201
|
Silkoff PE, Flavin S, Gordon R, Loza MJ, Sterk PJ, Lutter R, Diamant Z, Turner RB, Lipworth BJ, Proud D, Singh D, Eich A, Backer V, Gern JE, Herzmann C, Halperin SA, Mensinga TT, Del Vecchio AM, Branigan P, San Mateo L, Baribaud F, Barnathan ES, Johnston SL. Toll-like receptor 3 blockade in rhinovirus-induced experimental asthma exacerbations: A randomized controlled study. J Allergy Clin Immunol 2017; 141:1220-1230. [PMID: 28734844 DOI: 10.1016/j.jaci.2017.06.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 06/04/2017] [Accepted: 06/12/2017] [Indexed: 01/21/2023]
Abstract
BACKGROUND Human rhinoviruses (HRVs) commonly precipitate asthma exacerbations. Toll-like receptor 3, an innate pattern recognition receptor, is triggered by HRV, driving inflammation that can worsen asthma. OBJECTIVE We sought to evaluate an inhibitory mAb to Toll-like receptor 3, CNTO3157, on experimental HRV-16 inoculation in healthy subjects and asthmatic patients. METHODS In this double-blind, multicenter, randomized, parallel-group study in North America and Europe, healthy subjects and patients with mild-to-moderate stable asthma received single or multiple doses of CNTO3157 or placebo, respectively, and were then inoculated with HRV-16 within 72 hours. All subjects were monitored for respiratory symptoms, lung function, and nasal viral load. The primary end point was maximal decrease in FEV1 during 10 days after inoculation. RESULTS In asthmatic patients (n = 63) CNTO3157 provided no protection against FEV1 decrease (least squares mean: CNTO3157 [n = 30] = -7.08% [SE, 8.15%]; placebo [n = 25] = -5.98% [SE, 8.56%]) or symptoms after inoculation. In healthy subjects (n = 12) CNTO3157 versus placebo significantly attenuated upper (P = .03) and lower (P = .02) airway symptom scores, with area-under-the-curve increases of 9.1 (15.1) versus 34.9 (17.6) and 13.0 (18.4) versus 50.4 (25.9) for the CNTO3157 (n = 8) and placebo (n = 4) groups, respectively, after inoculation. All of the severe and 4 of the nonserious asthma exacerbations occurred while receiving CNTO3157. CONCLUSION In summary, CNTO3157 was ineffective in attenuating the effect of HRV-16 challenge on lung function, asthma control, and symptoms in asthmatic patients but suppressed cold symptoms in healthy subjects. Other approaches, including blockade of multiple pathways or antiviral agents, need to be sought for this high unmet medical need.
Collapse
Affiliation(s)
| | - Susan Flavin
- Janssen Research & Development LLC, Spring House, Pa
| | - Robert Gordon
- Janssen Research & Development LLC, Spring House, Pa
| | - Mathew J Loza
- Janssen Research & Development LLC, Spring House, Pa
| | - Peter J Sterk
- Department of Respiratory Medicine F5-259, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Rene Lutter
- Departments of Respiratory Medicine and Experimental Immunology, K0-150, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Zuzana Diamant
- Department of Respiratory Medicine & Allergology, Institute for Clinical Science, Skane University Hospital, Lund, and QPS Netherlands, Groningen, The Netherlands
| | - Ronald B Turner
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Va
| | - Brian J Lipworth
- Scottish Centre for Respiratory Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, United Kingdom
| | - David Proud
- Department of Physiology & Pharmacology, Snyder Institute for Chronic Diseases, University of Calgary Cumming School of Medicine, Calgary, Canada
| | - Dave Singh
- Centre for Respiratory Medicine and Allergy, Medicines Evaluation Unit, University Hospital of South Manchester Foundation Trust, University of Manchester, Manchester, United Kingdom
| | - Andreas Eich
- IKF Pneumologie Frankfurt, Clinical Research Center Respiratory Diseases, Frankfurt, Germany
| | - Vibeke Backer
- Department of Respiratory Medicine, Copenhagen, Denmark
| | - James E Gern
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wis
| | | | - Scott A Halperin
- Canadian Center for Vaccinology, Dalhousie University and the IWK Health Centre, Halifax, Canada
| | | | | | | | | | | | | | - Sebastian L Johnston
- Airway Disease Infection Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
202
|
Steinke JW, Borish L. Immune Responses in Rhinovirus-Induced Asthma Exacerbations. Curr Allergy Asthma Rep 2017; 16:78. [PMID: 27796793 DOI: 10.1007/s11882-016-0661-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Acute asthma exacerbations are responsible for urgent care visits and hospitalizations; they interfere with school and work productivity, thereby driving much of the morbidity and mortality associated with asthma. Approximately 80 to 85 % of asthma exacerbations in children, adolescents, and less frequently adults are associated with viral upper respiratory tract viral infections, and rhinovirus (RV) accounts for ∼60-70 % of these virus-associated exacerbations. Evidence suggests that it is not the virus itself but the nature of the immune response to RV that drives this untoward response. In particular, evidence supports the concept that RV acts to exacerbate an ongoing allergic inflammatory response to environmental allergens present at the time of the infection. The interaction of the ongoing IgE- and T cell-mediated response to allergen superimposed on the innate and adaptive immune responses to the virus and how this leads to triggering of an asthma exacerbation is discussed.
