1
|
Xie C, Yang J, Gul A, Li Y, Zhang R, Yalikun M, Lv X, Lin Y, Luo Q, Gao H. Immunologic aspects of asthma: from molecular mechanisms to disease pathophysiology and clinical translation. Front Immunol 2024; 15:1478624. [PMID: 39439788 PMCID: PMC11494396 DOI: 10.3389/fimmu.2024.1478624] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
In the present review, we focused on recent translational and clinical discoveries in asthma immunology, facilitating phenotyping and stratified or personalized interventions for patients with this condition. The immune processes behind chronic inflammation in asthma exhibit marked heterogeneity, with diverse phenotypes defining discernible features and endotypes illuminating the underlying molecular mechanisms. In particular, two primary endotypes of asthma have been identified: "type 2-high," characterized by increased eosinophil levels in the airways and sputum of patients, and "type 2-low," distinguished by increased neutrophils or a pauci-granulocytic profile. Our review encompasses significant advances in both innate and adaptive immunities, with emphasis on the key cellular and molecular mediators, and delves into innovative biological and targeted therapies for all the asthma endotypes. Recognizing that the immunopathology of asthma is dynamic and continuous, exhibiting spatial and temporal variabilities, is the central theme of this review. This complexity is underscored through the innumerable interactions involved, rather than being driven by a single predominant factor. Integrated efforts to improve our understanding of the pathophysiological characteristics of asthma indicate a trend toward an approach based on disease biology, encompassing the combined examination of the clinical, cellular, and molecular dimensions of the disease to more accurately correlate clinical traits with specific disease mechanisms.
Collapse
Affiliation(s)
- Cong Xie
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Jingyan Yang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Aman Gul
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
- Department of Respiratory Medicine, Uyghur Medicines Hospital of Xinjiang Uyghur Autonomous Region, Urumqi, China
- College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Yifan Li
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Rui Zhang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Maimaititusun Yalikun
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Xiaotong Lv
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuhan Lin
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qingli Luo
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Fudan Institutes of Integrative Medicine, Fudan University Shanghai Medical College, Shanghai, China
| | - Huijuan Gao
- Department of Endocrinology and Clinical Immunology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
2
|
Herrera-Luis E, Martin-Almeida M, Pino-Yanes M. Asthma-Genomic Advances Toward Risk Prediction. Clin Chest Med 2024; 45:599-610. [PMID: 39069324 PMCID: PMC11284279 DOI: 10.1016/j.ccm.2024.03.002] [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: 07/30/2024]
Abstract
Asthma is a common complex airway disease whose prediction of disease risk and most severe outcomes is crucial in clinical practice for adequate clinical management. This review discusses the latest findings in asthma genomics and current obstacles faced in moving forward to translational medicine. While genome-wide association studies have provided valuable insights into the genetic basis of asthma, there are challenges that must be addressed to improve disease prediction, such as the need for diverse representation, the functional characterization of genetic variants identified, variant selection for genetic testing, and refining prediction models using polygenic risk scores.
Collapse
Affiliation(s)
- Esther Herrera-Luis
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, 615 N Wolfe Street, Baltimore, MD 21205, USA.
| | - Mario Martin-Almeida
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez, s/n. Facultad de Ciencias, San Cristóbal de La Laguna, S/C de Tenerife La Laguna 38200, Tenerife, Spain
| | - Maria Pino-Yanes
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna (ULL), Avenida Astrofísico Francisco Sánchez, s/n. Facultad de Ciencias, San Cristóbal de La Laguna, S/C de Tenerife La Laguna 38200, Tenerife, Spain; CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid 28029, Spain; Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna (ULL), San Cristóbal de La Laguna 38200, Tenerife, Spain
| |
Collapse
|
3
|
Edris A, Voorhies K, Lutz SM, Iribarren C, Hall I, Wu AC, Tobin M, Fawcett K, Lahousse L. Asthma exacerbations and eosinophilia in the UK Biobank: a genome-wide association study. ERJ Open Res 2024; 10:00566-2023. [PMID: 38196893 PMCID: PMC10772900 DOI: 10.1183/23120541.00566-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/31/2023] [Indexed: 01/11/2024] Open
Abstract
Background Asthma exacerbations reflect disease severity, affect morbidity and mortality, and may lead to declining lung function. Inflammatory endotypes (e.g. T2-high (eosinophilic)) may play a key role in asthma exacerbations. We aimed to assess whether genetic susceptibility underlies asthma exacerbation risk and additionally tested for an interaction between genetic variants and eosinophilia on exacerbation risk. Methods UK Biobank data were used to perform a genome-wide association study of individuals with asthma and at least one exacerbation compared to individuals with asthma and no history of exacerbations. Individuals with asthma were identified using self-reported data, hospitalisation data and general practitioner records. Exacerbations were identified as either asthma-related hospitalisation, general practitioner record of asthma exacerbation or an oral corticosteroid burst prescription. A logistic regression model adjusted for age, sex, smoking status and genetic ancestry via principal components was used to assess the association between genetic variants and asthma exacerbations. We sought replication for suggestive associations (p<5×10-6) in the GERA cohort. Results In the UK Biobank, we identified 11 604 cases and 37 890 controls. While no variants reached genome-wide significance (p<5×10-8) in the primary analysis, 116 signals were suggestively significant (p<5×10-6). In GERA, two single nucleotide polymorphisms (rs34643691 and rs149721630) replicated (p<0.05), representing signals near the NTRK3 and ABCA13 genes. Conclusions Our study has identified reproducible associations with asthma exacerbations in the UK Biobank and GERA cohorts. Confirmation of these findings in different asthma subphenotypes in diverse ancestries and functional investigation will be required to understand their mechanisms of action and potentially inform therapeutic development.
Collapse
Affiliation(s)
- Ahmed Edris
- Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, UK
| | - Kirsten Voorhies
- Precision Medicine Translational Research Center, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Sharon M. Lutz
- Precision Medicine Translational Research Center, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Carlos Iribarren
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Ian Hall
- Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Ann Chen Wu
- Precision Medicine Translational Research Center, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Martin Tobin
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, UK
| | - Katherine Fawcett
- Genetic Epidemiology Group, Department of Health Sciences, University of Leicester, Leicester, UK
- These authors contributed equally
| | - Lies Lahousse
- Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
- These authors contributed equally
| |
Collapse
|
4
|
Lima DDS, de Morais RV, Rechenmacher C, Michalowski MB, Goldani MZ. Epigenetics, hypersensibility and asthma: what do we know so far? Clinics (Sao Paulo) 2023; 78:100296. [PMID: 38043345 DOI: 10.1016/j.clinsp.2023.100296] [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: 06/15/2023] [Revised: 09/26/2023] [Accepted: 10/04/2023] [Indexed: 12/05/2023] Open
Abstract
In this review, we describe recent advances in understanding the relationship between epigenetic changes, especially DNA methylation (DNAm), with hypersensitivity and respiratory disorders such as asthma in childhood. It is clearly described that epigenetic mechanisms can induce short to long-term changes in cells, tissues, and organs. Through the growing number of studies on the Origins of Health Development and Diseases, more and more data exist on how environmental and genomic aspects in early life can induce allergies and asthma. The lack of biomarkers, standardized assays, and access to more accessible tools for data collection and analysis are still a challenge for future studies. Through this review, the authors draw a panorama with the available information that can assist in the establishment of an epigenetic approach for the risk analysis of these pathologies.
Collapse
Affiliation(s)
- Douglas da Silva Lima
- Programa de Pós-Graduação em Saúde da Criança e do Adolescente, Departamento de Pediatria, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Pediatria Translacional, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Rahuany Velleda de Morais
- Laboratório de Pediatria Translacional, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Ciliana Rechenmacher
- Programa de Pós-Graduação em Saúde da Criança e do Adolescente, Departamento de Pediatria, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Pediatria Translacional, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Mariana Bohns Michalowski
- Programa de Pós-Graduação em Saúde da Criança e do Adolescente, Departamento de Pediatria, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Pediatria Translacional, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Serviço de Oncologia Pediátrica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil.
| | - Marcelo Zubaran Goldani
- Programa de Pós-Graduação em Saúde da Criança e do Adolescente, Departamento de Pediatria, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Laboratório de Pediatria Translacional, Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Faculdade de Medicina, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
5
|
Makrinioti H, Morita H, Nastouli E, Jartti T. Editorial: Bridging the gap between immunology, virology, genetics, and epigenetics in bronchiolitis: The multiomics pathway to asthma development. Front Immunol 2023; 14:1154121. [PMID: 36895569 PMCID: PMC9989251 DOI: 10.3389/fimmu.2023.1154121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/23/2023] Open
Affiliation(s)
- Heidi Makrinioti
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Hideaki Morita
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.,Allergy Center, National Center for Child Health and Development, Tokyo, Japan
| | - Eleni Nastouli
- Department of Infection, Immunity and Inflammation, University College London (UCL) Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Tuomas Jartti
- Research unit for Pediatrics, Pediatric Neurology, Pediatric Surgery, Child Psychiatry, Dermatology, Clinical Genetics, Obstetrics and Gynecology, Otorhinolaryngology and Opthalmology (PEDEGO) Research Unit, Medical Research Center, University of Oulu, Oulu, Finland.,Department of Pediatrics, Oulu University Hospital, Oulu, Finland.,Department of Paediatrics, Turku University Hospital and Turku University, Turku, Finland
| |
Collapse
|
6
|
Kim SR. Viral Infection and Airway Epithelial Immunity in Asthma. Int J Mol Sci 2022; 23:9914. [PMID: 36077310 PMCID: PMC9456547 DOI: 10.3390/ijms23179914] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 12/19/2022] Open
Abstract
Viral respiratory tract infections are associated with asthma development and exacerbation in children and adults. In the course of immune responses to viruses, airway epithelial cells are the initial platform of innate immunity against viral invasion. Patients with severe asthma are more vulnerable than those with mild to moderate asthma to viral infections. Furthermore, in most cases, asthmatic patients tend to produce lower levels of antiviral cytokines than healthy subjects, such as interferons produced from immune effector cells and airway epithelial cells. The epithelial inflammasome appears to contribute to asthma exacerbation through overactivation, leading to self-damage, despite its naturally protective role against infectious pathogens. Given the mixed and complex immune responses in viral-infection-induced asthma exacerbation, this review examines the diverse roles of airway epithelial immunity and related potential therapeutic targets and discusses the mechanisms underlying the heterogeneous manifestations of asthma exacerbations.
