanding endotypes will lead to not only the implementation of therapies that are tailored to the specific molecular network(s) underlying the patient's condition, but also to the development of therapeutic strategies that target individual endotypes, as well as to precision health, which will enable the prediction of disease onset with high accuracy from an early stage and the implementation of preventive strategies based on endotypes. Understanding of endotypes will pave the way for the practice of precision medicine in asthma care, moving away from 'one-size-fits-all' medicine and population-based prevention approaches that do not take individuals' susceptibility into account.
Collapse
Affiliation(s)
- Nobuyuki Hizawa
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.
| |
Collapse
|
31
|
Discerning asthma endotypes through comorbidity mapping. Nat Commun 2022; 13:6712. [PMID: 36344522 PMCID: PMC9640644 DOI: 10.1038/s41467-022-33628-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 09/27/2022] [Indexed: 11/09/2022] Open
Abstract
Asthma is a heterogeneous, complex syndrome, and identifying asthma endotypes has been challenging. We hypothesize that distinct endotypes of asthma arise in disparate genetic variation and life-time environmental exposure backgrounds, and that disease comorbidity patterns serve as a surrogate for such genetic and exposure variations. Here, we computationally discover 22 distinct comorbid disease patterns among individuals with asthma (asthma comorbidity subgroups) using diagnosis records for >151 M US residents, and re-identify 11 of the 22 subgroups in the much smaller UK Biobank. GWASs to discern asthma risk loci for individuals within each subgroup and in all subgroups combined reveal 109 independent risk loci, of which 52 are replicated in multi-ancestry meta-analysis across different ethnicity subsamples in UK Biobank, US BioVU, and BioBank Japan. Fourteen loci confer asthma risk in multiple subgroups and in all subgroups combined. Importantly, another six loci confer asthma risk in only one subgroup. The strength of association between asthma and each of 44 health-related phenotypes also varies dramatically across subgroups. This work reveals subpopulations of asthma patients distinguished by comorbidity patterns, asthma risk loci, gene expression, and health-related phenotypes, and so reveals different asthma endotypes.
Collapse
|
32
|
Schoettler N, Dissanayake E, Craven MW, Yee JS, Eliason J, Schauberger EM, Lemanske RF, Ober C, Gern JE. New Insights Relating Gasdermin B to the Onset of Childhood Asthma. Am J Respir Cell Mol Biol 2022; 67:430-437. [PMID: 35580164 PMCID: PMC9564923 DOI: 10.1165/rcmb.2022-0043ps] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/17/2022] [Indexed: 02/06/2023] Open
Abstract
Chromosome 17q12-q21 is the most replicated genetic locus for childhood-onset asthma. Polymorphisms in this locus containing ∼10 genes interact with a variety of environmental exposures in the home and outdoors to modify asthma risk. However, the functional basis for these associations and their linkages to the environment have remained enigmatic. Within this extended region, regulation of GSDMB (gasdermin B) expression in airway epithelial cells has emerged as the primary mechanism underlying the 17q12-q21 genome-wide association study signal. Asthma-associated SNPs influence the abundance of GSDMB transcripts as well as the functional properties of GSDMB protein in airway epithelial cells. GSDMB is a member of the gasdermin family of proteins, which regulate pyroptosis and inflammatory responses to microbial infections. The aims of this review are to synthesize recent studies on the relationship of 17q12-q21 SNPs to childhood asthma and the evidence pointing to GSDMB gene expression or protein function as the underlying mechanism and to explore the potential functions of GSDMB that may influence the risk of developing asthma during childhood.
Collapse
Affiliation(s)
| | | | - Mark W. Craven
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Jeremiah S. Yee
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Joshua Eliason
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | | | | | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, Illinois; and
| | | |
Collapse
|
33
|
Washington C, Dapas M, Biddanda A, Magnaye KM, Aneas I, Helling BA, Szczesny B, Boorgula MP, Taub MA, Kenny E, Mathias RA, Barnes KC, Khurana Hershey GK, Kercsmar CM, Gereige JD, Makhija M, Gruchalla RS, Gill MA, Liu AH, Rastogi D, Busse W, Gergen PJ, Visness CM, Gold DR, Hartert T, Johnson CC, Lemanske RF, Martinez FD, Miller RL, Ownby D, Seroogy CM, Wright AL, Zoratti EM, Bacharier LB, Kattan M, O'Connor GT, Wood RA, Nobrega MA, Altman MC, Jackson DJ, Gern JE, McKennan CG, Ober C. African-specific alleles modify risk for asthma at the 17q12-q21 locus in African Americans. Genome Med 2022; 14:112. [PMID: 36175932 PMCID: PMC9520885 DOI: 10.1186/s13073-022-01114-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 09/15/2022] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Asthma is the most common chronic disease in children, occurring at higher frequencies and with more severe disease in children with African ancestry. METHODS We tested for association with haplotypes at the most replicated and significant childhood-onset asthma locus at 17q12-q21 and asthma in European American and African American children. Following this, we used whole-genome sequencing data from 1060 African American and 100 European American individuals to identify novel variants on a high-risk African American-specific haplotype. We characterized these variants in silico using gene expression and ATAC-seq data from airway epithelial cells, functional annotations from ENCODE, and promoter capture (pc)Hi-C maps in airway epithelial cells. Candidate causal variants were then assessed for correlation with asthma-associated phenotypes in African American children and adults. RESULTS Our studies revealed nine novel African-specific common variants, enriched on a high-risk asthma haplotype, which regulated the expression of GSDMA in airway epithelial cells and were associated with features of severe asthma. Using ENCODE annotations, ATAC-seq, and pcHi-C, we narrowed the associations to two candidate causal variants that are associated with features of T2 low severe asthma. CONCLUSIONS Previously unknown genetic variation at the 17q12-21 childhood-onset asthma locus contributes to asthma severity in individuals with African ancestries. We suggest that many other population-specific variants that have not been discovered in GWAS contribute to the genetic risk for asthma and other common diseases.