Collapse
Affiliation(s)
- John W Steinke
- Department of Medicine, University of Virginia Health System, Charlottesville, VA, USA.,Carter Immunology Center, University of Virginia Health System, Charlottesville, VA, USA.,Asthma and Allergic Disease Center, University of Virginia Health System, Charlottesville, VA, 22908-1355, USA
| | - Larry Borish
- Department of Medicine, University of Virginia Health System, Charlottesville, VA, USA. .,Department of Microbiology, University of Virginia Health System, Charlottesville, VA, USA. .,Carter Immunology Center, University of Virginia Health System, Charlottesville, VA, USA. .,Asthma and Allergic Disease Center, University of Virginia Health System, Charlottesville, VA, 22908-1355, USA.
| |
Collapse
|
203
|
Miyauchi K. Helper T Cell Responses to Respiratory Viruses in the Lung: Development, Virus Suppression, and Pathogenesis. Viral Immunol 2017. [DOI: 10.1089/vim.2017.0018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Kosuke Miyauchi
- RIKEN Center for Integrative Medical Science, Yokohama, Japan
| |
Collapse
|
204
|
Pathogenesis of asthma: implications for precision medicine. Clin Sci (Lond) 2017; 131:1723-1735. [PMID: 28667070 DOI: 10.1042/cs20160253] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/02/2017] [Accepted: 05/08/2017] [Indexed: 01/03/2023]
Abstract
The pathogenesis of asthma is complex and multi-faceted. Asthma patients have a diverse range of underlying dominant disease processes and pathways despite apparent similarities in clinical expression. Here, we present the current understanding of asthma pathogenesis. We discuss airway inflammation (both T2HIGH and T2LOW), airway hyperresponsiveness (AHR) and airways remodelling as four key factors in asthma pathogenesis, and also outline other contributory factors such as genetics and co-morbidities. Response to current asthma therapies also varies greatly, which is probably related to the inter-patient differences in pathogenesis. Here, we also summarize how our developing understanding of detailed pathological processes potentially translates into the targeted treatment options we require for optimal asthma management in the future.