Collapse
Affiliation(s)
- So Ri Kim
- Division of Respiratory Medicine and Allergy, Department of Internal Medicine, Medical School of Jeonbuk National University, 20 Geonji-ro, Deokjin-gu, Jeonju 54907, Korea
| |
Collapse
|
7
|
Pischedda S, Rivero-Calle I, Gómez-Carballa A, Cebey-López M, Barral-Arca R, Gómez-Rial J, Pardo-Seco J, Curras-Tuala MJ, Viz-Lasheras S, Bello X, Crujeiras AB, Diaz-Lagares A, González-López MT, Martinón-Torres F, Salas A. Role and Diagnostic Performance of Host Epigenome in Respiratory Morbidity after RSV Infection: The EPIRESVi Study. Front Immunol 2022; 13:875691. [PMID: 35619695 PMCID: PMC9128527 DOI: 10.3389/fimmu.2022.875691] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/04/2022] [Indexed: 11/20/2022] Open
Abstract
Background Respiratory syncytial virus (RSV) infection has been associated with the subsequent development of recurrent wheezing and asthma, although the mechanisms involved are still unknown. We investigate the role of epigenetics in the respiratory morbidity after infection by comparing methylation patterns from children who develop recurrent wheezing (RW-RSV), subsequent asthma (AS-RVS), and those experiencing complete recovery (CR-RSV). Methods Prospective, observational study of infants aged < 2 years with RSV respiratory infection admitted to hospital and followed-up after discharge for at least three years. According to their clinical course, patients were categorized into subgroups: RW-RSV (n = 36), AS-RSV (n = 9), and CR-RSV (n = 32). The DNA genome-wide methylation pattern was analyzed in whole blood samples, collected during the acute phase of the infection, using the Illumina Infinium Methylation EPIC BeadChip (850K CpG sites). Differences in methylation were determined through a linear regression model adjusted for age, gender and cell composition. Results Patients who developed respiratory sequelae showed a statistically significant higher proportion of NK and CD8T cells (inferred through a deconvolution approach) than those with complete recovery. We identified 5,097 significant differentially methylated positions (DMPs) when comparing RW-RSV and AS-RVS together against CR-RSV. Methylation profiles affect several genes involved in airway inflammation processes. The most significant DMPs were found to be hypomethylated in cases and therefore generally leading to overexpression of affected genes. The lead CpG position (cg24509398) falls at the gene body of EYA3 (P-value = 2.77×10-10), a tyrosine phosphatase connected with pulmonary vascular remodeling, a key process in the asthma pathology. Logistic regression analysis resulted in a diagnostic epigenetic signature of 3-DMPs (involving genes ZNF2698, LOC102723354 and RPL15/NKIRAS1) that allows to efficiently differentiate sequelae cases from CR-RSV patients (AUC = 1.00). Enrichment pathway analysis reveals the role of the cell cycle checkpoint (FDR P-value = 4.71×10-2), DNA damage (FDP-value = 2.53×10-2), and DNA integrity checkpoint (FDR P-value = 2.56×10-2) in differentiating sequelae from CR-RSV patients. Conclusions Epigenetic mechanisms might play a fundamental role in the long-term sequelae after RSV infection, contributing to explain the different phenotypes observed.
Collapse
Affiliation(s)
- Sara Pischedda
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,GenPoB Research Group, Instituto de Investigacinó Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Unidade de Xenética, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Irene Rivero-Calle
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Alberto Gómez-Carballa
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,GenPoB Research Group, Instituto de Investigacinó Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Unidade de Xenética, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Miriam Cebey-López
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,GenPoB Research Group, Instituto de Investigacinó Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Unidade de Xenética, Santiago de Compostela, Spain
| | - Ruth Barral-Arca
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,GenPoB Research Group, Instituto de Investigacinó Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Unidade de Xenética, Santiago de Compostela, Spain
| | - Jose Gómez-Rial
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Jacobo Pardo-Seco
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,GenPoB Research Group, Instituto de Investigacinó Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Unidade de Xenética, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - María-José Curras-Tuala
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,GenPoB Research Group, Instituto de Investigacinó Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Unidade de Xenética, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Sandra Viz-Lasheras
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,GenPoB Research Group, Instituto de Investigacinó Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Unidade de Xenética, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Xabier Bello
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,GenPoB Research Group, Instituto de Investigacinó Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Unidade de Xenética, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Ana B Crujeiras
- Epigenomics in Endocrinology and Nutrition Group, Epigenomics Unit, Instituto De Investigación Sanitaria De Santiago De Compostela (IDIS), Complejo Hospitalario Universitario De Santiago De Compostela (CHUS/SERGAS), Santiago de Compostela, Spain.,Centro De Investigación Biomédica En Red Fisiopatología De La Obesidad Y Nutrición (Ciberobn), Madrid, Spain
| | - Angel Diaz-Lagares
- Cancer Epigenomics, Epigenomics Unit, Translational Medical Oncology (Oncomet), Instituto De Investigación Sanitaria De Santiago De Compostela (IDIS), Complejo Hospitalario Universitario De Santiago De Compostela (CHUS/SERGAS), Santiago De Compostela, Spain.,Centro De Investigación Biomédica En Red Cancer (CIBERONC), Madrid, Spain
| | | | - Federico Martinón-Torres
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Antonio Salas
- Genetics, Vaccines, Infectious Diseases and Pediatrics Research Group (GENVIP), Instituto de Investigación Sanitaria de Santiago, Santiago de Compostela, Santiago de Compostela, Spain.,GenPoB Research Group, Instituto de Investigacinó Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Unidade de Xenética, Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain.,Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | | |
Collapse
|
8
|
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: 40] [Impact Index Per Article: 13.3] [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
|
9
|
Noureddine N, Chalubinski M, Wawrzyniak P. The Role of Defective Epithelial Barriers in Allergic Lung Disease and Asthma Development. J Asthma Allergy 2022; 15:487-504. [PMID: 35463205 PMCID: PMC9030405 DOI: 10.2147/jaa.s324080] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/06/2022] [Indexed: 12/15/2022] Open
Abstract
The respiratory epithelium constitutes the physical barrier between the human body and the environment, thus providing functional and immunological protection. It is often exposed to allergens, microbial substances, pathogens, pollutants, and environmental toxins, which lead to dysregulation of the epithelial barrier and result in the chronic inflammation seen in allergic diseases and asthma. This epithelial barrier dysfunction results from the disturbed tight junction formation, which are multi-protein subunits that promote cell-cell adhesion and barrier integrity. The increasing interest and evidence of the role of impaired epithelial barrier function in allergy and asthma highlight the need for innovative approaches that can provide new knowledge in this area. Here, we review and discuss the current role and mechanism of epithelial barrier dysfunction in developing allergic diseases and the effect of current allergy therapies on epithelial barrier restoration.
Collapse
Affiliation(s)
- Nazek Noureddine
- Division of Clinical Chemistry and Biochemistry, University Children’s Hospital Zurich, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
- Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Maciej Chalubinski
- Department of Immunology and Allergy, Medical University of Lodz, Lodz, Poland
| | - Paulina Wawrzyniak
- Division of Clinical Chemistry and Biochemistry, University Children’s Hospital Zurich, Zurich, Switzerland
- Children’s Research Center, University Children’s Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
10
|
Bermick J, Schaller M. Epigenetic regulation of pediatric and neonatal immune responses. Pediatr Res 2022; 91:297-327. [PMID: 34239066 DOI: 10.1038/s41390-021-01630-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/01/2021] [Accepted: 06/09/2021] [Indexed: 02/06/2023]
Abstract
Epigenetic regulation of transcription is a collective term that refers to mechanisms known to regulate gene transcription without changing the underlying DNA sequence. These mechanisms include DNA methylation and histone tail modifications which influence chromatin accessibility, and microRNAs that act through post-transcriptional gene silencing. Epigenetics is known to regulate a variety of biological processes, and the role of epigtenetics in immunity and immune-mediated diseases is becoming increasingly recognized. While DNA methylation is the most widely studied, each of these systems play an important role in the development and maintenance of appropriate immune responses. There is clear evidence that epigenetic mechanisms contribute to developmental stage-specific immune responses in a cell-specific manner. There is also mounting evidence that prenatal exposures alter epigenetic profiles and subsequent immune function in exposed offspring. Early life exposures that are associated with poor long-term health outcomes also appear to impact immune specific epigenetic patterning. Finally, each of these epigenetic mechanisms contribute to the pathogenesis of a wide variety of diseases that manifest during childhood. This review will discuss each of these areas in detail. IMPACT: Epigenetics, including DNA methylation, histone tail modifications, and microRNA expression, dictate immune cell phenotypes. Epigenetics influence immune development and subsequent immune health. Prenatal, perinatal, and postnatal exposures alter immune cell epigenetic profiles and subsequent immune function. Numerous pediatric-onset diseases have an epigenetic component. Several successful strategies for childhood diseases target epigenetic mechanisms.
Collapse
Affiliation(s)
- Jennifer Bermick
- Department of Pediatrics, Division of Neonatology, University of Iowa, Iowa City, IA, USA. .,Iowa Inflammation Program, University of Iowa, Iowa City, IA, USA.
| | - Matthew Schaller
- Department of Pulmonary, Critical Care & Sleep Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
11
|
Hayashi K, Anzai N. L-type amino acid transporter 1 as a target for inflammatory disease and cancer immunotherapy. J Pharmacol Sci 2021; 148:31-40. [PMID: 34924127 DOI: 10.1016/j.jphs.2021.09.006] [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] [Received: 06/22/2021] [Revised: 08/11/2021] [Accepted: 09/21/2021] [Indexed: 12/23/2022] Open
Abstract
Ingestion of amino acids is fundamental for cellular activity. Amino acids are important components for protein synthesis but are also crucial for intracellular metabolic reactions and signal transduction. Following activation, immune cells induce metabolic reprogramming to generate adequate energy and constitutive substances. Hence, the delivery of amino acids by transporters is necessary for the progression of metabolic rewiring. In this review, we discuss how amino acids and their transporters regulate immune cell functions, with emphasis on LAT1, a transporter of large neutral amino acids. Furthermore, we explore the possibility of targeting amino acid transporters to improve immune disorders and cancer immune therapies.
Collapse
Affiliation(s)
- Keitaro Hayashi
- Department of Pharmacology and Toxicology, Dokkyo Medical University School of Medicine, Shimotsuga, Japan.
| | - Naohiko Anzai
- Department of Pharmacology and Toxicology, Dokkyo Medical University School of Medicine, Shimotsuga, Japan; Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
12
|
Watkinson RL, Looi K, Laing IA, Cianferoni A, Kicic A. Viral Induced Effects on a Vulnerable Epithelium; Lessons Learned From Paediatric Asthma and Eosinophilic Oesophagitis. Front Immunol 2021; 12:773600. [PMID: 34912343 PMCID: PMC8666438 DOI: 10.3389/fimmu.2021.773600] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/05/2021] [Indexed: 01/07/2023] Open
Abstract
The epithelium is integral to the protection of many different biological systems and for the maintenance of biochemical homeostasis. Emerging evidence suggests that particular children have epithelial vulnerabilities leading to dysregulated barrier function and integrity, that resultantly contributes to disease pathogenesis. These epithelial vulnerabilities likely develop in utero or in early life due to various genetic, epigenetic and environmental factors. Although various epithelia are uniquely structured with specific function, prevalent allergic-type epithelial diseases in children potentially have common or parallel disease processes. These include inflammation and immune response dysregulation stemming from atypical epithelial barrier function and integrity. Two diseases where aetiology and pathogenesis are potentially linked to epithelial vulnerabilities include Paediatric Asthma and Eosinophilic Oesophagitis (EoE). For example, rhinovirus C (RV-C) is a known risk factor for paediatric asthma development and is known to disrupt respiratory epithelial barrier function causing acute inflammation. In addition, EoE, a prevalent atopic condition of the oesophageal epithelium, is characterised by similar innate immune and epithelial responses to viral injury. This review examines the current literature and identifies the gaps in the field defining viral-induced effects on a vulnerable respiratory epithelium and resulting chronic inflammation, drawing from knowledge generated in acute wheezing illness, paediatric asthma and EoE. Besides highlighting the importance of epithelial structure and barrier function in allergic disease pathogenesis regardless of specific epithelial sub-types, this review focuses on the importance of examining other parallel allergic-type disease processes that may uncover commonalities driving disease pathogenesis. This in turn may be beneficial in the development of common therapeutics for current clinical management and disease prevention in the future.