Collapse
Affiliation(s)
- Charles Washington
- Department of Human Genetics, The University of Chicago, 928 E. 58th St. CLSC 507C, Chicago, IL, 60637, USA
| | - Matthew Dapas
- Department of Human Genetics, The University of Chicago, 928 E. 58th St. CLSC 507C, Chicago, IL, 60637, USA
| | - Arjun Biddanda
- Department of Human Genetics, The University of Chicago, 928 E. 58th St. CLSC 507C, Chicago, IL, 60637, USA
| | - Kevin M Magnaye
- Department of Human Genetics, The University of Chicago, 928 E. 58th St. CLSC 507C, Chicago, IL, 60637, USA
| | - Ivy Aneas
- Department of Human Genetics, The University of Chicago, 928 E. 58th St. CLSC 507C, Chicago, IL, 60637, USA
| | - Britney A Helling
- Department of Human Genetics, The University of Chicago, 928 E. 58th St. CLSC 507C, Chicago, IL, 60637, USA
| | - Brooke Szczesny
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | | | - Margaret A Taub
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Eimear Kenny
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rasika A Mathias
- Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Kathleen C Barnes
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA
| | | | - Carolyn M Kercsmar
- Division of Asthma Research, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Jessica D Gereige
- Department of Medicine, Division of Pulmonary, Allergy, Sleep, and Critical Care Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Melanie Makhija
- Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | | | - Michelle A Gill
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andrew H Liu
- Children's Hospital Colorado and University of Colorado School of Medicine, Aurora, CO, USA
| | - Deepa Rastogi
- Children's National Hospital and George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - William Busse
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | | | - Diane R Gold
- The Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Tina Hartert
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Christine C Johnson
- Department of Public Health Sciences, Henry Ford Health Systems, Detroit, MI, USA
| | - Robert F Lemanske
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Fernando D Martinez
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, USA
| | - Rachel L Miller
- Department of Medicine, Division of Clinical Immunology Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dennis Ownby
- Department of Public Health Sciences, Henry Ford Health Systems, Detroit, MI, USA
| | - Christine M Seroogy
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Anne L Wright
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, USA
| | - Edward M Zoratti
- Department of Medicine, Henry Ford Health Systems, Detroit, MI, USA
| | - Leonard B Bacharier
- Department of Pediatrics, Monroe Carell Jr Children's Hospital at Vanderbilt University Medical Center, Nashville, TN, USA
| | - Meyer Kattan
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - George T O'Connor
- Pulmonary Center, Boston University School of Medicine, Boston, MA, USA
| | - Robert A Wood
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD, USA
| | - Marcelo A Nobrega
- Department of Human Genetics, The University of Chicago, 928 E. 58th St. CLSC 507C, Chicago, IL, 60637, USA
| | - Matthew C Altman
- Immunology Division, Benaroya Research Institute Systems, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Daniel J Jackson
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - James E Gern
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | - Carole Ober
- Department of Human Genetics, The University of Chicago, 928 E. 58th St. CLSC 507C, Chicago, IL, 60637, USA.
| |
Collapse
|
34
|
Kulkarni A, Kediya DA. A Multi-Point View of Genetic Factors Affecting Hereditary Transmissibility of Asthma. Cureus 2022; 14:e28768. [PMID: 36225476 PMCID: PMC9531716 DOI: 10.7759/cureus.28768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 09/04/2022] [Indexed: 11/05/2022] Open
|
35
|
Eliasen AU, Pedersen CET, Rasmussen MA, Wang N, Soverini M, Fritz A, Stokholm J, Chawes BL, Morin A, Bork-Jensen J, Grarup N, Pedersen O, Hansen T, Linneberg A, Mortensen PB, Hougaard DM, Bybjerg-Grauholm J, Bækvad-Hansen M, Mors O, Nordentoft M, Børglum AD, Werge T, Agerbo E, Söderhall C, Altman MC, Thysen AH, McKennan CG, Brix S, Gern JE, Ober C, Ahluwalia TS, Bisgaard H, Pedersen AG, Bønnelykke K. Genome-wide study of early and severe childhood asthma identifies interaction between CDHR3 and GSDMB. J Allergy Clin Immunol 2022; 150:622-630. [PMID: 35381269 DOI: 10.1016/j.jaci.2022.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 03/11/2022] [Accepted: 03/18/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Asthma with severe exacerbation is one of the most common causes of hospitalization among young children. Exacerbations are typically triggered by respiratory infections, but the host factors causing recurrent infections and exacerbations in some children are poorly understood. As a result, current treatment options and preventive measures are inadequate. OBJECTIVE We sought to identify genetic interaction associated with the development of childhood asthma. METHODS We performed an exhaustive search for pairwise interaction between genetic single nucleotide polymorphisms using 1204 cases of a specific phenotype of early childhood asthma with severe exacerbations in patients aged 2 to 6 years combined with 5328 nonasthmatic controls. Replication was attempted in 3 independent populations, and potential underlying immune mechanisms were investigated in the COPSAC2010 and COPSAC2000 birth cohorts. RESULTS We found evidence of interaction, including replication in independent populations, between the known childhood asthma loci CDHR3 and GSDMB. The effect of CDHR3 was dependent on the GSDMB genotype, and this interaction was more pronounced for severe and early onset of disease. Blood immune analyses suggested a mechanism related to increased IL-17A production after viral stimulation. CONCLUSIONS We found evidence of interaction between CDHR3 and GSDMB in development of early childhood asthma, possibly related to increased IL-17A response to viral infections. This study demonstrates the importance of focusing on specific disease subtypes for understanding the genetic mechanisms of asthma.
Collapse
Affiliation(s)
- Anders U Eliasen
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Health Technology, Section for Bioinformatics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Casper Emil T Pedersen
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Morten A Rasmussen
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Food Science, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Ni Wang
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Matteo Soverini
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Amelie Fritz
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Health Technology, Section for Bioinformatics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Jakob Stokholm
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Bo L Chawes
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Andréanne Morin
- Department of Human Genetics, University of Chicago, Chicago, Ill
| | - Jette Bork-Jensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Copenhagen, Denmark
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Copenhagen, Denmark
| | - Allan Linneberg
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, The Capital Region, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Preben B Mortensen
- iPSYCH, The Lundbeck Foundation Initiative for Integrated Psychiatric Research, Aarhus, Denmark; National Center for Register-Based Research (NCRR), Business and Social Sciences, Aarhus University, Aarhus, Denmark; Center for Integrated Register-Based Research (CIRRAU), Aarhus University, Aarhus, Denmark
| | - David M Hougaard
- iPSYCH, The Lundbeck Foundation Initiative for Integrated Psychiatric Research, Aarhus, Denmark; Department for Congenital Disorders, Statens Serum Institut (SSI), Copenhagen, Denmark; Den Neonatale Screenings Biobank, SSI, Copenhagen, Denmark
| | - Jonas Bybjerg-Grauholm
- iPSYCH, The Lundbeck Foundation Initiative for Integrated Psychiatric Research, Aarhus, Denmark; Den Neonatale Screenings Biobank, SSI, Copenhagen, Denmark
| | - Marie Bækvad-Hansen
- iPSYCH, The Lundbeck Foundation Initiative for Integrated Psychiatric Research, Aarhus, Denmark; Department for Congenital Disorders, Statens Serum Institut (SSI), Copenhagen, Denmark
| | - Ole Mors
- iPSYCH, The Lundbeck Foundation Initiative for Integrated Psychiatric Research, Aarhus, Denmark; Psychosis Research Unit, Aarhus University Hospital-Psychiatry, Risskov, Denmark
| | - Merete Nordentoft
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; iPSYCH, The Lundbeck Foundation Initiative for Integrated Psychiatric Research, Aarhus, Denmark; Mental Health Center Copenhagen, Capital Region of Denmark, Copenhagen University Hospital, Copenhagen, Denmark
| | - Anders D Børglum
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; iPSYCH, The Lundbeck Foundation Initiative for Integrated Psychiatric Research, Aarhus, Denmark; Center for Integrative Sequencing, Department of Biomedicine and iSEQ, Aarhus University, Aarhus, Denmark