Collapse
|
205
|
Becerra-Díaz M, Wills-Karp M, Heller NM. New perspectives on the regulation of type II inflammation in asthma. F1000Res 2017; 6:1014. [PMID: 28721208 PMCID: PMC5497827 DOI: 10.12688/f1000research.11198.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/16/2017] [Indexed: 12/12/2022] Open
Abstract
Asthma is a chronic inflammatory disease of the lungs which has been thought to arise as a result of inappropriately directed T helper type-2 (Th2) immune responses of the lungs to otherwise innocuous inhaled antigens. Current asthma therapeutics are directed towards the amelioration of downstream consequences of type-2 immune responses (i.e. β-agonists) or broad-spectrum immunosuppression (i.e. corticosteroids). However, few approaches to date have been focused on the primary prevention of immune deviation. Advances in molecular phenotyping reveal heterogeneity within the asthmatic population with multiple endotypes whose varying expression depends on the interplay between numerous environmental factors and the inheritance of a broad range of susceptibility genes. The most common endotype is one described as "type-2-high" (i.e. high levels of interleukin [IL]-13, eosinophilia, and periostin). The identification of multiple endotypes has provided a potential explanation for the observations that therapies directed at typical Th2 cytokines (IL-4, IL-5, and IL-13) and their receptors have often fallen short when they were tested in a diverse group of asthmatic patients without first stratifying based on disease endotype or severity. However, despite the incorporation of endotype-dependent stratification schemes into clinical trial designs, variation in drug responses are still apparent, suggesting that additional genetic/environmental factors may be contributing to the diversity in drug efficacy. Herein, we will review recent advances in our understanding of the complex pathways involved in the initiation and regulation of type-2-mediated immune responses and their modulation by host factors (genetics, metabolic status, and the microbiome). Particular consideration will be given to how this knowledge could pave the way for further refinement of disease endotypes and/or the development of novel therapeutic strategies for the treatment of asthma .
Collapse
Affiliation(s)
- Mireya Becerra-Díaz
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Marsha Wills-Karp
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Nicola M. Heller
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
206
|
Tworek D, Smith SG, Salter BM, Baatjes AJ, Scime T, Watson R, Obminski C, Gauvreau GM, O'Byrne PM. IL-25 Receptor Expression on Airway Dendritic Cells after Allergen Challenge in Subjects with Asthma. Am J Respir Crit Care Med 2017; 193:957-64. [PMID: 26625138 DOI: 10.1164/rccm.201509-1751oc] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
RATIONALE IL-25 is an epithelial-derived cytokine, whose effects are mediated by the IL-25 receptor (IL-17RB), and that has been implicated in the pathogenesis of allergic disease and airway viral responses. Airway myeloid dendritic cells (mDCs) and plasmacytoid dendritic cells (pDCs) are professional antigen-presenting cells. pDCs may play a protective role in asthma and are key players in the innate immune response through recognition of microbial products via Toll-like receptors (TLRs). The effects of inhaled allergens on the expression of IL-17RB by mDCs and pDCs, and the effects of IL-25 on pDCs, are unknown. OBJECTIVES To evaluate allergen-induced changes in IL-17RB expression by mDCs and pDCs and to investigate the effects of IL-25 on pDCs. METHODS Patients with mild atopic asthma (n = 13) were challenged with inhaled allergen. Blood and sputum DCs were enumerated and IL-17RB expression was determined by flow cytometry before and 7 and 24 hours after allergen challenge. The effects of IL-25 on pDCs in vitro were also assessed. MEASUREMENTS AND MAIN RESULTS Inhaled allergen significantly increased mDC and pDC numbers in sputum but not in blood. The percentage of IL-17RB(+) mDCs and pDCs was significantly increased in blood and sputum 24 hours after challenge. IL-25 up-regulated TLR9 expression by pDCs and orchestrated the responses to TLR9 ligation. CONCLUSIONS IL-17RB is up-regulated on blood and sputum mDCs and pDCs after allergen inhalation. IL-25 modulates pDC function through an effect on TLR9 expression.