Collapse
Affiliation(s)
- Rebecca L Watkinson
- Division of Paediatrics, Medical School, The University of Western Australia, Nedlands, WA, Australia.,Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Kevin Looi
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA, Australia.,School of Public Health, Curtin University, Bentley, WA, Australia
| | - Ingrid A Laing
- Division of Paediatrics, Medical School, The University of Western Australia, Nedlands, WA, Australia.,Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Antonella Cianferoni
- Pediatrics Department, Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Anthony Kicic
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA, Australia.,School of Public Health, Curtin University, Bentley, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
13
|
Role of Epigenetics in the Pathogenesis, Treatment, Prediction, and Cellular Transformation of Asthma. Mediators Inflamm 2021; 2021:9412929. [PMID: 34566492 PMCID: PMC8457970 DOI: 10.1155/2021/9412929] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/27/2021] [Indexed: 12/15/2022] Open
Abstract
Asthma is a mysterious disease with heterogeneity in etiology, pathogenesis, and clinical phenotypes. Although ongoing studies have provided a better understanding of asthma, its natural history, progression, pathogenesis, diversified phenotypes, and even the exact epigenetic linkage between childhood asthma and adult-onset/old age asthma remain elusive in many aspects. Asthma heritability has been established through genetic studies, but genetics is not the only influencing factor in asthma. The increasing incidence and some unsolved queries suggest that there may be other elements related to asthma heredity. Epigenetic mechanisms link genetic and environmental factors with developmental trajectories in asthma. This review provides an overview of asthma epigenetics and its components, including several epigenetic studies on asthma, and discusses the epigenetic linkage between childhood asthma and adult-onset/old age asthma. Studies involving asthma epigenetics present valuable novel approaches to solve issues related to asthma. Asthma epigenetic research guides us towards gene therapy and personalized T cell therapy, directs the discovery of new therapeutic agents, predicts long-term outcomes in severe cases, and is also involved in the cellular transformation of childhood asthma to adult-onset/old age asthma.
Collapse
|
14
|
Combined prenatal Lactobacillus reuteri and ω-3 supplementation synergistically modulates DNA methylation in neonatal T helper cells. Clin Epigenetics 2021; 13:135. [PMID: 34193262 PMCID: PMC8247185 DOI: 10.1186/s13148-021-01115-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/15/2021] [Indexed: 02/08/2023] Open
Abstract
Background Environmental exposures may alter DNA methylation patterns of T helper cells. As T helper cells are instrumental for allergy development, changes in methylation patterns may constitute a mechanism of action for allergy preventive interventions. While epigenetic effects of separate perinatal probiotic or ω-3 fatty acid supplementation have been studied previously, the combined treatment has not been assessed. We aimed to investigate epigenome-wide DNA methylation patterns from a sub-group of children in an on-going randomised double-blind placebo-controlled allergy prevention trial using pre- and postnatal combined Lactobacillus reuteri and ω-3 fatty acid treatment. To this end, > 866000 CpG sites (MethylationEPIC 850K array) in cord blood CD4+ T cells were examined in samples from all four study arms (double-treatment: n = 18, single treatments: probiotics n = 16, ω-3 n = 15, and double placebo: n = 14). Statistical and bioinformatic analyses identified treatment-associated differentially methylated CpGs and genes, which were used to identify putatively treatment-induced network modules. Pathway analyses inferred biological relevance, and comparisons were made to an independent allergy data set. Results Comparing the active treatments to the double placebo group, most differentially methylated CpGs and genes were hypermethylated, possibly suggesting induction of transcriptional inhibition. The double-treated group showed the largest number of differentially methylated CpGs, of which many were unique, suggesting synergy between interventions. Clusters within the double-treated network module consisted of immune-related pathways, including T cell receptor signalling, and antigen processing and presentation, with similar pathways revealed for the single-treatment modules. CpGs derived from differential methylation and network module analyses were enriched in an independent allergy data set, particularly in the double-treatment group, proposing treatment-induced DNA methylation changes as relevant for allergy development. Conclusion Prenatal L. reuteri and/or ω-3 fatty acid treatment results in hypermethylation and affects immune- and allergy-related pathways in neonatal T helper cells, with potentially synergistic effects between the interventions and relevance for allergic disease. Further studies need to address these findings on a transcriptional level, and whether the results associate to allergy development in the children. Understanding the role of DNA methylation in regulating effects of perinatal probiotic and ω-3 interventions may provide essential knowledge in the development of efficacious allergy preventive strategies. Trial registration ClinicalTrials.gov, ClinicalTrials.gov-ID: NCT01542970. Registered 27th of February 2012—Retrospectively registered, https://clinicaltrials.gov/ct2/show/NCT01542970. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-021-01115-4.
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW Asthma is the most common chronic disease in pediatric age. Childhood-onset asthma, as opposed to adult-onset asthma, is typically characterized by a personal and often a family history of atopy and related markers of type 2-mediated inflammation. However, the interplay between atopy and asthma development is more complex than a linear dose-response relationship. RECENT FINDINGS Family and personal history of atopic diseases have been confirmed as major risk factors for asthma occurrence and persistence in children. Early life and multiple sensitizations to aeroallergens significantly increase the risk of asthma development in school age. Early life lower respiratory tract viral infections, especially caused by rhinovirus, also increase the susceptibility to atopic asthma in childhood. Human rhinovirus type C receptor CDHR3 polymorphisms have been shown to affect receptor epithelial expression, activation, and asthma development and exacerbation severity in children. Atopic sensitization and respiratory viral infections can synergistically enhance the susceptibility to asthma through multiple mechanisms, including the IgE-mediated inhibition of innate antiviral responses to rhinovirus. Emerging evidence shows that several nonatopic factors are also involved in the asthma pathogenesis in genetically predisposed individuals, including early life exposure to environmental factors, and lung and gut microbiome composition. SUMMARY The current review outlines recent data on the complex role of atopy in asthma pathogenesis and persistence, and addresses new research topics such as the role of epigenetics and the lung microbiome.
Collapse
|
16
|
Nemani SSP, Vermeulen CJ, Pech M, Faiz A, Oliver BGG, van den Berge M, Burgess JK, Kopp MV, Weckmann M. COL4A3 expression in asthmatic epithelium depends on intronic methylation and ZNF263 binding. ERJ Open Res 2021; 7:00802-2020. [PMID: 34109240 PMCID: PMC8181658 DOI: 10.1183/23120541.00802-2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 03/18/2021] [Indexed: 11/23/2022] Open
Abstract
Background Reduction of COL4A3, one of the six isoforms of collagen 4, in asthmatic airways results in increased inflammation and angiogenesis, implicating it as a central part of asthma pathogenesis. However, to date, the path underlying these diminished COL4A3 levels has been elusive. This study investigated a possible mechanism underlying the reduction of COL4A3 expression. Methods Bronchial biopsies of 76 patients with asthma and 83 controls were subjected to RNA-sequencing and DNA methylation bead arrays to identify expression and methylation changes. The binding of ZNF263 was analysed by chromatin-immunoprecipitation sequencing coupled with quantitative (q)PCR. Effects of ZNF263 silencing, using small interfering RNA, on the COL4A3 expression were studied using qPCR. Results COL4A3 expression was significantly reduced in bronchial biopsies compared to healthy controls, whereas DNA methylation levels at cg11797365 were increased. COL4A3 expression levels were significantly low in asthmatics without inhaled corticosteroid (ICS) use, whereas the expression was not statistically different between asthmatics using ICS and controls. Methylation levels at cg11797365 in vitro were increased upon consecutive rhinovirus infections. Conclusion Our data indicate an epigenetic modification as a contributing factor for the loss of COL4A3 expression in asthmatic airway epithelium. An epigenetic modification interrupts ZNF263 binding, which may contribute to the loss of COL4A3 expression in asthmatic airway epitheliumhttps://bit.ly/39cZbyn
Collapse
Affiliation(s)
- Sai Sneha Priya Nemani
- Division of Paediatric Pneumology and Allergology, University Medical Centre Schleswig-Holstein, Airway Research Centre North, member of the German Centre for Lung Research (DZL), Lübeck, Germany
| | - Cornelis Joseph Vermeulen
- Dept of Pulmonary Diseases, University Medical Centre Groningen, GRIAC (Groningen Research Institute for Asthma and COPD), University of Groningen, Groningen, The Netherlands
| | - Martin Pech
- Division of Paediatric Pneumology and Allergology, University Medical Centre Schleswig-Holstein, Airway Research Centre North, member of the German Centre for Lung Research (DZL), Lübeck, Germany
| | - Alen Faiz
- Dept of Pulmonary Diseases, University Medical Centre Groningen, GRIAC (Groningen Research Institute for Asthma and COPD), University of Groningen, Groningen, The Netherlands.,Dept of Pathology and Medical Biology, University Medical Centre Groningen, GRIAC, University of Groningen, Groningen, The Netherlands.,Woolcock Institute of Medical Research, The University of Sydney, Glebe, NSW, Australia.,School of Life Sciences, University of Technology, Sydney, NSW, Australia
| | - Brian George G Oliver
- Woolcock Institute of Medical Research, The University of Sydney, Glebe, NSW, Australia.,School of Life Sciences, University of Technology, Sydney, NSW, Australia
| | - Maarten van den Berge
- Dept of Pulmonary Diseases, University Medical Centre Groningen, GRIAC (Groningen Research Institute for Asthma and COPD), University of Groningen, Groningen, The Netherlands
| | - Janette Kay Burgess
- Dept of Pathology and Medical Biology, University Medical Centre Groningen, GRIAC, University of Groningen, Groningen, The Netherlands.,Woolcock Institute of Medical Research, The University of Sydney, Glebe, NSW, Australia.,Discipline of Pharmacology, Faculty of Medicine, The University of Sydney, NSW, Australia
| | - Matthias V Kopp
- Division of Paediatric Pneumology and Allergology, University Medical Centre Schleswig-Holstein, Airway Research Centre North, member of the German Centre for Lung Research (DZL), Lübeck, Germany.,Pediatric Respiratory Medicine, Dept of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Markus Weckmann
- Division of Paediatric Pneumology and Allergology, University Medical Centre Schleswig-Holstein, Airway Research Centre North, member of the German Centre for Lung Research (DZL), Lübeck, Germany
| |
Collapse
|
17
|
Fiuza BSD, Fonseca HF, Meirelles PM, Marques CR, da Silva TM, Figueiredo CA. Understanding Asthma and Allergies by the Lens of Biodiversity and Epigenetic Changes. Front Immunol 2021; 12:623737. [PMID: 33732246 PMCID: PMC7957070 DOI: 10.3389/fimmu.2021.623737] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Exposure to different organisms (bacteria, mold, virus, protozoan, helminths, among others) can induce epigenetic changes affecting the modulation of immune responses and consequently increasing the susceptibility to inflammatory diseases. Epigenomic regulatory features are highly affected during embryonic development and are responsible for the expression or repression of different genes associated with cell development and targeting/conducting immune responses. The well-known, "window of opportunity" that includes maternal and post-natal environmental exposures, which include maternal infections, microbiota, diet, drugs, and pollutant exposures are of fundamental importance to immune modulation and these events are almost always accompanied by epigenetic changes. Recently, it has been shown that these alterations could be involved in both risk and protection of allergic diseases through mechanisms, such as DNA methylation and histone modifications, which can enhance Th2 responses and maintain memory Th2 cells or decrease Treg cells differentiation. In addition, epigenetic changes may differ according to the microbial agent involved and may even influence different asthma or allergy phenotypes. In this review, we discuss how exposure to different organisms, including bacteria, viruses, and helminths can lead to epigenetic modulations and how this correlates with allergic diseases considering different genetic backgrounds of several ancestral populations.