| | - Thomas Werge
- iPSYCH, The Lundbeck Foundation Initiative for Integrated Psychiatric Research, Aarhus, Denmark; Institute of Biological Psychiatry, Copenhagen Mental Health Services, Copenhagen, Denmark; Institute of Clinical Medicine and GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| | - Esben Agerbo
- iPSYCH, The Lundbeck Foundation Initiative for Integrated Psychiatric Research, Aarhus, Denmark; National Center for Register-Based Research (NCRR), Business and Social Sciences, Aarhus University, Aarhus, Denmark; Center for Integrated Register-Based Research (CIRRAU), Aarhus University, Aarhus, Denmark
| | - Cilla Söderhall
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Wash
| | - Matthew C Altman
- Department of Medicine, University of Washington, Seattle, Sweden
| | - Anna H Thysen
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark; Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Chris G McKennan
- Department of Statistics, University of Pittsburgh, Pittsburgh, Pa
| | - Susanne Brix
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - James E Gern
- Department of Pediatrics, University of Wisconsin, Madison, Wis
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, Ill
| | - Tarunveer S Ahluwalia
- Steno Diabetes Center Copenhagen, Gentofte, Denmark; Bioinformatics Center, University of Copenhagen, Copenhagen, Denmark
| | - Hans Bisgaard
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Anders G Pedersen
- Department of Health Technology, Section for Bioinformatics, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Klaus Bønnelykke
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
36
|
Augustine T, Al-Aghbar MA, Al-Kowari M, Espino-Guarch M, van Panhuys N. Asthma and the Missing Heritability Problem: Necessity for Multiomics Approaches in Determining Accurate Risk Profiles. Front Immunol 2022; 13:822324. [PMID: 35693821 PMCID: PMC9174795 DOI: 10.3389/fimmu.2022.822324] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 04/25/2022] [Indexed: 11/20/2022] Open
Abstract
Asthma is ranked among the most common chronic conditions and has become a significant public health issue due to the recent and rapid increase in its prevalence. Investigations into the underlying genetic factors predict a heritable component for its incidence, estimated between 35% and 90% of causation. Despite the application of large-scale genome-wide association studies (GWAS) and admixture mapping approaches, the proportion of variants identified accounts for less than 15% of the observed heritability of the disease. The discrepancy between the predicted heritable component of disease and the proportion of heritability mapped to the currently identified susceptibility loci has been termed the ‘missing heritability problem.’ Here, we examine recent studies involving both the analysis of genetically encoded features that contribute to asthma and also the role of non-encoded heritable characteristics, including epigenetic, environmental, and developmental aspects of disease. The importance of vertical maternal microbiome transfer and the influence of maternal immune factors on fetal conditioning in the inheritance of disease are also discussed. In order to highlight the broad array of biological inputs that contribute to the sum of heritable risk factors associated with allergic disease incidence that, together, contribute to the induction of a pro-atopic state. Currently, there is a need to develop in-depth models of asthma risk factors to overcome the limitations encountered in the interpretation of GWAS results in isolation, which have resulted in the missing heritability problem. Hence, multiomics analyses need to be established considering genetic, epigenetic, and functional data to create a true systems biology-based approach for analyzing the regulatory pathways that underlie the inheritance of asthma and to develop accurate risk profiles for disease.
Collapse
Affiliation(s)
- Tracy Augustine
- Laboratory of Immunoregulation, Systems Biology and Immunology Department, Sidra Medicine, Doha, Qatar
| | - Mohammad Ameen Al-Aghbar
- Laboratory of Immunoregulation, Systems Biology and Immunology Department, Sidra Medicine, Doha, Qatar
| | - Moza Al-Kowari
- Laboratory of Immunoregulation, Systems Biology and Immunology Department, Sidra Medicine, Doha, Qatar
| | - Meritxell Espino-Guarch
- Laboratory of Immunoregulation, Systems Biology and Immunology Department, Sidra Medicine, Doha, Qatar
| | - Nicholas van Panhuys
- Laboratory of Immunoregulation, Systems Biology and Immunology Department, Sidra Medicine, Doha, Qatar
| |
Collapse
|
37
|
Li L, Holloway JW, Ewart S, Arshad SH, Relton CL, Karmaus W, Zhang H. Newborn DNA methylation and asthma acquisition across adolescence and early adulthood. Clin Exp Allergy 2022; 52:658-669. [PMID: 34995380 PMCID: PMC9050758 DOI: 10.1111/cea.14091] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/16/2021] [Accepted: 01/03/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Little is known about the association of newborn DNA methylation (DNAm) with asthma acquisition across adolescence and early adult life. OBJECTIVE We aim to identify epigenetic biomarkers in newborns for asthma acquisition during adolescence or young adulthood. METHODS The Isle of Wight Birth Cohort (IOWBC) (n = 1456) data at ages 10, 18 and 26 years were assessed. To screen cytosine-phosphate-guanine site (CpGs) potentially associated with asthma acquisition, at the genome scale, we examined differentially methylated regions (DMR) using dmrff R package and individual CpG sites using linear regression on such associations. For CpGs that passed screening, we examined their enrichment in biological pathways using their mapping genes and tested their associations with asthma acquisitions using logistic regressions. Findings in IOWBC were tested in an independent cohort, the Avon Longitudinal Study of Parents and Children (ALSPAC) cohort. RESULTS In total, 2636 unique CpGs passed screening, based on which we identified one biological pathway linked to asthma acquisition during adolescence in females (FDR adjusted p-value = .003 in IOWBC). Via logistic regressions, for females, four CpGs were shown to be associated with asthma acquisition during adolescence, and another four CpGs with asthma acquisition in young adulthood (FDR adjusted p-value < .05 in IOWBC) and these eight CpGs were replicated in ALSPAC (all p-values < .05). DNAm at all the identified CpGs was shown to be temporally consistent, and at six of the CpGs was associated with expressions of adjacent or mapping genes in females (all p-values < .05). For males, 622 CpGs were identified in IOWBC (FDR = 0.01), but these were not tested in ALSPAC due to small sample sizes. CONCLUSION AND CLINICAL RELEVANCE Eight CpGs on LHX5, IL22RA2, SOX11, CBX4, ACPT, CFAP46, MUC4, and ATP1B2 genes have the potential to serve as candidate epigenetic biomarkers in newborns for asthma acquisition in females during adolescence or young adulthood.
Collapse
Affiliation(s)
- Liang Li
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA
| | - John W. Holloway
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Susan Ewart
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - S. Hasan Arshad
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
- The David Hide Asthma and Allergy Research Centre, St Mary’s, Hospital, Parkhurst Road, Newport, Isle of Wight PO30 5TG, UK
| | - Caroline L. Relton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 2BN, UK
| | - Wilfried Karmaus
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA
| | - Hongmei Zhang
- Division of Epidemiology, Biostatistics, and Environmental Health, School of Public Health, University of Memphis, Memphis, TN, USA
| |
Collapse
|
38
|
Hernandez-Pacheco N, Kere M, Melén E. Gene-environment interactions in childhood asthma revisited; expanding the interaction concept. Pediatr Allergy Immunol 2022; 33:e13780. [PMID: 35616899 PMCID: PMC9325482 DOI: 10.1111/pai.13780] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 04/13/2022] [Indexed: 01/04/2023]
Abstract
Investigation of gene-environment interactions (GxE) may provide important insights into the gene regulatory framework in response to environmental factors of relevance for childhood asthma. Over the years, different methodological strategies have been applied, more recently using genome-wide approaches. The best example to date is the major asthma locus on the 17q12-21 chromosome region, viral infections, and airway epithelium processes where recent studies have shed much light on mechanisms in childhood asthma. However, there are challenges with the traditional single variant-single exposure interaction models, as they do not encompass the complexity and cumulative effects of multiple exposures or multiple genetic variants. As such, we need to redefine our traditional GxE thinking, and we propose in this review to expand the GxE concept by also evaluating other omics layers, such as epigenetics, transcriptomics, metabolomics, and proteomics. In addition, host factors such as age, gender, and other exposures are very likely to influence GxE effects and need firmly to be considered in future studies.