Collapse
Affiliation(s)
- Damian Tworek
- 1 Firestone Institute of Respiratory Health and the Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada; and.,2 Department of Internal Medicine, Asthma and Allergy, Medical University of Lodz, Lodz, Poland
| | - Steven G Smith
- 1 Firestone Institute of Respiratory Health and the Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Brittany M Salter
- 1 Firestone Institute of Respiratory Health and the Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Adrian J Baatjes
- 1 Firestone Institute of Respiratory Health and the Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Tara Scime
- 1 Firestone Institute of Respiratory Health and the Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Rick Watson
- 1 Firestone Institute of Respiratory Health and the Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Caitlin Obminski
- 1 Firestone Institute of Respiratory Health and the Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Gail M Gauvreau
- 1 Firestone Institute of Respiratory Health and the Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| | - Paul M O'Byrne
- 1 Firestone Institute of Respiratory Health and the Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada; and
| |
Collapse
|
207
|
Toussaint M, Jackson DJ, Swieboda D, Guedán A, Tsourouktsoglou TD, Ching YM, Radermecker C, Makrinioti H, Aniscenko J, Bartlett NW, Edwards MR, Solari R, Farnir F, Papayannopoulos V, Bureau F, Marichal T, Johnston SL. Host DNA released by NETosis promotes rhinovirus-induced type-2 allergic asthma exacerbation. Nat Med 2017; 23:681-691. [PMID: 28459437 PMCID: PMC5821220 DOI: 10.1038/nm.4332] [Citation(s) in RCA: 257] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 04/04/2017] [Indexed: 02/06/2023]
Abstract
Respiratory viral infections represent the most common cause of allergic asthma exacerbations. Amplification of the type-2 immune response is strongly implicated in asthma exacerbation, but how virus infection boosts type-2 responses is poorly understood. We report a significant correlation between the release of host double-stranded DNA (dsDNA) following rhinovirus infection and the exacerbation of type-2 allergic inflammation in humans. In a mouse model of allergic airway hypersensitivity, we show that rhinovirus infection triggers dsDNA release associated with the formation of neutrophil extracellular traps (NETs), known as NETosis. We further demonstrate that inhibiting NETosis by blocking neutrophil elastase or by degrading NETs with DNase protects mice from type-2 immunopathology. Furthermore, the injection of mouse genomic DNA alone is sufficient to recapitulate many features of rhinovirus-induced type-2 immune responses and asthma pathology. Thus, NETosis and its associated extracellular dsDNA contribute to the pathogenesis and may represent potential therapeutic targets of rhinovirus-induced asthma exacerbations.
Collapse
Affiliation(s)
- Marie Toussaint
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - David J Jackson
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
- Imperial College Healthcare NHS Trust, London, UK
- Guy's and St Thomas' NHS Trust, London, UK
| | - Dawid Swieboda
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Anabel Guedán
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | | | - Yee Man Ching
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Coraline Radermecker
- Laboratory of Cellular and Molecular Immunology, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), University of Liège, Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Heidi Makrinioti
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Julia Aniscenko
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Nathan W Bartlett
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Michael R Edwards
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Roberto Solari
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
| | - Frédéric Farnir
- Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
- Fundamental and Applied Research for Animals &Health, University of Liège, Liège, Belgium
| | | | - Fabrice Bureau
- Laboratory of Cellular and Molecular Immunology, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), University of Liège, Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
- WELBIO, Walloon Excellence in Life Sciences and Biotechnology, Wallonia, Belgium
| | - Thomas Marichal
- Laboratory of Cellular and Molecular Immunology, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), University of Liège, Liège, Belgium
- Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Sebastian L Johnston
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College London, London, UK
- Medical Research Council (MRC) and Asthma UK Centre in Allergic Mechanisms of Asthma, London, UK
- Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
208
|
Heymann PW, Nguyen HT, Steinke JW, Turner RB, Woodfolk JA, Platts-Mills TAE, Martin L, He H, Biagini Myers J, Lindsey M, Sivaprasad U, Medvedovic M, Mahi N, Carper H, Murphy DD, Patrie J, Khurana Hershey GK. Rhinovirus infection results in stronger and more persistent genomic dysregulation: Evidence for altered innate immune response in asthmatics at baseline, early in infection, and during convalescence. PLoS One 2017; 12:e0178096. [PMID: 28552993 PMCID: PMC5446117 DOI: 10.1371/journal.pone.0178096] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 05/07/2017] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Rhinovirus (HRV) is associated with the large majority of virus-induced asthma exacerbations in children and young adults, but the mechanisms remain poorly defined. METHODS Asthmatics and non-asthmatic controls were inoculated with HRV-A16, and nasal epithelial samples were obtained 7 days before, 36 hours after, and 7 days after viral inoculation. RNA was extracted and subjected to RNA-seq analysis. RESULTS At baseline, 57 genes were differentially expressed between asthmatics and controls, and the asthmatics had decreased expression of viral replication inhibitors and increased expression of genes involved in inflammation. At 36 hours (before the emergence of peak symptoms), 1329 genes were significantly altered from baseline in the asthmatics compared to 62 genes in the controls. At this time point, asthmatics lacked an increase in IL-10 signaling observed in the controls. At 7 days following HRV inoculation, 222 genes were significantly dysregulated in the asthmatics, whereas only 4 genes were dysregulated among controls. At this time point, the controls but not asthmatics demonstrated upregulation of SPINK5. CONCLUSIONS As judged by the magnitude and persistence of dysregulated genes, asthmatics have a substantially different host response to HRV-A16 infection compared with non-asthmatic controls. Gene expression differences illuminate biologically plausible mechanisms that contribute to a better understanding of the pathogenesis of HRV-induced asthma exacerbations.