Collapse
Affiliation(s)
| | | | - Pedro Milet Meirelles
- Instituto de Biologia, Universidade Federal da Bahia, Salvador, Brazil
- Instituto Nacional de Ciência e Tecnologia em Estudos Interdisciplinares e Transdisciplinares em Ecologia e Evolução (IN-TREE), Salvador, Brazil
| | - Cintia Rodrigues Marques
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista, Brazil
| | | | | |
Collapse
|
18
|
Acevedo N, Alashkar Alhamwe B, Caraballo L, Ding M, Ferrante A, Garn H, Garssen J, Hii CS, Irvine J, Llinás-Caballero K, López JF, Miethe S, Perveen K, Pogge von Strandmann E, Sokolowska M, Potaczek DP, van Esch BCAM. Perinatal and Early-Life Nutrition, Epigenetics, and Allergy. Nutrients 2021; 13:724. [PMID: 33668787 PMCID: PMC7996340 DOI: 10.3390/nu13030724] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/08/2021] [Accepted: 02/12/2021] [Indexed: 02/08/2023] Open
Abstract
Epidemiological studies have shown a dramatic increase in the incidence and the prevalence of allergic diseases over the last several decades. Environmental triggers including risk factors (e.g., pollution), the loss of rural living conditions (e.g., farming conditions), and nutritional status (e.g., maternal, breastfeeding) are considered major contributors to this increase. The influences of these environmental factors are thought to be mediated by epigenetic mechanisms which are heritable, reversible, and biologically relevant biochemical modifications of the chromatin carrying the genetic information without changing the nucleotide sequence of the genome. An important feature characterizing epigenetically-mediated processes is the existence of a time frame where the induced effects are the strongest and therefore most crucial. This period between conception, pregnancy, and the first years of life (e.g., first 1000 days) is considered the optimal time for environmental factors, such as nutrition, to exert their beneficial epigenetic effects. In the current review, we discussed the impact of the exposure to bacteria, viruses, parasites, fungal components, microbiome metabolites, and specific nutritional components (e.g., polyunsaturated fatty acids (PUFA), vitamins, plant- and animal-derived microRNAs, breast milk) on the epigenetic patterns related to allergic manifestations. We gave insight into the epigenetic signature of bioactive milk components and the effects of specific nutrition on neonatal T cell development. Several lines of evidence suggest that atypical metabolic reprogramming induced by extrinsic factors such as allergens, viruses, pollutants, diet, or microbiome might drive cellular metabolic dysfunctions and defective immune responses in allergic disease. Therefore, we described the current knowledge on the relationship between immunometabolism and allergy mediated by epigenetic mechanisms. The knowledge as presented will give insight into epigenetic changes and the potential of maternal and post-natal nutrition on the development of allergic disease.
Collapse
Affiliation(s)
- Nathalie Acevedo
- Institute for Immunological Research, University of Cartagena, Cartagena 130014, Colombia; (N.A.); (L.C.); (K.L.-C.); (J.F.L.)
| | - Bilal Alashkar Alhamwe
- Institute of Tumor Immunology, Clinic for Hematology, Oncology and Immunology, Center for Tumor Biology and Immunology, Philipps University Marburg, 35043 Marburg, Germany; (B.A.A.); (E.P.v.S.)
- College of Pharmacy, International University for Science and Technology (IUST), Daraa 15, Syria
| | - Luis Caraballo
- Institute for Immunological Research, University of Cartagena, Cartagena 130014, Colombia; (N.A.); (L.C.); (K.L.-C.); (J.F.L.)
| | - Mei Ding
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos, Switzerland; (M.D.); (M.S.)
- Christine Kühne-Center for Allergy Research and Education, 7265 Davos, Switzerland
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Antonio Ferrante
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia; (A.F.); (C.S.H.); (J.I.); (K.P.)
- Adelaide School of Medicine and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
- School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Holger Garn
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (H.G.); (S.M.)
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands;
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands
| | - Charles S. Hii
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia; (A.F.); (C.S.H.); (J.I.); (K.P.)
- Adelaide School of Medicine and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - James Irvine
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia; (A.F.); (C.S.H.); (J.I.); (K.P.)
- Adelaide School of Medicine and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - Kevin Llinás-Caballero
- Institute for Immunological Research, University of Cartagena, Cartagena 130014, Colombia; (N.A.); (L.C.); (K.L.-C.); (J.F.L.)
| | - Juan Felipe López
- Institute for Immunological Research, University of Cartagena, Cartagena 130014, Colombia; (N.A.); (L.C.); (K.L.-C.); (J.F.L.)
| | - Sarah Miethe
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (H.G.); (S.M.)
| | - Khalida Perveen
- Department of Immunopathology, SA Pathology at the Women’s and Children’s Hospital, North Adelaide, SA 5006, Australia; (A.F.); (C.S.H.); (J.I.); (K.P.)
- Adelaide School of Medicine and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - Elke Pogge von Strandmann
- Institute of Tumor Immunology, Clinic for Hematology, Oncology and Immunology, Center for Tumor Biology and Immunology, Philipps University Marburg, 35043 Marburg, Germany; (B.A.A.); (E.P.v.S.)
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos, Switzerland; (M.D.); (M.S.)
- Christine Kühne-Center for Allergy Research and Education, 7265 Davos, Switzerland
| | - Daniel P. Potaczek
- Translational Inflammation Research Division & Core Facility for Single Cell Multiomics, Medical Faculty, Philipps University Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center, 35043 Marburg, Germany; (H.G.); (S.M.)
| | - Betty C. A. M. van Esch
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CG Utrecht, The Netherlands;
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands
| |
Collapse
|
19
|
Cevhertas L, Ogulur I, Maurer DJ, Burla D, Ding M, Jansen K, Koch J, Liu C, Ma S, Mitamura Y, Peng Y, Radzikowska U, Rinaldi AO, Satitsuksanoa P, Globinska A, Veen W, Sokolowska M, Baerenfaller K, Gao Y, Agache I, Akdis M, Akdis CA. Advances and recent developments in asthma in 2020. Allergy 2020; 75:3124-3146. [PMID: 32997808 DOI: 10.1111/all.14607] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022]
Abstract
In this review, we discuss recent publications on asthma and review the studies that have reported on the different aspects of the prevalence, risk factors and prevention, mechanisms, diagnosis, and treatment of asthma. Many risk and protective factors and molecular mechanisms are involved in the development of asthma. Emerging concepts and challenges in implementing the exposome paradigm and its application in allergic diseases and asthma are reviewed, including genetic and epigenetic factors, microbial dysbiosis, and environmental exposure, particularly to indoor and outdoor substances. The most relevant experimental studies further advancing the understanding of molecular and immune mechanisms with potential new targets for the development of therapeutics are discussed. A reliable diagnosis of asthma, disease endotyping, and monitoring its severity are of great importance in the management of asthma. Correct evaluation and management of asthma comorbidity/multimorbidity, including interaction with asthma phenotypes and its value for the precision medicine approach and validation of predictive biomarkers, are further detailed. Novel approaches and strategies in asthma treatment linked to mechanisms and endotypes of asthma, particularly biologicals, are critically appraised. Finally, due to the recent pandemics and its impact on patient management, we discuss the challenges, relationships, and molecular mechanisms between asthma, allergies, SARS-CoV-2, and COVID-19.