Collapse
Affiliation(s)
- Natalia Hernandez-Pacheco
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden.,CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Maura Kere
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Erik Melén
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden.,Sachs' Children's Hospital, South General Hospital, Stockholm, Sweden
| |
Collapse
|
39
|
Chen R, Michaeloudes C, Liang Y, Bhavsar PK, Chung KF, Ip MSM, Mak JCW. ORMDL3 regulates cigarette smoke-induced endoplasmic reticulum stress in airway smooth muscle cells. J Allergy Clin Immunol 2022; 149:1445-1457.e5. [PMID: 34624393 DOI: 10.1016/j.jaci.2021.09.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/19/2021] [Accepted: 09/29/2021] [Indexed: 01/12/2023]
Abstract
BACKGROUND Orosomucoid 1-like protein 3 (ORMDL3), a transmembrane protein localized in the endoplasmic reticulum (ER), has been genetically associated with chronic obstructive pulmonary disease (COPD), in addition to childhood-onset asthma. However, the functional role of ORMDL3 in the pathogenesis of COPD is still unknown. OBJECTIVE Because cigarette smoke is the major risk factor for COPD, we aimed to investigate the role of ORMDL3 in cigarette smoke-induced human airway smooth muscle cell (HASMC) injury. METHODS The mRNA and protein expression of ORMDL3 was examined in HASMCs from nonsmokers and smokers without or with COPD. Knockdown of ORMDL3 in primary healthy HASMCs was performed using small interfering RNA before exposure to cigarette smoke medium (CSM) for 24 hours. Inflammatory, proliferative/apoptotic, ER stress, and mitochondrial markers were evaluated. RESULTS Elevation of ORMDL3 mRNA and protein expression was observed in HASMCs of smokers without or with COPD. CSM caused significant upregulation of ORMDL3 expression in healthy nonsmokers. ORMDL3 knockdown regulated CSM-induced inflammation, cell proliferation, and apoptosis. Silencing ORMDL3 led to reduction of CSM-induced ER stress via inhibition of unfolded protein response pathways such as activating transcription factor 6 and protein kinase RNA-like ER kinase. ORMDL3 was also involved in CSM-induced mitochondrial dysfunction via the mitochondrial fission process. CONCLUSIONS We report the induction of ORMDL3 in HASMCs after cigarette smoke exposure. ORMDL3 may mediate cigarette smoke-induced activation of unfolded protein response pathways during airway smooth muscle cell injury.
Collapse
Affiliation(s)
- Rui Chen
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Charalambos Michaeloudes
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital Respiratory Division, Shenzhen, China
| | - Yingmin Liang
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China; Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital Respiratory Division, Shenzhen, China
| | - Pankaj K Bhavsar
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital Respiratory Division, Shenzhen, China
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital Respiratory Division, Shenzhen, China
| | - Mary S M Ip
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China; Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital Respiratory Division, Shenzhen, China
| | - Judith C W Mak
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China; Respiratory Medicine, The University of Hong Kong-Shenzhen Hospital Respiratory Division, Shenzhen, China; Department of Pharmacology & Pharmacy, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
40
|
Sunde RB, Thorsen J, Pedersen CET, Stokholm J, Bønnelykke K, Chawes B, Bisgaard H. Prenatal tobacco exposure and risk of asthma and allergy outcomes in childhood. Eur Respir J 2022; 59:2100453. [PMID: 34244319 DOI: 10.1183/13993003.00453-2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 06/21/2021] [Indexed: 11/05/2022]
Abstract
BACKGROUND Harmful effects of prenatal tobacco exposure and possible interaction with 17q12-21 genetic variants have been shown for some asthma outcomes in childhood, whereas findings related to allergy outcomes are more inconsistent. This study aimed to examine the effect of prenatal tobacco exposure and relation to 17q12-21 genotype on a wide array of asthma and allergy-related outcomes in early childhood. METHODS Prenatal tobacco exposure was determined by maternal smoking during the third trimester (yes/no) in 411 children from the phenotyped Copenhagen Prospective Studies on Asthma in Childhood 2000 (COPSAC2000) birth cohort with clinical follow-up to age 7 years. The rs7216389 single nucleotide polymorphism was used as main representative of the 17q12-21 locus. Asthma end-points included asthma diagnosis, exacerbations, episodes with troublesome lung symptoms and lower respiratory tract infections, spirometry, plethysmography, bronchial responsiveness to methacholine, exercise and cold dry air. Allergy-related endpoints included aeroallergen sensitisation, allergic rhinitis, fractional exhaled nitric oxide, blood eosinophil count and urine eosinophil protein X levels. Statistical analyses were done using Cox regression, linear regression, logistic regression and quasi-Poisson regression. RESULTS Prenatal tobacco exposure increased the risk of asthma (adjusted hazard ratio (aHR) 2.05, 95% CI 1.13-3.73; p=0.02), exacerbations (aHR 3.76, 95% CI 2.05-6.91; p<0.001), number of LRTIs (adjusted incidence rate ratio 1.87, 95% CI 1.34-2.55; p<0.001), and was associated with decreased spirometry indices (forced expiratory volume in 1 s (FEV1) adjusted mean difference (aMD) -0.07 L, 95% CI -0.13- -0.005 L, p=0.03; maximal mid-expiratory flow aMD -0.19 L·s-1, -0.34- -0.04 L·s-1, p=0.01) and increased bronchial responsiveness to methacholine (provocative dose of methacholine causing a 20% drop in FEV1 adjusted geometric mean ratio 0.55, 95% CI 0.31-0.96; p=0.04). In contrast, there was no association with any allergy-related end-points. The effect on asthma depended on 17q12-21 genotype with an increased risk only among children without risk alleles. CONCLUSION Prenatal tobacco exposure was associated with asthma dependent on 17q12-21 genotype and with exacerbations, lung function and bronchial responsiveness, but not with any allergy-related outcomes. This suggests that tobacco exposure in utero leads to adverse lung developmental/structural effects rather than susceptibility to develop allergy and type 2 inflammation.