Collapse
Affiliation(s)
- Peter W. Heymann
- Division of Allergy, Immunology and Respiratory Medicine, Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Internal Medicine, Asthma and Allergic Diseases Center, University of Virginia, Charlottesville, Virginia, United States of America
| | - Huyen-Tran Nguyen
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - John W. Steinke
- Department of Internal Medicine, Asthma and Allergic Diseases Center, University of Virginia, Charlottesville, Virginia, United States of America
| | - Ronald B. Turner
- Division of Infectious Diseases, Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Judith A. Woodfolk
- Department of Internal Medicine, Asthma and Allergic Diseases Center, University of Virginia, Charlottesville, Virginia, United States of America
| | - Thomas A. E. Platts-Mills
- Department of Internal Medicine, Asthma and Allergic Diseases Center, University of Virginia, Charlottesville, Virginia, United States of America
| | - Lisa Martin
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Hua He
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Jocelyn Biagini Myers
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Mark Lindsey
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Umasundari Sivaprasad
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Mario Medvedovic
- Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Naim Mahi
- Department of Environmental Health, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Holliday Carper
- Division of Allergy, Immunology and Respiratory Medicine, Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Deborah D. Murphy
- Division of Allergy, Immunology and Respiratory Medicine, Department of Pediatrics, University of Virginia, Charlottesville, Virginia, United States of America
| | - James Patrie
- Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, United States of America
| | - Gurjit K. Khurana Hershey
- Division of Asthma Research, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
| |
Collapse
|
209
|
Turkeltaub PC, Cheon J, Friedmann E, Lockey RF. The Influence of Asthma and/or Hay Fever on Pregnancy: Data from the 1995 National Survey of Family Growth. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2017; 5:1679-1690. [PMID: 28550983 DOI: 10.1016/j.jaip.2017.03.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 03/19/2017] [Accepted: 03/31/2017] [Indexed: 01/17/2023]
Abstract
BACKGROUND Asthma is associated with adverse pregnancy outcomes. At the same time there is a worldwide increase in asthma and hay fever. OBJECTIVE This study addresses whether asthma and/or hay fever adversely influence pregnancy outcomes. METHODS Data from the 1995 National Survey of Family Growth that include a history of diagnosed asthma, hay fever, and adverse pregnancy outcomes in 10,847 women representative of the US population aged 15 to 44 years were analyzed. RESULTS Women with the allergic phenotypes asthma and hay fever and hay fever only had no significant increase in adverse pregnancy outcomes (spontaneous pregnancy loss, preterm birth, infant low birth weight), whereas women with the nonatopic phenotype asthma only (without hay fever) did. The study did not evaluate endotypes. CONCLUSIONS This study provides new data that the allergic phenotypes, asthma and hay fever and hay fever only, are compatible with healthy pregnancy, whereas the nonatopic asthma phenotype, asthma only, adversely impacts pregnancy.