Collapse
Affiliation(s)
- Lacin Cevhertas
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne‐Center for Allergy Research and Education (CK‐CARE) Davos Switzerland
- Department of Medical Immunology Institute of Health Sciences, Bursa Uludag University Bursa Turkey
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Faculty of Medicine, Division of Pediatric Allergy and Immunology Marmara University Istanbul Turkey
| | - Debbie J. Maurer
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Daniel Burla
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Mei Ding
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne‐Center for Allergy Research and Education (CK‐CARE) Davos Switzerland
- Department of Allergology Zhongnan Hospital of Wuhan University Wuhan Hubei China
| | - Kirstin Jansen
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Jana Koch
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Swiss Institute for Bioinformatics (SIB) Davos Switzerland
| | - Chengyao Liu
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Department of Otolaryngology Head and Neck Surgery Beijing TongRen HospitalCapital Medical University Beijing China
| | - Siyuan Ma
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Department of Otolaryngology Head and Neck Surgery Beijing TongRen HospitalCapital Medical University Beijing China
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Yaqi Peng
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Otorhinolaryngology HospitalThe First Affiliated HospitalSun Yat‐sen University Guangzhou China
| | - Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne‐Center for Allergy Research and Education (CK‐CARE) Davos Switzerland
- Department of Regenerative Medicine and Immune Regulation Medical University of Bialystok Bialystok Poland
| | - Arturo O. Rinaldi
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Pattraporn Satitsuksanoa
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne‐Center for Allergy Research and Education (CK‐CARE) Davos Switzerland
| | - Anna Globinska
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Willem Veen
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne‐Center for Allergy Research and Education (CK‐CARE) Davos Switzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Katja Baerenfaller
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Swiss Institute for Bioinformatics (SIB) Davos Switzerland
| | - Ya‐dong Gao
- Department of Allergology Zhongnan Hospital of Wuhan University Wuhan Hubei China
| | - Ioana Agache
- Faculty of Medicine Transylvania University Brasov Romania
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne‐Center for Allergy Research and Education (CK‐CARE) Davos Switzerland
| |
Collapse
|
20
|
Wirz OF, Üzülmez Ö, Jansen K, Veen W, Lammela A, Kainulainen L, Vuorinen T, Breiteneder H, Akdis CA, Jartti T, Akdis M. Increased antiviral response in circulating lymphocytes from hypogammaglobulinemia patients. Allergy 2020; 75:3147-3158. [PMID: 32533713 DOI: 10.1111/all.14445] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/21/2020] [Accepted: 04/29/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND B cells play a crucial role during rhinovirus (RV) infections by production of virus-neutralizing antibodies. A main feature of common variable immunodeficiency (CVID) is hypogammaglobulinemia (HG). HG patients have severely reduced levels of antibody-producing B cells and suffer from prolonged virus infections. Here, we addressed whether antiviral response of peripheral blood lymphocytes differs between HG patients and healthy individuals during natural RV infection. METHODS Using fluorescence-activated cell sorting, B-cell subsets were analyzed. Simultaneously, CD19 + B cells, CD14 + monocytes, and CD3 + T cells were sorted from frozen peripheral blood mononuclear cells from 11 RV-infected hypogammaglobulinemia patients, 7 RV-infected control subjects, and 14 noninfected control subjects. Real-time PCR was used to study expression of antiviral genes. A pan-RV PCR was used to detect RV genome in all samples. RESULTS In HG patients, total B-cell numbers, as well as IgA + and IgG + switched memory B cells, were reduced while naïve B cells and T cells were increased. STAT1 expression was increased in HG patients compared to controls in all lymphocyte subsets analyzed. The expression of antiviral genes IFITM1 and MX1 correlated with STAT1 expression in B cells and monocytes. RV RNA was found in 88.9% of monocytes from infected HG patients, 85.7% of monocytes from infected controls, and 7.1% of monocytes from uninfected controls. CONCLUSIONS We demonstrate an increased antiviral response in B cells and monocytes in HG patients and their correlation with STAT1 expression. Monocytes of infected HG patients and infected non-HG controls carry RV RNA.
Collapse
Affiliation(s)
- Oliver F. Wirz
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Öykü Üzülmez
- Department of Pathophysiology and Allergy Research Medical University of Vienna Vienna Austria
| | - Kirstin Jansen
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Willem Veen
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne—Center for Allergy Research and Education (CK‐CARE) Davos Switzerland
| | - Auli Lammela
- Department of Pediatrics and Adolescent Medicine Turku University Hospital and University of Turku Turku Finland
| | - Leena Kainulainen
- Department of Pediatrics and Adolescent Medicine Turku University Hospital and University of Turku Turku Finland
| | - Tytti Vuorinen
- Department of Clinical Virology Turku University Hospital Turku Finland
- Department of Virology University of Turku Turku Finland
| | - Heimo Breiteneder
- Department of Pathophysiology and Allergy Research Medical University of Vienna Vienna Austria
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne—Center for Allergy Research and Education (CK‐CARE) Davos Switzerland
| | - Tuomas Jartti
- Department of Pediatrics and Adolescent Medicine Turku University Hospital and University of Turku Turku Finland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| |
Collapse
|
21
|
The Airway Epithelium-A Central Player in Asthma Pathogenesis. Int J Mol Sci 2020; 21:ijms21238907. [PMID: 33255348 PMCID: PMC7727704 DOI: 10.3390/ijms21238907] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/20/2020] [Accepted: 11/21/2020] [Indexed: 12/11/2022] Open
Abstract
Asthma is a chronic inflammatory airway disease characterized by variable airflow obstruction in response to a wide range of exogenous stimuli. The airway epithelium is the first line of defense and plays an important role in initiating host defense and controlling immune responses. Indeed, increasing evidence indicates a range of abnormalities in various aspects of epithelial barrier function in asthma. A central part of this impairment is a disruption of the airway epithelial layer, allowing inhaled substances to pass more easily into the submucosa where they may interact with immune cells. Furthermore, many of the identified susceptibility genes for asthma are expressed in the airway epithelium. This review focuses on the biology of the airway epithelium in health and its pathobiology in asthma. We will specifically discuss external triggers such as allergens, viruses and alarmins and the effect of type 2 inflammatory responses on airway epithelial function in asthma. We will also discuss epigenetic mechanisms responding to external stimuli on the level of transcriptional and posttranscriptional regulation of gene expression, as well the airway epithelium as a potential treatment target in asthma.
Collapse
|
22
|
Asthma genomics and pharmacogenomics. Curr Opin Immunol 2020; 66:136-142. [PMID: 33171417 DOI: 10.1016/j.coi.2020.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 10/04/2020] [Indexed: 12/16/2022]
Abstract
In this review, we summarize recent published work interrogating the relationship between genetic variation or gene expression regulation across the genome and asthma or asthma treatment outcomes. This includes 11 genome-wide association studies of asthma phenotypes that collectively identified 64 novel loci; transcriptome-wide asthma association studies which identified genes involved in virus recognition, bacterial infection, lung tissue remodeling, eosinophilic and neutrophilic inflammation and genes in the chromosome 17q12 asthma susceptibility locus; and three epigenome-wide studies of asthma that had robust sample sizes and replicated findings. We also highlight pharmacogenomic studies of corticosteroids, bronchodilator response to albuterol and zileuton, although finding from these studies may still be preliminary due to their relatively small sample sizes and limited availability of replication cohorts.
Collapse
|
23
|
Groth EE, Weber M, Bahmer T, Pedersen F, Kirsten A, Börnigen D, Rabe KF, Watz H, Ammerpohl O, Goldmann T. Exploration of the sputum methylome and omics deconvolution by quadratic programming in molecular profiling of asthma and COPD: the road to sputum omics 2.0. Respir Res 2020; 21:274. [PMID: 33076907 PMCID: PMC7574293 DOI: 10.1186/s12931-020-01544-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/11/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND To date, most studies involving high-throughput analyses of sputum in asthma and COPD have focused on identifying transcriptomic signatures of disease. No whole-genome methylation analysis of sputum cells has been performed yet. In this context, the highly variable cellular composition of sputum has potential to confound the molecular analyses. METHODS Whole-genome transcription (Agilent Human 4 × 44 k array) and methylation (Illumina 450 k BeadChip) analyses were performed on sputum samples of 9 asthmatics, 10 healthy and 10 COPD subjects. RNA integrity was checked by capillary electrophoresis and used to correct in silico for bias conferred by RNA degradation during biobank sample storage. Estimates of cell type-specific molecular profiles were derived via regression by quadratic programming based on sputum differential cell counts. All analyses were conducted using the open-source R/Bioconductor software framework. RESULTS A linear regression step was found to perform well in removing RNA degradation-related bias among the main principal components of the gene expression data, increasing the number of genes detectable as differentially expressed in asthma and COPD sputa (compared to controls). We observed a strong influence of the cellular composition on the results of mixed-cell sputum analyses. Exemplarily, upregulated genes derived from mixed-cell data in asthma were dominated by genes predominantly expressed in eosinophils after deconvolution. The deconvolution, however, allowed to perform differential expression and methylation analyses on the level of individual cell types and, though we only analyzed a limited number of biological replicates, was found to provide good estimates compared to previously published data about gene expression in lung eosinophils in asthma. Analysis of the sputum methylome indicated presence of differential methylation in genomic regions of interest, e.g. mapping to a number of human leukocyte antigen (HLA) genes related to both major histocompatibility complex (MHC) class I and II molecules in asthma and COPD macrophages. Furthermore, we found the SMAD3 (SMAD family member 3) gene, among others, to lie within differentially methylated regions which has been previously reported in the context of asthma. CONCLUSIONS In this methodology-oriented study, we show that methylation profiling can be easily integrated into sputum analysis workflows and exhibits a strong potential to contribute to the profiling and understanding of pulmonary inflammation. Wherever RNA degradation is of concern, in silico correction can be effective in improving both sensitivity and specificity of downstream analyses. We suggest that deconvolution methods should be integrated in sputum omics analysis workflows whenever possible in order to facilitate the unbiased discovery and interpretation of molecular patterns of inflammation.
Collapse
Affiliation(s)
- Espen E Groth
- LungenClinic Grosshansdorf, Großhansdorf, Germany. .,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany. .,Department of Internal Medicine I, Pneumology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany. .,Department of Oncology, Hematology and BMT with Section Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Melanie Weber
- Program in Applied and Computational Mathematics, Princeton University, Princeton, NJ, USA
| | - Thomas Bahmer
- LungenClinic Grosshansdorf, Großhansdorf, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany.,Department of Internal Medicine I, Pneumology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Frauke Pedersen
- LungenClinic Grosshansdorf, Großhansdorf, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany.,Pulmonary Research Institute at LungenClinic Grosshansdorf, Großhansdorf, Germany
| | - Anne Kirsten
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany.,Pulmonary Research Institute at LungenClinic Grosshansdorf, Großhansdorf, Germany
| | - Daniela Börnigen
- Bioinformatics Core Unit, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus F Rabe
- LungenClinic Grosshansdorf, Großhansdorf, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany
| | - Henrik Watz
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany.,Pulmonary Research Institute at LungenClinic Grosshansdorf, Großhansdorf, Germany
| | - Ole Ammerpohl
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany.,Institute of Human Genetics, University Medical Center Ulm, Ulm, Germany
| | - Torsten Goldmann
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany.,Research Center Borstel, Pathology, Borstel, Germany
| |
Collapse
|
24
|
Heijink IH, Kuchibhotla VNS, Roffel MP, Maes T, Knight DA, Sayers I, Nawijn MC. Epithelial cell dysfunction, a major driver of asthma development. Allergy 2020; 75:1902-1917. [PMID: 32460363 PMCID: PMC7496351 DOI: 10.1111/all.14421] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 05/04/2020] [Accepted: 05/12/2020] [Indexed: 12/13/2022]
Abstract
Airway epithelial barrier dysfunction is frequently observed in asthma and may have important implications. The physical barrier function of the airway epithelium is tightly interwoven with its immunomodulatory actions, while abnormal epithelial repair responses may contribute to remodelling of the airway wall. We propose that abnormalities in the airway epithelial barrier play a crucial role in the sensitization to allergens and pathogenesis of asthma. Many of the identified susceptibility genes for asthma are expressed in the airway epithelium, supporting the notion that events at the airway epithelial surface are critical for the development of the disease. However, the exact mechanisms by which the expression of epithelial susceptibility genes translates into a functionally altered response to environmental risk factors of asthma are still unknown. Interactions between genetic factors and epigenetic regulatory mechanisms may be crucial for asthma susceptibility. Understanding these mechanisms may lead to identification of novel targets for asthma intervention by targeting the airway epithelium. Moreover, exciting new insights have come from recent studies using single‐cell RNA sequencing (scRNA‐Seq) to study the airway epithelium in asthma. This review focuses on the role of airway epithelial barrier function in the susceptibility to develop asthma and novel insights in the modulation of epithelial cell dysfunction in asthma.