Collapse
Affiliation(s)
- Rikke Bjersand Sunde
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Dept of Pediatrics, Slagelse Sygehus, Slagelse, Denmark
| | - Jonathan Thorsen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Casper-Emil Tingskov Pedersen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Stokholm
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Dept of Pediatrics, Slagelse Sygehus, Slagelse, Denmark
| | - Klaus Bønnelykke
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Bo Chawes
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Hans Bisgaard
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
41
|
Illi S, Depner M, Pfefferle PI, Renz H, Roduit C, Taft DH, Kalanetra KM, Mills DA, Farquharson FM, Louis P, Schmausser-Hechfellner E, Divaret-Chauveau A, Lauener R, Karvonen AM, Pekkanen J, Kirjavainen PV, Roponen M, Riedler J, Kabesch M, Schaub B, von Mutius E. Immune Responsiveness to LPS Determines Risk of Childhood Wheeze and Asthma in 17q21 Risk Allele Carriers. Am J Respir Crit Care Med 2021; 205:641-650. [PMID: 34919021 DOI: 10.1164/rccm.202106-1458oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND In murine models microbial exposures induce protection from experimental allergic asthma through innate immunity. Our aim was to assess the association of early life innate immunity with the development of asthma in children at risk. METHODS In the PASTURE farm birth cohort innate, Th2, Th1 and Th17 cytokine expression at age 1 year was measured after stimulation of PBMCs with lipopolysaccharide (LPS) in N=445 children. Children at risk of asthma were defined based on single-nucleotide polymorphisms at the 17q21 asthma gene locus. Specifically, we used the SNP rs7216389 in the GSDMB gene. Wheeze in the 1st year of life was assessed by weekly diaries and asthma by questionnaire at age 6 years. RESULTS Not all cytokines were detectable in all children after LPS-stimulation. When classifying detectability of cytokines by latent class analysis, carrying the 17q21 risk allele rs7216389 was associated with risk of wheeze only in the class with the lowest level of LPS-induced activation, odds ratio (OR)=1.89, 95%-CI 1.13-3.16, p=0.015. In contrast, in children with high cytokine activation after LPS-stimulation no association of the 17q21 risk allele with wheeze (OR=0.63, 95%-CI 0.29-1.40, p=0.258, p=0.034 for interaction) or school age asthma was observed. In these children consumption of unprocessed cow's milk was associated with higher cytokine activation (OR=3.37, 95%-CI 1.56-7.30, p=0.002), which was in part mediated by the gut microbiome. CONCLUSIONS These findings suggest that within the 17q21 genotype asthma risk can be mitigated by activated immune responses after innate stimulation, which is partly mediated by a gut-immune axis.
Collapse
Affiliation(s)
- Sabina Illi
- Helmholtz Zentrum Munchen Deutsches Forschungszentrum fur Gesundheit und Umwelt, 9150, Institute of Asthma and Allergy Prevention, Neuherberg, Germany.,German Center for Lung Research, 542891, Giessen, Germany;
| | - Martin Depner
- Helmholtz Zentrum Munchen Deutsches Forschungszentrum fur Umwelt und Gesundheit, 9150, Institute of Asthma and Allergy Prevention, Neuherberg, Germany
| | - Petra Ina Pfefferle
- Philipps-Universität Marburg Fachbereich Medizin, 98594, Comprehensive Biobank Marburg CBBM, Marburg, Germany.,German Center for Lung Research, 542891, Giessen, Germany
| | - Harald Renz
- Philipps-Universität Marburg, 9377, Institute of Laboratory Medicine, Marburg, Germany.,Sechenov University, 68477, Department of Clinical Immunology and Allergology, Laboratory of Immunopathology, Moskva, Russian Federation.,German Center for Lung Research, 542891, Giessen, Germany
| | - Caroline Roduit
- University of Zurich, Children's Hospital, Zurich, Switzerland.,Christine Kühne Center for Allergy Research and Education (CK-CARE) , Davos, Switzerland.,Children's Hospital of Eastern Switzerland, St. Gallen, Switzerland
| | - Diana Hazard Taft
- University of California Davis, 8789, Department of Food Science & Technology, Davis, California, United States
| | - Karen M Kalanetra
- University of California Davis, 8789, Department of Food Science & Technology, Davis, California, United States
| | - David A Mills
- University of California Davis, 8789, Department of Food Science & Technology, Davis, California, United States
| | - Freda M Farquharson
- University of Aberdeen, 1019, The Rowett Institute, Aberdeen, United Kingdom of Great Britain and Northern Ireland
| | - Petra Louis
- University of Aberdeen, 1019, The Rowett Institute, Aberdeen, United Kingdom of Great Britain and Northern Ireland
| | - Elisabeth Schmausser-Hechfellner
- Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt, 9150, Institute of Asthma and Allergy Prevention, Neuherberg, Germany
| | - Amandine Divaret-Chauveau
- Burgundy Franche-Comté University, 439716, UMR 6249 Chrono-environment , Besancon, France.,University of Lorraine, 137665, EA3450 Développement Adaptation et Handicap (DevAH) , Nancy, France.,University Hospital Centre Nancy, 26920, Pediatric Allergy Department, Nancy, France
| | - Roger Lauener
- Children's Hospital of Eastern Switzerland, Children's Hospital of Eastern Switzerland, St. Gallen, Switzerland.,Christine Kühne Center for Allergy Research and Education (CK-CARE) , Davos, Switzerland
| | - Anne M Karvonen
- Finnish Institute for Health and Welfare, 3837, Department of Health Security, Helsinki, Finland
| | - Juha Pekkanen
- University of Helsinki, Department of Public Health, Helsinki, Finland.,Finnish Institute for Health and Welfare, 3837, Department of Health Security, Helsinki, Finland
| | - Pirkka V Kirjavainen
- Finnish Institute for Health and Welfare, 3837, Department of Health Security, Kuopio, Finland.,University of Eastern Finland, 163043, Institute of Public Health and Clinical Nutrition, Kuopio, Finland
| | - Marjut Roponen
- University of Eastern Finland, 163043, Department of Environmental and Biological Sciences, Kuopio, Finland
| | - Josef Riedler
- Children's Hospital Schwarzach, Children's Hospital Schwarzach, Schwarzach, Austria
| | - Michael Kabesch
- University Children's Hospital Regensburg (KUNO), Department of Pediatric Pneumology and Allergy, Campus St. Hedwig, Regensburg, Germany
| | - Bianca Schaub
- Ludwig-Maximilians-Universitat Munchen, 9183, Dr. von Hauner Children's Hospital, Munchen, Germany.,German Center for Lung Research, 542891, Giessen, Germany
| | - Erika von Mutius
- Helmholtz Zentrum Munchen Deutsches Forschungszentrum fur Gesundheit und Umwelt, 9150, Institute of Asthma and Allergy Prevention, Neuherberg, Germany.,Ludwig-Maximilians-Universitat Munchen, 9183, Dr. von Hauner Children's Hospital, München, Germany.,German Center for Lung Research, 542891, Giessen, Germany
| | | |
Collapse
|
42
|
Zou J, Zheng Y, Huang Y, Tang D, Kang R, Chen R. The Versatile Gasdermin Family: Their Function and Roles in Diseases. Front Immunol 2021; 12:751533. [PMID: 34858408 PMCID: PMC8632255 DOI: 10.3389/fimmu.2021.751533] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 10/25/2021] [Indexed: 12/22/2022] Open
Abstract
The gasdermin (GSDM) family, a novel group of structure-related proteins, consists of GSDMA, GSDMB, GSDMC, GSDMD, GSDME/DNFA5, and PVJK/GSDMF. GSDMs possess a C-terminal repressor domain, cytotoxic N-terminal domain, and flexible linker domain (except for GSDMF). The GSDM-NT domain can be cleaved and released to form large oligomeric pores in the membrane that facilitate pyroptosis. The emerging roles of GSDMs include the regulation of various physiological and pathological processes, such as cell differentiation, coagulation, inflammation, and tumorigenesis. Here, we introduce the basic structure, activation, and expression patterns of GSDMs, summarize their biological and pathological functions, and explore their regulatory mechanisms in health and disease. This review provides a reference for the development of GSDM-targeted drugs to treat various inflammatory and tissue damage-related conditions.