Collapse
Affiliation(s)
| | - Jooyoung Cheon
- Sungshin Women's University College of Nursing, Seoul, Korea
| | | | | |
Collapse
|
210
|
To KKW, Yip CCY, Yuen KY. Rhinovirus - From bench to bedside. J Formos Med Assoc 2017; 116:496-504. [PMID: 28495415 DOI: 10.1016/j.jfma.2017.04.009] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 04/07/2017] [Accepted: 04/17/2017] [Indexed: 11/16/2022] Open
Abstract
Rhinovirus has been neglected in the past because it was generally perceived as a respiratory virus only capable of causing mild common cold. Contemporary epidemiological studies using molecular assays have shown that rhinovirus is frequently detected in adult and pediatric patients with upper or lower respiratory tract infections. Severe pulmonary and extrapulmonary complications are increasingly recognized. Contrary to popular belief, some rhinoviruses can actually replicate well at 37 °C and infect the lower airway in humans. The increasing availability of multiplex PCR panels allows rapid detection of rhinovirus and provides the opportunity for timely treatment and early recognition of outbreaks. Recent advances in the understanding of host factors for viral attachment and replication, and the host immunological response in both asthmatic and non-asthmatic individuals, have provided important insights into rhinovirus infection which are crucial in the development of antiviral treatment. The identification of novel drugs has been accelerated by repurposing clinically-approved drugs. As humoral antibodies induced by past exposure and vaccine antigen of a particular serotype cannot provide full coverage for all rhinovirus serotypes, novel vaccination strategies are required for inducing protective response against all rhinoviruses.
Collapse
Affiliation(s)
- Kelvin K W To
- State Key Laboratory for Emerging Infectious Diseases, Hong Kong Special Administrative Region; Carol Yu Centre for Infection, Hong Kong Special Administrative Region; Research Centre of Infection and Immunology, Hong Kong Special Administrative Region; Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Cyril C Y Yip
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Kwok-Yung Yuen
- State Key Laboratory for Emerging Infectious Diseases, Hong Kong Special Administrative Region; Carol Yu Centre for Infection, Hong Kong Special Administrative Region; Research Centre of Infection and Immunology, Hong Kong Special Administrative Region; Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
211
|
Thiriou D, Morianos I, Xanthou G, Samitas K. Innate immunity as the orchestrator of allergic airway inflammation and resolution in asthma. Int Immunopharmacol 2017; 48:43-54. [PMID: 28463786 DOI: 10.1016/j.intimp.2017.04.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 04/15/2017] [Accepted: 04/24/2017] [Indexed: 12/31/2022]
Abstract
The respiratory system is constantly in direct contact with the environment and, has therefore, developed strong innate and adaptive immune responses to combat pathogens. Unlike adaptive immunity which is mounted later in the course of the immune response and is naive at the outset, innate immunity provides the first line of defense against microbial agents, while also promoting resolution of inflammation. In the airways, innate immune effector cells mainly consist of eosinophils, neutrophils, mast cells, basophils, macrophages/monocytes, dendritic cells and innate lymphoid cells, which attack pathogens directly or indirectly through the release of inflammatory cytokines and antimicrobial peptides, and coordinate T and B cell-mediated adaptive immunity. Airway epithelial cells are also critically involved in shaping both the innate and adaptive arms of the immune response. Chronic allergic airway inflammation and linked asthmatic disease is often considered a result of aberrant activation of type 2 T helper cells (Th2) towards innocuous environmental allergens; however, innate immune cells are increasingly recognized as key players responsible for the initiation and the perpetuation of allergic responses. Moreover, innate cells participate in immune response regulation through the release of anti-inflammatory mediators, and guide tissue repair and the maintenance of airway homeostasis. The scope of this review is to outline existing knowledge on innate immune responses involved in allergic airway inflammation, highlight current gaps in our understanding of the underlying molecular and cellular mechanisms and discuss the potential use of innate effector cells in new therapeutic avenues.
Collapse
Affiliation(s)
- Despoina Thiriou
- 2(nd) Respiratory Medicine Dept., Athens Chest Hospital "Sotiria", Athens, Greece
| | - Ioannis Morianos
- Cellular Immunology Laboratory, Division of Cell Biology, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Greece
| | - Georgina Xanthou
- Cellular Immunology Laboratory, Division of Cell Biology, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Greece
| | - Konstantinos Samitas
- Cellular Immunology Laboratory, Division of Cell Biology, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, Greece; 7(th) Respiratory Medicine Dept. and Asthma Center, Athens Chest Hospital "Sotiria", Athens, Greece.