Collapse
Affiliation(s)
- Irene H. Heijink
- Department of Pathology & Medical Biology GRIAC Research Institute University Medical Center Groningen University of Groningen Groningen The Netherlands
- Department of Pulmonology University Medical Center Groningen University of Groningen Groningen The Netherlands
| | - Virinchi N. S. Kuchibhotla
- Department of Pathology & Medical Biology GRIAC Research Institute University Medical Center Groningen University of Groningen Groningen The Netherlands
- School of Biomedical Sciences and Pharmacy University of Newcastle Callaghan NSW Australia
| | - Mirjam P. Roffel
- Department of Pathology & Medical Biology GRIAC Research Institute University Medical Center Groningen University of Groningen Groningen The Netherlands
- Department of Respiratory Medicine Laboratory for Translational Research in Obstructive Pulmonary Diseases Ghent University Hospital Ghent University Ghent Belgium
| | - Tania Maes
- Department of Respiratory Medicine Laboratory for Translational Research in Obstructive Pulmonary Diseases Ghent University Hospital Ghent University Ghent Belgium
| | - Darryl A. Knight
- School of Biomedical Sciences and Pharmacy University of Newcastle Callaghan NSW Australia
- UBC Providence Health Care Research Institute Vancouver BC Canada
- Department of Anesthesiology, Pharmacology and Therapeutics University of British Columbia Vancouver BC Canada
| | - Ian Sayers
- Division of Respiratory Medicine National Institute for Health Research Nottingham Biomedical Research Centre University of Nottingham Biodiscovery Institute University of Nottingham Nottingham UK
| | - Martijn C. Nawijn
- Department of Pathology & Medical Biology GRIAC Research Institute University Medical Center Groningen University of Groningen Groningen The Netherlands
| |
Collapse
|
25
|
Forsberg A, Huoman J, Söderholm S, Bhai Mehta R, Nilsson L, Abrahamsson TR, Ernerudh J, Gustafsson M, Jenmalm MC. Pre- and postnatal Lactobacillus reuteri treatment alters DNA methylation of infant T helper cells. Pediatr Allergy Immunol 2020; 31:544-553. [PMID: 32150651 DOI: 10.1111/pai.13240] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/20/2019] [Accepted: 01/27/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Perinatal childhood exposures, including probiotic supplementation, may affect epigenetic modifications and impact on immune maturation and allergy development. The aim of this study was to assess the effects of pre- and postnatal Lactobacillus reuteri supplementation on DNA methylation in relation to immune maturation and allergy development. METHODS DNA methylation patterns were investigated for allergy-related T helper subsets using a locus-specific method and at a genome-wide scale using the Illumina 450K array. From a randomised, double-blind, placebo-controlled allergy prevention trial with pre- and postnatal probiotic supplementation, CD4+ T helper cells were obtained at birth (from cord blood), and 12 and 24 months of age (total (placebo/probiotics); locus-specific method: CB = 32 (17/15), 12 months = 24 (9/15), 24 months = 35 (15/20); Illumina: CB = 19 (10/9), 12 months = 10 (6/4), 24 months = 19(11/8)). RESULTS Comparing probiotics to placebo, the greatest genome-wide differential DNA methylation was observed at birth, where the majority of sites were hypomethylated, indicating transcriptional accessibility in the probiotic group. Bioinformatic analyses, including network analyses, revealed a module containing 91 genes, enriched for immune-related pathways such as chemotaxis, PI3K-Akt, MAPK and TGF-β signalling. A majority of the module genes were associated with atopic manifestations (OR = 1.43, P = 2.4 × 10-6 ), and a classifier built on this model could predict allergy development (AUC = 0.78, P = 3.0 × 10e-3 ). Pathways such as IFN-γ signalling and T-cell activation were more hypermethylated at birth compared with later in life in both intervention groups over time, in line with DNA methylation patterns in the IFNG locus obtained by the locus-specific methodology. CONCLUSION Maternal L. reuteri supplementation during pregnancy alters DNA methylation patterns in CD4+ T cells towards enhanced immune activation at birth, which may affect immune maturation and allergy development.
Collapse
Affiliation(s)
- Anna Forsberg
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Johanna Huoman
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Simon Söderholm
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Wallenberg Centre for Molecular Medicine, Linköping University, Linköping, Sweden
| | - Ratnesh Bhai Mehta
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Lennart Nilsson
- Allergy Center, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Thomas R Abrahamsson
- Crown Princess Victoria's Child and Youth Hospital, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Jan Ernerudh
- Department of Clinical Immunology and Transfusion Medicine, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Mika Gustafsson
- Department of Physics, Chemistry and Biology, Bioinformatics, Linköping University, Linköping, Sweden
| | - Maria C Jenmalm
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
26
|
Thom CS, Voight BF. Genetic colocalization atlas points to common regulatory sites and genes for hematopoietic traits and hematopoietic contributions to disease phenotypes. BMC Med Genomics 2020; 13:89. [PMID: 32600345 PMCID: PMC7325014 DOI: 10.1186/s12920-020-00742-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/17/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Genetic associations link hematopoietic traits and disease end-points, but most causal variants and genes underlying these relationships are unknown. Here, we used genetic colocalization to nominate loci and genes related to shared genetic signal for hematopoietic, cardiovascular, autoimmune, neuropsychiatric, and cancer phenotypes. METHODS Our aim was to identify colocalization sites for human traits among established genome-wide significant loci. Using genome-wide association study (GWAS) summary statistics, we determined loci where multiple traits colocalized at a false discovery rate < 5%. We then identified quantitative trait loci among colocalization sites to highlight related genes. In addition, we used Mendelian randomization analysis to further investigate certain trait relationships genome-wide. RESULTS Our findings recapitulated developmental hematopoietic lineage relationships, identified loci that linked traits with causal genetic relationships, and revealed novel trait associations. Out of 2706 loci with genome-wide significant signal for at least 1 blood trait, we identified 1779 unique sites (66%) with shared genetic signal for 2+ hematologic traits. We could assign some sites to specific developmental cell types during hematopoiesis based on affected traits, including those likely to impact hematopoietic progenitor cells and/or megakaryocyte-erythroid progenitor cells. Through an expanded analysis of 70 human traits, we defined 2+ colocalizing traits at 2123 loci from an analysis of 9852 sites (22%) containing genome-wide significant signal for at least 1 GWAS trait. In addition to variants and genes underlying shared genetic signal between blood traits and disease phenotypes that had been previously related through Mendelian randomization studies, we defined loci and related genes underlying shared signal between eosinophil percentage and eczema. We also identified colocalizing signals in a number of clinically relevant coding mutations, including sites linking PTPN22 with Crohn's disease, NIPA with coronary artery disease and platelet trait variation, and the hemochromatosis gene HFE with altered lipid levels. Finally, we anticipate potential off-target effects on blood traits related novel therapeutic targets, including TRAIL. CONCLUSIONS Our findings provide a road map for gene validation experiments and novel therapeutics related to hematopoietic development, and offer a rationale for pleiotropic interactions between hematopoietic loci and disease end-points.
Collapse
Affiliation(s)
- Christopher S Thom
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, USA
- Institute of Translational Medicine and Therapeutics, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, USA
| | - Benjamin F Voight
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, USA.
- Department of Genetics, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, USA.
- Institute of Translational Medicine and Therapeutics, University of Pennsylvania - Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
27
|
Genome‑wide analysis of DNA methylation and gene expression changes in an ovalbumin‑induced asthma mouse model. Mol Med Rep 2020; 22:1709-1716. [PMID: 32705270 PMCID: PMC7411290 DOI: 10.3892/mmr.2020.11245] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 02/04/2020] [Indexed: 12/12/2022] Open
Abstract
The aim of the present study was to establish an integrated network of DNA methylation and RNA expression in an ovalbumin (OVA)-induced asthma model, and to investigate the epigenetically-regulated genes involved in asthma development. Genome-wide CpG-DNA methylation profiling was conducted through the use of a methylated DNA immunoprecipitation microarray and RNA sequencing was performed using three lung samples from mice with OVA-induced asthma. A total of 35,401 differentially methylated regions (DMRs) were identified between mice with OVA-induced asthma and control mice. Of these, 3,060 were located in promoter regions and 370 of the genes containing these DMRs demonstrated an inverse correlation between methylation and gene expression. Kyoto Encyclopedia of Genes and Genomes pathway analysis identified that 368 genes were upregulated or downregulated in OVA-induced asthma samples, including genes involved in ‘chemokine signalling pathway’, ‘focal adhesion’, ‘leukocyte transendothelial migration’ and ‘vascular smooth muscle contraction signaling’ pathways. Integrated network analysis identified four hub genes, consisting of three upregulated genes [forkhead box O1 (FOXO1), SP1 transcription factor (SP1) and amyloid β precursor protein (APP)], and one downregulated gene [RUNX family transcription factor 1 (RUNX1)], all of which demonstrated an association between DNA methylation and gene expression. These genes were highly interconnected nodes in the Ingenuity Pathway Analysis module and were functionally significant. A total of four interconnected hub genes, FOXO1, RUNX1, SP1 and APP, were identified from the integrated DNA methylation and gene expression networks involved in asthma development. These results suggested that modulating these four genes could effectively control the development of asthma.
Collapse
|
28
|
Sugita K, Soyka MB, Wawrzyniak P, Rinaldi AO, Mitamura Y, Akdis M, Akdis CA. Outside-in hypothesis revisited: The role of microbial, epithelial, and immune interactions. Ann Allergy Asthma Immunol 2020; 125:517-527. [PMID: 32454094 DOI: 10.1016/j.anai.2020.05.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/14/2020] [Accepted: 05/17/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Our understanding of the origin of allergic diseases has increased in recent years, highlighting the importance of microbial dysbiosis and epithelial barrier dysfunction in affected tissues. Exploring the microbial-epithelial-immune crosstalk underlying the mechanisms of allergic diseases will allow the development of novel prevention and treatment strategies for allergic diseases. DATA SOURCES This review summarizes the recent advances in microbial, epithelial, and immune interactions in atopic dermatitis, allergic rhinitis, chronic rhinosinusitis, and asthma. STUDY SELECTIONS We performed a literature search, identifying relevant recent primary articles and review articles. RESULTS Dynamic crosstalk between the environmental factors and microbial, epithelial, and immune cells in the development of atopic dermatitis, allergic rhinitis, chronic rhinosinusitis, and asthma underlies the pathogenesis of these diseases. There is substantial evidence in the literature suggesting that environmental factors directly affect barrier function of the epithelium. In addition, T-helper 2 (TH2) cells, type 2 innate lymphoid cells, and their cytokine interleukin 13 (IL-13) damage skin and lung barriers. The effects of environmental factors may at least in part be mediated by epigenetic mechanisms. Histone deacetylase activation by type 2 immune response has a major effect on leaky barriers and blocking of histone deacetylase activity corrects the defective barrier in human air-liquid interface cultures and mouse models of allergic asthma with rhinitis. We also present and discuss a novel device to detect and monitor skin barrier dysfunction, which provides an opportunity to rapidly and robustly assess disease severity. CONCLUSION A complex interplay between environmental factors, epithelium, and the immune system is involved in the development of systemic allergic diseases.