Collapse
Affiliation(s)
- Ju Zou
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Yixiang Zheng
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Yan Huang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, United States
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, United States
| | - Ruochan Chen
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
43
|
Wasserman E, Gomi R, Sharma A, Hong S, Bareja R, Gu J, Balaji U, Veerappan A, Kim BI, Wu W, Heras A, Perez-Zoghbi J, Sung B, Gueye-Ndiaye S, Worgall TS, Worgall S. Human Rhinovirus Infection of the Respiratory Tract Affects Sphingolipid Synthesis. Am J Respir Cell Mol Biol 2021; 66:302-311. [PMID: 34851798 DOI: 10.1165/rcmb.2021-0443oc] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The 17q21 asthma susceptibility locus includes asthma risk alleles associated with decreased sphingolipid synthesis, likely resulting from increased expression of ORMDL3. ORMDL3 inhibits serine-palmitoyl transferase (SPT), the rate limiting enzyme of de novo sphingolipid synthesis. There is evidence that decreased sphingolipid synthesis is critical to asthma pathogenesis. Children with asthma and 17q21 asthma risk alleles display decreased sphingolipid synthesis in blood cells. Reduced SPT activity results in airway hyperreactivity, a hallmark feature of asthma. 17q21 asthma risk alleles are also linked to childhood infections with human rhinovirus (RV). This study evaluates the interaction of RV with the de novo sphingolipid synthesis pathway, and the alterative effects of concurrent SPT inhibition in SPT-deficient mice and human airway epithelial cells. In mice, RV infection shifted lung sphingolipid synthesis gene expression to a pattern that resembles genetic SPT deficiency, including decreased expression of Sptssa, a small SPT subunit. This pattern was pronounced in lung EpCAM+ epithelial cells and reproduced in human bronchial epithelial cells. RV did not affect Sptssa expression in lung CD45+ immune cells. RV increased sphingolipids unique to the de novo synthesis pathway in mouse lung and human airway epithelial cells. Interestingly, these de novo sphingolipid species were reduced in the blood of RV infected, wild-type mice. RV exacerbated SPT-deficiency-associated airway hyperreactivity. Airway inflammation was similar in RV-infected wild-type and SPT deficient mice. This study reveals the effects of RV infection on the de novo sphingolipid synthesis pathway, elucidating a potential mechanistic link between 17q21 asthma risk alleles and rhinoviral infection.
Collapse
Affiliation(s)
- Emily Wasserman
- Weill Cornell Medical College, 12295, Pediatrics, New York, New York, United States
| | - Rika Gomi
- Weill Cornell Medical College, 12295, Pediatrics, New York, New York, United States
| | - Anurag Sharma
- Weill Cornell Medical College, 12295, Pediatrics, New York, New York, United States
| | - Seunghee Hong
- Weill Cornell Medical College, 12295, Pediatrics, New York, New York, United States
| | - Rohan Bareja
- Weill Cornell Medical College, 12295, Precision Medicine, New York, New York, United States
| | - Jinghua Gu
- Weill Cornell Medical College, 12295, Pediatrics, New York, New York, United States
| | - Uthra Balaji
- Weill Cornell Medical College, 12295, Pediatrics, New York, New York, United States
| | - Arul Veerappan
- New York University, 5894, Medicine, New York, New York, United States
| | - Benjamin I Kim
- Columbia University, 5798, Pathology, New York, New York, United States
| | - Wenzhu Wu
- Weill Cornell Medical College, 12295, New York, New York, United States
| | - Andrea Heras
- Weill Cornell Medical College, 12295, Pediatrics , New York, New York, United States
| | - Jose Perez-Zoghbi
- Columbia University, 5798, Department of Anesthesiology , New York, New York, United States
| | - Biin Sung
- Weill Cornell Medical College, 12295, Pediatrics, New York, New York, United States
| | - Seyni Gueye-Ndiaye
- Weill Cornell Medical College, 12295, Pediatrics, New York, New York, United States
| | - Tilla S Worgall
- Columbia University Irving Medical Center, 21611, Dept. of Pathology, New York, New York, United States
| | - Stefan Worgall
- Weill Cornell Medical College, 12295, Pediatrics, New York, New York, United States;
| |
Collapse
|
44
|
Zack DE, Stern DA, Willis AL, Kim AS, Mansfield CJ, Reed DR, Brooks SG, Adappa ND, Palmer JN, Cohen NA, Chiu AG, Song BH, Le CH, Chang EH. The GSDMB rs7216389 SNP is associated with chronic rhinosinusitis in a multi-institutional cohort. Int Forum Allergy Rhinol 2021; 11:1647-1653. [PMID: 34076350 PMCID: PMC8636513 DOI: 10.1002/alr.22824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/23/2021] [Accepted: 05/05/2021] [Indexed: 11/11/2022]
Abstract
BACKGROUND Chronic rhinosinusitis (CRS) is a multifactorial disease with a high co-occurrence with asthma. In this multicohort study, we tested whether single nucleotide polymorphisms (SNPs) associated with childhood asthma and rhinovirus (RV)-associated disease are related to an increased susceptibility to adult CRS in a multicohort retrospective case-control study. METHODS Participants at two tertiary academic rhinology centers, University of Arizona (UofA) and University of Pennsylvania (UPenn) were recruited. Cases were defined as those with physician diagnosed CRS (UofA, n = 149; UPenn, n = 250), and healthy controls were those without CRS (UofA, n = 66; UPenn, n = 275). Genomic DNA was screened for the GSDMB rs7216389 SNP and CDHR3 rs6967330 SNP. Gene dosage, or the number of combined risk alleles in a single subject was calculated. Meta-analysis of the association between GSDMB or CDHR3 genotypes and CRS was performed and additive gene dosage effect for each population calculated using p for trend. RESULTS A meta-analysis revealed a combined increased risk for CRS in subjects with the GSDMB rs7216389 SNP (odds ratio [OR] 1.40; 95% confidence interval [CI], 1.16-1.76; p = 0.004). Both the UofA (OR 1.73; 95% CI, 1.23-2.43; p = 0.002) and UPenn (OR 1.27; 95% CI, 1.02-1.58; p = 0.035) populations showed a significant positive association between the number of combined risk alleles of GSDMB rs7216389 SNP and CDHR3 rs6967330 SNP and risk for CRS. CONCLUSION Carriers of the GSDMB rs7216389 SNP and CDHR3 rs6967330 SNP are at increased susceptibility for CRS. These data suggest that therapeutic approaches to target aberrant responses to RV infection may play a role in the treatment of unified airway disease.