| |
Collapse
|
212
|
Edwards MR, Saglani S, Schwarze J, Skevaki C, Smith JA, Ainsworth B, Almond M, Andreakos E, Belvisi MG, Chung KF, Cookson W, Cullinan P, Hawrylowicz C, Lommatzsch M, Jackson D, Lutter R, Marsland B, Moffatt M, Thomas M, Virchow JC, Xanthou G, Edwards J, Walker S, Johnston SL. Addressing unmet needs in understanding asthma mechanisms: From the European Asthma Research and Innovation Partnership (EARIP) Work Package (WP)2 collaborators. Eur Respir J 2017; 49:49/5/1602448. [PMID: 28461300 DOI: 10.1183/13993003.02448-2016] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 03/13/2017] [Indexed: 12/27/2022]
Abstract
Asthma is a heterogeneous, complex disease with clinical phenotypes that incorporate persistent symptoms and acute exacerbations. It affects many millions of Europeans throughout their education and working lives and puts a heavy cost on European productivity. There is a wide spectrum of disease severity and control. Therapeutic advances have been slow despite greater understanding of basic mechanisms and the lack of satisfactory preventative and disease modifying management for asthma constitutes a significant unmet clinical need. Preventing, treating and ultimately curing asthma requires co-ordinated research and innovation across Europe. The European Asthma Research and Innovation Partnership (EARIP) is an FP7-funded programme which has taken a co-ordinated and integrated approach to analysing the future of asthma research and development. This report aims to identify the mechanistic areas in which investment is required to bring about significant improvements in asthma outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Rene Lutter
- Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Benjamin Marsland
- University of Lausanne, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | | | | | | | - Georgina Xanthou
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | | | | | | | | |
Collapse
|
213
|
Maltby S, Tay HL, Yang M, Foster PS. Mouse models of severe asthma: Understanding the mechanisms of steroid resistance, tissue remodelling and disease exacerbation. Respirology 2017; 22:874-885. [PMID: 28401621 DOI: 10.1111/resp.13052] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 02/28/2017] [Accepted: 03/09/2017] [Indexed: 02/07/2023]
Abstract
Severe asthma has significant disease burden and results in high healthcare costs. While existing therapies are effective for the majority of asthma patients, treatments for individuals with severe asthma are often ineffective. Mouse models are useful to identify mechanisms underlying disease pathogenesis and for the preclinical assessment of new therapies. In fact, existing mouse models have contributed significantly to our understanding of allergic/eosinophilic phenotypes of asthma and facilitated the development of novel targeted therapies (e.g. anti-IL-5 and anti-IgE). These therapies are effective in relevant subsets of severe asthma patients. Unfortunately, non-allergic/non-eosinophilic asthma, steroid resistance and disease exacerbation remain areas of unmet clinical need. No mouse model encompasses all features of severe asthma. However, mouse models can provide insight into pathogenic pathways that are relevant to severe asthma. In this review, as examples, we highlight models relevant to understanding steroid resistance, chronic tissue remodelling and disease exacerbation. Although these models highlight the complexity of the immune pathways that may underlie severe asthma, they also provide insight into new potential therapeutic approaches.