Collapse
Affiliation(s)
- Kazunari Sugita
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland; Division of Dermatology, Department of Medicine of Sensory and Motor Organs, Faculty of Medicine, Tottori University, Yonago, Japan.
| | - Michael B Soyka
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Department of Otorhinolaryngology, Head and Neck Surgery, University and University Hospital of Zurich, Zurich, Switzerland
| | - Paulina Wawrzyniak
- Division of Clinical Chemistry and Biochemistry and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Arturo O Rinaldi
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| |
Collapse
|
29
|
Frey A, Lunding LP, Ehlers JC, Weckmann M, Zissler UM, Wegmann M. More Than Just a Barrier: The Immune Functions of the Airway Epithelium in Asthma Pathogenesis. Front Immunol 2020; 11:761. [PMID: 32411147 PMCID: PMC7198799 DOI: 10.3389/fimmu.2020.00761] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/03/2020] [Indexed: 12/11/2022] Open
Abstract
Allergic bronchial asthma is a chronic disease of the airways that is characterized by symptoms like respiratory distress, chest tightness, wheezing, productive cough, and acute episodes of broncho-obstruction. This symptom-complex arises on the basis of chronic allergic inflammation of the airway wall. Consequently, the airway epithelium is central to the pathogenesis of this disease, because its multiple abilities directly have an impact on the inflammatory response and thus the formation of the disease. In turn, its structure and functions are markedly impaired by the inflammation. Hence, the airway epithelium represents a sealed, self-cleaning barrier, that prohibits penetration of inhaled allergens, pathogens, and other noxious agents into the body. This barrier is covered with mucus that further contains antimicrobial peptides and antibodies that are either produced or specifically transported by the airway epithelium in order to trap these particles and to remove them from the body by a process called mucociliary clearance. Once this first line of defense of the lung is overcome, airway epithelial cells are the first cells to get in contact with pathogens, to be damaged or infected. Therefore, these cells release a plethora of chemokines and cytokines that not only induce an acute inflammatory reaction but also have an impact on the alignment of the following immune reaction. In case of asthma, all these functions are impaired by the already existing allergic immune response that per se weakens the barrier integrity and self-cleaning abilities of the airway epithelium making it more vulnerable to penetration of allergens as well as of infection by bacteria and viruses. Recent studies indicate that the history of allergy- and pathogen-derived insults can leave some kind of memory in these cells that can be described as imprinting or trained immunity. Thus, the airway epithelium is in the center of processes that lead to formation, progression and acute exacerbation of asthma.
Collapse
Affiliation(s)
- Andreas Frey
- Division of Mucosal Immunology and Diagnostics, Research Center Borstel, Borstel, Germany.,Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany
| | - Lars P Lunding
- Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany.,Division of Asthma Exacerbation & Regulation, Research Center Borstel, Borstel, Germany
| | - Johanna C Ehlers
- Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany.,Division of Experimental Pneumology, Research Center Borstel, Borstel, Germany
| | - Markus Weckmann
- Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany.,Department of Pediatric Pulmonology and Allergology, University Children's Hospital, Lübeck, Germany
| | - Ulrich M Zissler
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany.,Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| | - Michael Wegmann
- Airway Research Center North, German Center for Lung Research (DZL), Borstel, Germany.,Division of Asthma Exacerbation & Regulation, Research Center Borstel, Borstel, Germany
| |
Collapse
|
30
|
Fang L, Sun Q, Roth M. Immunologic and Non-Immunologic Mechanisms Leading to Airway Remodeling in Asthma. Int J Mol Sci 2020; 21:ijms21030757. [PMID: 31979396 PMCID: PMC7037330 DOI: 10.3390/ijms21030757] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 01/18/2020] [Accepted: 01/20/2020] [Indexed: 02/07/2023] Open
Abstract
Asthma increases worldwide without any definite reason and patient numbers double every 10 years. Drugs used for asthma therapy relax the muscles and reduce inflammation, but none of them inhibited airway wall remodeling in clinical studies. Airway wall remodeling can either be induced through pro-inflammatory cytokines released by immune cells, or direct binding of IgE to smooth muscle cells, or non-immunological stimuli. Increasing evidence suggests that airway wall remodeling is initiated early in life by epigenetic events that lead to cell type specific pathologies, and modulate the interaction between epithelial and sub-epithelial cells. Animal models are only available for remodeling in allergic asthma, but none for non-allergic asthma. In human asthma, the mechanisms leading to airway wall remodeling are not well understood. In order to improve the understanding of this asthma pathology, the definition of “remodeling” needs to be better specified as it summarizes a wide range of tissue structural changes. Second, it needs to be assessed if specific remodeling patterns occur in specific asthma pheno- or endo-types. Third, the interaction of the immune cells with tissue forming cells needs to be assessed in both directions; e.g., do immune cells always stimulate tissue cells or are inflamed tissue cells calling immune cells to the rescue? This review aims to provide an overview on immunologic and non-immunologic mechanisms controlling airway wall remodeling in asthma.
Collapse
Affiliation(s)
- Lei Fang
- Pulmonary Cell Research & Pneumology, University Hospital & University of Basel, Petersgraben 4, CH-4031 Basel, Switzerland;
| | - Qinzhu Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China;
| | - Michael Roth
- Pulmonary Cell Research & Pneumology, University Hospital & University of Basel, Petersgraben 4, CH-4031 Basel, Switzerland;
- Correspondence: ; Tel.: +41-61-265-2337
| |
Collapse
|
31
|
Bousquet J, Akdis CA, Grattan C, Eigenmann PA, Hoffmann‐Sommergruber K, Agache I, Jutel M. Highlights and recent developments in airway diseases in EAACI journals (2018). Allergy 2019; 74:2329-2341. [PMID: 31573676 DOI: 10.1111/all.14068] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 09/24/2019] [Accepted: 09/27/2019] [Indexed: 12/12/2022]
Abstract
The European Academy of Allergy and Clinical Immunology (EAACI) supports three journals: Allergy, Pediatric Allergy and Immunology, and Clinical and Translational Allergy. EAACI's major goals include supporting the promotion of health, in which the prevention of allergy and asthma plays a critical role, and disseminating the knowledge of allergic disease to all stakeholders. In 2018, the remarkable progress in the identification of basic mechanisms of allergic and respiratory diseases as well as the translation of these findings into clinical practice were observed. Last year's highlights include publication of EAACI guidelines for allergen immunotherapy, many EAACI Position Papers covering important aspects for the specialty, better understanding of molecular and cellular mechanisms, identification of biomarkers for disease prediction and progress monitoring, novel prevention and intervention studies, elucidation of mechanisms of multimorbidities, introduction of new drugs to the clinics, recently completed phase three clinical studies, and publication of a large number of allergen immunotherapy studies and meta-analyses.
Collapse
Affiliation(s)
- Jean Bousquet
- Fondation partenariale FMC VIA‐LR MACVIA‐France Montpellier France
- INSERM U 1168 VIMA: Ageing and Chronic Diseases Epidemiological and Public Health Approaches Villejuif France
- UMR‐S 1168 Université Versailles St‐Quentin‐en‐Yvelines Montigny le Bretonneux France
- EUFOREA Brussels Belgium
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
- Christine Kühne‐Center for Allergy Research and Education Davos Switzerland
| | - Clive Grattan
- St John's Institute of Dermatology Guy's Hospital London UK
| | | | | | - Ioana Agache
- Department of Allergy and Clinical Immunology Faculty of Medicine Transylvania University Brasov Brasov Romania
| | - Marek Jutel
- Department of Clinical Immunology ALL‐MED Medical Research Institute Wroclaw Medical University Wrocław Poland
| |
Collapse
|
32
|
Larenas-Linnemann D, Romero-Tapia SJ, Virgen C, Mallol J, Baeza Bacab MA, García-Marcos L. Risk factors for wheezing in primary health care settings in the tropics. Ann Allergy Asthma Immunol 2019; 124:179-184.e1. [PMID: 31734332 DOI: 10.1016/j.anai.2019.11.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 10/29/2019] [Accepted: 11/06/2019] [Indexed: 10/25/2022]
Abstract
BACKGROUND The International Study of Wheezing in Infants (EISL) is a cross-sectional, population-based study, based on ISAAC (http://www.isaac.auckland.ac.nz). It uses a validated questionnaire on early wheezing and risk/protective factors. OBJECTIVE To apply the EISL questionnaire regarding wheezing events in 0- to 12-month-old infants with or without atopic background searching for risk factors in the tropics. METHODS The population was toddlers coming in for a checkup or 12-months' vaccination in primary health care clinics of a tropical city. Apart from child factors (eg, daycare attendance), we evaluated home factors (eg, air conditioning, bathroom, carpet, >6 persons, pollution) and mothers' factors (eg, education level, employment, cellphone). Data analysis was descriptive and case-control, with as cases atopic (AW) or non-atopic (NAW) wheezing children vs healthy controls. Wheezing-associated factors were evaluated using multivariate analysis, adjusted for the relation of AW/NAW with factors that were significant in prior univariate analysis. RESULTS The study included 999 toddlers. Any wheeze: 31.3%, recurrent wheeze (≥3 episodes): 12.1%. Major risk factors for AW (OR; 95%CI) included smoking (11.39; 2.36-54.99), common cold before 3 months of life (3.72; 2.59-5.36), mold (3.48; 2.28-5.30), kitchen indoors (2.40; 1.27-4.54), and pets (1.69; 1.09-2.62); breastfeeding was almost protective. For NAW, common cold and pets were risk factors, but cesarean section (0.44; 0.23-0.82), more than 1 sibling (0.33; 0.18-0.61), and breastfeeding for longer than 3 months (0.50; 0.28-0.91) were protective. CONCLUSION Wheezing is a health care burden. We found potential new risk factors for AW, some possibly unique for tropical climates. We suggest testing several hypotheses: could early AW be reduced in the tropics by attacking mold growth? Enhancing cooking place ventilation? Keeping pets outside? Or by postponing daycare attendance until after 4 months of age and avoiding (passive) smoking during pregnancy?