Collapse
Affiliation(s)
- Dana E Zack
- Department of Otolaryngology, University of Arizona, Tucson, Arizona, USA
| | - Debra A Stern
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Arizona, USA
| | - Amanda L Willis
- Department of Otolaryngology, University of Arizona, Tucson, Arizona, USA
| | - Alexander S Kim
- Department of Otolaryngology, University of Arizona, Tucson, Arizona, USA
| | - Corinne J Mansfield
- Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Danielle R Reed
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Steven G Brooks
- Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nithin D Adappa
- Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James N Palmer
- Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Noam A Cohen
- Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Monell Chemical Senses Center, Philadelphia, Pennsylvania, USA
| | - Alexander G Chiu
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Brian H Song
- Department of Otolaryngology, University of Arizona, Tucson, Arizona, USA
| | - Chris H Le
- Department of Otolaryngology, University of Arizona, Tucson, Arizona, USA
| | - Eugene H Chang
- Department of Otolaryngology, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
45
|
Soliai MM, Kato A, Helling BA, Stanhope CT, Norton JE, Naughton KA, Klinger AI, Thompson EE, Clay SM, Kim S, Celedón JC, Gern JE, Jackson DJ, Altman MC, Kern RC, Tan BK, Schleimer RP, Nicolae DL, Pinto JM, Ober C. Multi-omics colocalization with genome-wide association studies reveals a context-specific genetic mechanism at a childhood onset asthma risk locus. Genome Med 2021; 13:157. [PMID: 34629083 PMCID: PMC8504130 DOI: 10.1186/s13073-021-00967-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/10/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Genome-wide association studies (GWASs) have identified thousands of variants associated with asthma and other complex diseases. However, the functional effects of most of these variants are unknown. Moreover, GWASs do not provide context-specific information on cell types or environmental factors that affect specific disease risks and outcomes. To address these limitations, we used an upper airway epithelial cell (AEC) culture model to assess transcriptional and epigenetic responses to rhinovirus (RV), an asthma-promoting pathogen, and provide context-specific functional annotations to variants discovered in GWASs of asthma. METHODS Genome-wide genetic, gene expression, and DNA methylation data in vehicle- and RV-treated upper AECs were collected from 104 individuals who had a diagnosis of airway disease (n=66) or were healthy participants (n=38). We mapped cis expression and methylation quantitative trait loci (cis-eQTLs and cis-meQTLs, respectively) in each treatment condition (RV and vehicle) in AECs from these individuals. A Bayesian test for colocalization between AEC molecular QTLs and adult onset asthma and childhood onset asthma GWAS SNPs, and a multi-ethnic GWAS of asthma, was used to assign the function to variants associated with asthma. We used Mendelian randomization to demonstrate DNA methylation effects on gene expression at asthma colocalized loci. RESULTS Asthma and allergic disease-associated GWAS SNPs were specifically enriched among molecular QTLs in AECs, but not in GWASs from non-immune diseases, and in AEC eQTLs, but not among eQTLs from other tissues. Colocalization analyses of AEC QTLs with asthma GWAS variants revealed potential molecular mechanisms of asthma, including QTLs at the TSLP locus that were common to both the RV and vehicle treatments and to both childhood onset and adult onset asthma, as well as QTLs at the 17q12-21 asthma locus that were specific to RV exposure and childhood onset asthma, consistent with clinical and epidemiological studies of these loci. CONCLUSIONS This study provides evidence of functional effects for asthma risk variants in AECs and insight into RV-mediated transcriptional and epigenetic response mechanisms that modulate genetic effects in the airway and risk for asthma.
Collapse
Affiliation(s)
- Marcus M Soliai
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA.
- Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL, USA.
| | - Atsushi Kato
- Departments of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Britney A Helling
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA
| | | | - James E Norton
- Departments of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Aiko I Klinger
- Departments of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Emma E Thompson
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Selene M Clay
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Soyeon Kim
- Division of Pediatric Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Juan C Celedón
- Division of Pediatric Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - James E Gern
- Department of Pediatrics, University of Wisconsin, School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Daniel J Jackson
- Department of Pediatrics, University of Wisconsin, School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Matthew C Altman
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA, USA
- Systems Immunology Program, Benaroya Research Institute, Seattle, WA, USA
| | - Robert C Kern
- Department of Otolaryngology-Head and Neck Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Bruce K Tan
- Department of Otolaryngology-Head and Neck Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Robert P Schleimer
- Departments of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Dan L Nicolae
- Department of Statistics, University of Chicago, Chicago, IL, USA
| | - Jayant M Pinto
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Carole Ober
- Departments of Human Genetics, University of Chicago, Chicago, IL, USA.
- Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
46
|
Kitazawa H, Masuko H, Kanazawa J, Shigemasa R, Hyodo K, Yamada H, Yatagai Y, Kaneko Y, Iijima H, Naito T, Saito T, Noguchi E, Konno S, Hirota T, Tamari M, Sakamoto T, Hizawa N. ORMDL3/GSDMB genotype is associated with distinct phenotypes of adult asthma. Allergol Int 2021; 70:495-497. [PMID: 33941434 DOI: 10.1016/j.alit.2021.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/27/2021] [Accepted: 04/07/2021] [Indexed: 11/24/2022] Open
|
47
|
Maroteau C, Espuela-Ortiz A, Herrera-Luis E, Srinivasan S, Carr F, Tavendale R, Wilson K, Hernandez-Pacheco N, Chalmers JD, Turner S, Mukhopadhyay S, Maitland-van der Zee AH, Burchard EG, Pino-Yanes M, Young S, Lassi G, Platt A, Palmer CNA. LTA4H rs2660845 association with montelukast response in early and late-onset asthma. PLoS One 2021; 16:e0257396. [PMID: 34550981 PMCID: PMC8457475 DOI: 10.1371/journal.pone.0257396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 08/31/2021] [Indexed: 12/15/2022] Open
Abstract
Leukotrienes play a central pathophysiological role in both paediatric and adult asthma. However, 35% to 78% of asthmatics do not respond to leukotriene inhibitors. In this study we tested the role of the LTA4H regulatory variant rs2660845 and age of asthma onset in response to montelukast in ethnically diverse populations. We identified and genotyped 3,594 asthma patients treated with montelukast (2,514 late-onset and 1,080 early-onset) from seven cohorts (UKBiobank, GoSHARE, BREATHE, Tayside RCT, PAGES, GALA II and SAGE). Individuals under montelukast treatment experiencing at least one exacerbation in a 12-month period were compared against individuals with no exacerbation, using logistic regression for each cohort and meta-analysis. While no significant association was found with European late-onset subjects, a meta-analysis of 523 early-onset individuals from European ancestry demonstrated the odds of experiencing asthma exacerbations by carriers of at least one G allele, despite montelukast treatment, were increased (odds-ratio = 2.92, 95%confidence interval (CI): 1.04-8.18, I2 = 62%, p = 0.0412) compared to those in the AA group. When meta-analysing with other ethnic groups, no significant increased risk of asthma exacerbations was found (OR = 1.60, 95% CI: 0.61-4.19, I2 = 85%, p = 0.342). Our study demonstrates that genetic variation in LTA4H, together with timing of asthma onset, may contribute to variability in montelukast response. European individuals with early-onset (≤18y) carrying at least one copy of rs2660845 have increased odd of exacerbation under montelukast treatment, presumably due to the up-regulation of LTA4H activity. These findings support a precision medicine approach for the treatment of asthma with montelukast.