Collapse
Affiliation(s)
| | | | - Cesar Virgen
- Pediatric Private practice, Villahermosa, Mexico
| | - Javier Mallol
- Department of Pediatric Respiratory Medicine, Faculty of Medical Sciences, Hospital CRS El Pino, University of Santiago de Chile (USACH)
| | | | - Luis García-Marcos
- Research unit, Department of Paediatrics at the "Virgen de la Arrixaca" University Children's Hospital, El Palmar, Spain
| |
Collapse
|
33
|
Qi C, Xu CJ, Koppelman GH. The role of epigenetics in the development of childhood asthma. Expert Rev Clin Immunol 2019; 15:1287-1302. [PMID: 31674254 DOI: 10.1080/1744666x.2020.1686977] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: The development of childhood asthma is caused by a combination of genetic factors and environmental exposures. Epigenetics describes mechanisms of (heritable) regulation of gene expression that occur without changes in DNA sequence. Epigenetics is strongly related to aging, is cell-type specific, and includes DNA methylation, noncoding RNAs, and histone modifications.Areas covered: This review summarizes recent epigenetic studies of childhood asthma in humans, which mostly involve studies of DNA methylation published in the recent five years. Environmental exposures, in particular cigarette smoking, have significant impact on epigenetic changes, but few of these epigenetic signals are also associated with asthma. Several asthma-associated genetic variants relate to DNA methylation. Epigenetic signals can be better understood by studying their correlation with gene expression, which revealed higher presence and activation of blood eosinophils in asthma. Strong associations of nasal methylation signatures and atopic asthma were identified, which were replicable across different populations.Expert commentary: Epigenetic markers have been strongly associated with asthma, and might serve as biomarker of asthma. The causal and longitudinal relationships between epigenetics and disease, and between environmental exposures and epigenetic changes need to be further investigated. Efforts should be made to understand cell-type-specific epigenetic mechanisms in asthma.
Collapse
Affiliation(s)
- Cancan Qi
- Dept. of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Cheng-Jian Xu
- Dept. of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Gastroenterology, Hepatology and Endocrinology, CiiM, Centre for individualised infection medicine, A joint venture between Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany.,TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Gerard H Koppelman
- Dept. of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,GRIAC Research Institute, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
34
|
Edris A, den Dekker HT, Melén E, Lahousse L. Epigenome-wide association studies in asthma: A systematic review. Clin Exp Allergy 2019; 49:953-968. [PMID: 31009112 DOI: 10.1111/cea.13403] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/29/2019] [Accepted: 04/10/2019] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Asthma is a common chronic respiratory airway disease influenced by environmental factors and possibly their interaction with the human genome causing epigenetic changes. Epigenome-wide association studies (EWAS) have mainly investigated DNA methylation and its association with disease or traits, exposure factors or gene expression. This systematic review aimed to identify all EWAS assessing differentially methylated sites associated with asthma in humans. DESIGN Structured systematic literature search following PRISMA guidelines, Newcastle-Ottawa Scale (NOS) for cohort studies was used for bias assessment. DATA SOURCES We searched PubMed and Embase databases from 2005 to 2019. ELIGIBILITY CRITERIA Epigenome-wide association studies testing association between differential methylation and asthma in humans. RESULTS Overall, we identified 16 EWAS studies complying with our search criteria. Twelve studies were conducted on children, and 10 were conducted on sample sizes <150 subjects. Four hundred and nineteen CpGs were reported in children studies after correction for multiple testing. In the adult studies, thousands of differentially methylated sites were identified. Differential methylation in inflammatory-related genes correlated with higher levels of gene expressions of inflammatory modulators in asthma. Differentially methylated genes associated with asthma included SMAD3, SERPINC1, PROK1, IL13, RUNX3 and TIGIT. Forty-one CpGs were replicated at least once in blood samples, and 28 CpGs were replicated in nasal samples. CONCLUSION Although many differentially methylated CpGs in genes known to be involved in asthma have been identified in EWAS to date, we conclude that further studies of larger sample sizes and analyses of differential methylation between different phenotypes are needed in order to comprehensively evaluate the role of epigenetic factors in the pathophysiology and heterogeneity of asthma, and the potential clinical utility to predict or classify patients with asthma.
Collapse
Affiliation(s)
- Ahmed Edris
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Herman T den Dekker
- The Generation R Study Group, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands.,Department of Epidemiology, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands.,Division of Respiratory Medicine and Allergology, Department of Pediatrics, Erasmus MC, University Medical Centre, Rotterdam, The Netherlands
| | - Erik Melén
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Sachs' Children's Hospital, Södersjukhuset, Stockholm, Sweden.,Centre for Occupational and Environmental Medicine, Stockholm County Council, Stockholm, Sweden
| | - Lies Lahousse
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
35
|
Hernandez-Pacheco N, Pino-Yanes M, Flores C. Genomic Predictors of Asthma Phenotypes and Treatment Response. Front Pediatr 2019; 7:6. [PMID: 30805318 PMCID: PMC6370703 DOI: 10.3389/fped.2019.00006] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 01/10/2019] [Indexed: 12/11/2022] Open
Abstract
Asthma is a complex respiratory disease considered as the most common chronic condition in children. A large genetic contribution to asthma susceptibility is predicted by the clustering of asthma and allergy symptoms among relatives and the large disease heritability estimated from twin studies, ranging from 55 to 90%. Genetic basis of asthma has been extensively investigated in the past 40 years using linkage analysis and candidate-gene association studies. However, the development of dense arrays for polymorphism genotyping has enabled the transition toward genome-wide association studies (GWAS), which have led the discovery of several unanticipated asthma genes in the last 11 years. Despite this, currently known risk variants identified using many thousand samples from distinct ethnicities only explain a small proportion of asthma heritability. This review examines the main findings of the last 2 years in genomic studies of asthma using GWAS and admixture mapping studies, as well as the direction of studies fostering integrative perspectives involving omics data. Additionally, we discuss the need for assessing the whole spectrum of genetic variation in association studies of asthma susceptibility, severity, and treatment response in order to further improve our knowledge of asthma genes and predictive biomarkers. Leveraging the individual's genetic information will allow a better understanding of asthma pathogenesis and will facilitate the transition toward a more precise diagnosis and treatment.
Collapse
Affiliation(s)
- Natalia Hernandez-Pacheco
- Research Unit, Hospital Universitario N.S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain.,Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | - Maria Pino-Yanes
- Research Unit, Hospital Universitario N.S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain.,Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna, Santa Cruz de Tenerife, Spain.,CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Flores
- Research Unit, Hospital Universitario N.S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain.,CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
| |
Collapse
|
36
|
Forno E, Celedón JC. Epigenomics and Transcriptomics in the Prediction and Diagnosis of Childhood Asthma: Are We There Yet? Front Pediatr 2019; 7:115. [PMID: 31001502 PMCID: PMC6454089 DOI: 10.3389/fped.2019.00115] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/11/2019] [Indexed: 01/15/2023] Open
Abstract
Asthma is the most common non-communicable chronic disease of childhood. Despite its high prevalence, to date we lack methods that are both efficient and accurate in diagnosing asthma. Most traditional approaches have been based on garnering clinical evidence, such as risk factors and exposures. Given the high heritability of asthma, more recent approaches have looked at genetic polymorphisms as potential "risk factors." However, genetic variants explain only a small proportion of asthma risk, and have been less than optimal at predicting risk for individual subjects. Epigenomic studies offer significant advantages over previous approaches. Epigenetic regulation is highly tissue-specific, and can induce both short- and long-term changes in gene expression. Such changes can start in utero, can vary throughout the life span, and in some instances can be passed on from one generation to another. Most importantly, the epigenome can be modified by environmental factors and exposures, and thus epigenetic and transcriptomic profiling may yield the most accurate risk estimates for a given patient by incorporating environmental (and treatment) effects throughout the lifespan. Here we will review the most recent advances in the use of epigenetic and transcriptomic analysis for the early diagnosis of asthma and atopy, as well as challenges and future directions in the field as it moves forward. We will particularly focus on DNA methylation, the most studied mechanism of epigenetic regulation.
Collapse
Affiliation(s)
- Erick Forno
- Division of Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, United States.,Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Juan C Celedón
- Division of Pulmonary Medicine, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, United States.,Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
37
|
Lund RJ, Osmala M, Malonzo M, Lukkarinen M, Leino A, Salmi J, Vuorikoski S, Turunen R, Vuorinen T, Akdis C, Lähdesmäki H, Lahesmaa R, Jartti T. Atopic asthma after rhinovirus-induced wheezing is associated with DNA methylation change in the SMAD3 gene promoter. Allergy 2018; 73:1735-1740. [PMID: 29729188 PMCID: PMC6055882 DOI: 10.1111/all.13473] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2018] [Indexed: 12/22/2022]
Abstract
Children with rhinovirus‐induced severe early wheezing have an increased risk of developing asthma later in life. The exact molecular mechanisms for this association are still mostly unknown. To identify potential changes in the transcriptional and epigenetic regulation in rhinovirus‐associated atopic or nonatopic asthma, we analyzed a cohort of 5‐year‐old children (n = 45) according to the virus etiology of the first severe wheezing episode at the mean age of 13 months and to 5‐year asthma outcome. The development of atopic asthma in children with early rhinovirus‐induced wheezing was associated with DNA methylation changes at several genomic sites in chromosomal regions previously linked to asthma. The strongest changes in atopic asthma were detected in the promoter region of SMAD3 gene at chr 15q22.33 and introns of DDO/METTL24 genes at 6q21. These changes were validated to be present also at the average age of 8 years.
Collapse
Affiliation(s)
- R. J. Lund
- Turku Centre for Biotechnology; University of Turku and Åbo Akademi University; Turku Finland
| | - M. Osmala
- Department of Information and Computer Science; Aalto University; Helsinki Finland
| | - M. Malonzo
- Department of Information and Computer Science; Aalto University; Helsinki Finland
| | - M. Lukkarinen
- Department of Paediatrics and Adolescent Medicine; Turku University Hospital; University of Turku; Turku Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine; University of Turku; Turku Finland
| | - A. Leino
- Department of Paediatrics and Adolescent Medicine; Turku University Hospital; University of Turku; Turku Finland
| | - J. Salmi
- Turku Centre for Biotechnology; University of Turku and Åbo Akademi University; Turku Finland
| | - S. Vuorikoski
- Turku Centre for Biotechnology; University of Turku and Åbo Akademi University; Turku Finland
| | - R. Turunen
- Department of Paediatrics and Adolescent Medicine; Turku University Hospital; University of Turku; Turku Finland
- Department of Virology; University of Turku; Turku Finland
| | - T. Vuorinen
- Department of Virology; University of Turku; Turku Finland
- Department of Clinical Virology; Turku University Hospital; Turku Finland
| | - C. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF); Christine Kühne-Center for Allergy Research and Education (CK-CARE); University of Zürich; Davos Switzerland
| | - H. Lähdesmäki
- Department of Information and Computer Science; Aalto University; Helsinki Finland
| | - R. Lahesmaa
- Turku Centre for Biotechnology; University of Turku and Åbo Akademi University; Turku Finland
| | - T. Jartti
- Department of Paediatrics and Adolescent Medicine; Turku University Hospital; University of Turku; Turku Finland
| |
Collapse
|