Collapse
Affiliation(s)
- Cyrielle Maroteau
- Centre for Genomics Research, Discovery Science, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Antonio Espuela-Ortiz
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, San Cristobal de La Laguna, Santa Cruz de Tenerife, Spain
| | - Esther Herrera-Luis
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, San Cristobal de La Laguna, Santa Cruz de Tenerife, Spain
| | - Sundararajan Srinivasan
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Fiona Carr
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Roger Tavendale
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Karen Wilson
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Natalia Hernandez-Pacheco
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, San Cristobal de La Laguna, Santa Cruz de Tenerife, Spain
- Research Unit, Hospital Universitario N.S. de Candelaria, Universidad de La Laguna, San Cristobal de La Laguna, Spain
| | - James D. Chalmers
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Steve Turner
- Child Health, University of Aberdeen, Aberdeen, United Kingdom
| | - Somnath Mukhopadhyay
- Academic Department of Pediatrics, Brighton and Sussex Medical School, Royal Alexandra Children’s Hospital, Brighton, United Kingdom
| | - Anke-Hilse Maitland-van der Zee
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, Utrecht, The Netherlands
- Department of Respiratory Medicine, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- Department of Pediatric Respiratory Medicine and Allergy, Emma’s Children Hospital, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, The Netherlands
| | - Esteban G. Burchard
- Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Maria Pino-Yanes
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, San Cristobal de La Laguna, Santa Cruz de Tenerife, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Comunidad de Madrid, Spain
- Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - Simon Young
- BioPharmaceutical Precision Medicine, Oncology R&D, AstraZeneca, Cambridge, United Kingdom
- Research & Development Centre, Sysmex, Cambridge, United Kingdom
| | - Glenda Lassi
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Adam Platt
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Colin N. A. Palmer
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, United Kingdom
| | | |
Collapse
|
48
|
Wang N, Brix S, Larsen JM, Thysen AH, Rasmussen MA, Workman CT, Stokholm J, Bønnelykke K, Bisgaard H, Chawes BL. Innate IL-23/Type 17 immune responses mediate the effect of the 17q21 locus on childhood asthma. Clin Exp Allergy 2021; 51:892-901. [PMID: 33987892 DOI: 10.1111/cea.13900] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 04/16/2021] [Accepted: 04/22/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND Several childhood asthma risk loci that relate to immune function have been identified by genome-wide association studies (GWAS), but the underlying mechanisms remain unknown. OBJECTIVE Here, we examined whether perturbed innate immune responses mediate the association between known genetic risk variants and development of childhood asthma. METHODS Peripheral blood mononuclear cells from 336 six-month-old infants from the Copenhagen Prospective Studies on Asthma in Childhood (COPSAC2000 ) cohort were stimulated in vitro with six different innate ligands (LPS, CpG, poly(I:C), R848, HDMAPP and aluminium hydroxide together with low levels of LPS) followed by quantification of 18 released cytokines and chemokines 40 h after the stimulations. The innate immune response profiles were decomposed by principal component (PC) analysis, and PC1-5 were used in mediation analyses of the effect of 25 known genetic risk variants on childhood asthma until age 7. RESULTS The effects of two variants from the 17q21 locus (rs7216389, rs2305480) on asthma and exacerbation risk were significantly mediated by immune parameters induced in response to ligands mimicking intracellular colonization; bacterial DNA (CpG) and double-stranded viral RNA (poly(I:C)). The Th17 and innate lymphoid cell type 3-amplifying cytokine IL-23 was the most prominent cytokine involved. CONCLUSION The 17q21 effect on childhood asthma and exacerbations was partly mediated by deregulation of IL-23 in response to intracellular microbial ligands, which may suggest ineffective clearance of intracellular pathogens in the lungs.
Collapse
Affiliation(s)
- Ni Wang
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark.,Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Susanne Brix
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Jeppe M Larsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Anna H Thysen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark.,Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Morten A Rasmussen
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark.,Faculty of Life Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Christopher T Workman
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Jakob Stokholm
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
| | - Klaus Bønnelykke
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
| | - Hans Bisgaard
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
| | - Bo L Chawes
- COPSAC, Copenhagen Prospective Studies on Asthma in Childhood, Herlev and Gentofte Hospital, University of Copenhagen, Gentofte, Denmark
| |
Collapse
|
49
|
Ziani M, Henry AP, Hall IP. Association study between asthma and single nucleotide polymorphisms of ORMDL3, GSDMB, and IL1RL1 genes in an Algerian population. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2021. [DOI: 10.1186/s43042-021-00163-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Abstract
Background
Genetic variation has a key role in the development of asthma, but genetic influences may vary between different populations. In this study, we looked for evidence of association of key asthma SNPs, namely, rs1420101 and rs10192157 within the IL1RL1 gene, rs2305480 in GSDMB gene, and the rs3744246 polymorphism in the ORMDL3 gene, in the Algerian population. We included 266 unrelated subjects of an Algerian population in a case-control study, with 125 adult asthmatic and 141 healthy controls. DNA was extracted and genotypes determined by the Taqman PCR technique for characterization of the different genetic variants.
Results
The results show that there were no significant differences in allele frequencies for 3 of the chosen SNPs in the ORMDL3, GSDMB, and IL1RL1 genes between the asthmatic and control groups with respective P values of 0.922, 0.331, and 0.937. However the T allele of rs10192157 of the IL1RL1gene was associated with protection from asthma (P value=0.010).
Conclusion
These results indicate that there is no marked effect of rs3744246, rs2305480, and rs1420101 polymorphisms of the ORMDL3, GSDMB, and IL1RL1 genes on asthma risk in the Algerian population. However, a protective effect of the rs10192157 polymorphism of the IL1RL1 gene was found.
Collapse
|
50
|
Syreeni A, Sandholm N, Sidore C, Cucca F, Haukka J, Harjutsalo V, Groop PH. Genome-wide search for genes affecting the age at diagnosis of type 1 diabetes. J Intern Med 2021; 289:662-674. [PMID: 33179336 PMCID: PMC8247053 DOI: 10.1111/joim.13187] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/07/2020] [Accepted: 09/09/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Type 1 diabetes (T1D) is an autoimmune disease affecting individuals in the early years of life. Although previous studies have identified genetic loci influencing T1D diagnosis age, these studies did not investigate the genome with high resolution. OBJECTIVE AND METHODS We performed a genome-wide meta-analysis for age at diagnosis with cohorts from Finland (Finnish Diabetic Nephropathy Study), the United Kingdom (UK Genetic Resource Investigating Diabetes) and Sardinia. Through SNP associations, transcriptome-wide association analysis linked T1D diagnosis age and gene expression. RESULTS We identified two chromosomal regions associated with T1D diagnosis age: multiple independent variants in the HLA region on chromosome 6 and a locus on chromosome 17q12. We performed gene-level association tests with transcriptome prediction models from two whole blood datasets, lymphocyte cell line, spleen, pancreas and small intestine tissues. Of the non-HLA genes, lower PNMT expression in whole blood, and higher IKZF3 and ZPBP2, and lower ORMDL3 and GSDMB transcription levels in multiple tissues were associated with lower T1D diagnosis age (FDR = 0.05). These genes lie on chr17q12 which is associated with T1D, other autoimmune diseases, and childhood asthma. Additionally, higher expression of PHF20L1, a gene not previously implicated in T1D, was associated with lower diagnosis age in lymphocytes, pancreas, and spleen. Altogether, the non-HLA associations were enriched in open chromatin in various blood cells, blood vessel tissues and foetal thymus tissue. CONCLUSION Multiple genes on chr17q12 and PHF20L1 on chr8 were associated with T1D diagnosis age and only further studies may elucidate the role of these genes for immunity and T1D onset.
Collapse
Affiliation(s)
- A Syreeni
- From the, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - N Sandholm
- From the, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - C Sidore
- Instituto di Ricerca Genetica e Biomedica, CNR, Monserrato, Italy
| | - F Cucca
- Instituto di Ricerca Genetica e Biomedica, CNR, Monserrato, Italy.,Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, Sassari, Italy
| | - J Haukka
- From the, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - V Harjutsalo
- From the, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland.,National Institute for Health and Welfare, Helsinki, Finland
| | - P-H Groop
- From the, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland.,Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | | |
Collapse
|