Collapse
Affiliation(s)
- Steven Maltby
- Hunter Medical Research Institute, Priority Research Centre for Healthy Lungs, Newcastle, New South Wales, Australia.,Department of Microbiology and Immunology, School of Biomedical Sciences and Pharmacy, Faculty of Health, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Hock L Tay
- Hunter Medical Research Institute, Priority Research Centre for Healthy Lungs, Newcastle, New South Wales, Australia.,Department of Microbiology and Immunology, School of Biomedical Sciences and Pharmacy, Faculty of Health, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Ming Yang
- Hunter Medical Research Institute, Priority Research Centre for Healthy Lungs, Newcastle, New South Wales, Australia.,Department of Microbiology and Immunology, School of Biomedical Sciences and Pharmacy, Faculty of Health, The University of Newcastle, Newcastle, New South Wales, Australia
| | - Paul S Foster
- Hunter Medical Research Institute, Priority Research Centre for Healthy Lungs, Newcastle, New South Wales, Australia.,Department of Microbiology and Immunology, School of Biomedical Sciences and Pharmacy, Faculty of Health, The University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
214
|
Hansel TT, Tunstall T, Trujillo-Torralbo MB, Shamji B, Del-Rosario A, Dhariwal J, Kirk PDW, Stumpf MPH, Koopmann J, Telcian A, Aniscenko J, Gogsadze L, Bakhsoliani E, Stanciu L, Bartlett N, Edwards M, Walton R, Mallia P, Hunt TM, Hunt TL, Hunt DG, Westwick J, Edwards M, Kon OM, Jackson DJ, Johnston SL. A Comprehensive Evaluation of Nasal and Bronchial Cytokines and Chemokines Following Experimental Rhinovirus Infection in Allergic Asthma: Increased Interferons (IFN-γ and IFN-λ) and Type 2 Inflammation (IL-5 and IL-13). EBioMedicine 2017; 19:128-138. [PMID: 28373098 PMCID: PMC5440599 DOI: 10.1016/j.ebiom.2017.03.033] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/09/2017] [Accepted: 03/24/2017] [Indexed: 01/04/2023] Open
Abstract
Background Rhinovirus infection is a major cause of asthma exacerbations. Objectives We studied nasal and bronchial mucosal inflammatory responses during experimental rhinovirus-induced asthma exacerbations. Methods We used nasosorption on days 0, 2–5 and 7 and bronchosorption at baseline and day 4 to sample mucosal lining fluid to investigate airway mucosal responses to rhinovirus infection in patients with allergic asthma (n = 28) and healthy non-atopic controls (n = 11), by using a synthetic absorptive matrix and measuring levels of 34 cytokines and chemokines using a sensitive multiplex assay. Results Following rhinovirus infection asthmatics developed more upper and lower respiratory symptoms and lower peak expiratory flows compared to controls (all P < 0.05). Asthmatics also developed higher nasal lining fluid levels of an anti-viral pathway (including IFN-γ, IFN-λ/IL-29, CXCL11/ITAC, CXCL10/IP10 and IL-15) and a type 2 inflammatory pathway (IL-4, IL-5, IL-13, CCL17/TARC, CCL11/eotaxin, CCL26/eotaxin-3) (area under curve day 0–7, all P < 0.05). Nasal IL-5 and IL-13 were higher in asthmatics at day 0 (P < 0.01) and levels increased by days 3 and 4 (P < 0.01). A hierarchical correlation matrix of 24 nasal lining fluid cytokine and chemokine levels over 7 days demonstrated expression of distinct interferon-related and type 2 pathways in asthmatics. In asthmatics IFN-γ, CXCL10/IP10, CXCL11/ITAC, IL-15 and IL-5 increased in bronchial lining fluid following viral infection (all P < 0.05). Conclusions Precision sampling of mucosal lining fluid identifies robust interferon and type 2 responses in the upper and lower airways of asthmatics during an asthma exacerbation. Nasosorption and bronchosorption have potential to define asthma endotypes in stable disease and at exacerbation. Following rhinovirus infection asthmatics have increased interferons and type 2 inflammation in airway mucosal lining fluid. Nasosorption cytokines and chemokines showed distinct pathways of interferon and type 2 inflammation in asthma. Precision mucosal sampling has potential for stratifying molecular endotypes of asthma. Validation of nasosorption and bronchosorption will be required for selection of asthmatics for therapy with biologics.
Experimental human rhinovirus (HRV) infection causes more severe upper and lower respiratory tract symptoms in allergic asthmatics than in healthy controls. There is greater induction of cytokines and chemokines in nasal and bronchial mucosal lining fluid (MLF) of asthmatics: with distinct pathways of type 2 and anti-viral/regulatory inflammation. Subject to further validation, analysis of MLF may prove useful in stratification of patients with asthma, and the definition of molecular endotypes. Interpretation Nasosorption and bronchosorption are precision sampling methods with potential for widespread application in respiratory and other mucosal diseases (e.g. gastrointestinal diseases). Biomarkers identified in nasosorption and bronchosorption samples will need to be validated compared to established airway sampling methods, in a range of asthma phenotypes, and with current and novel therapies.
Collapse
Affiliation(s)
- Trevor T Hansel
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK; Imperial College Healthcare NHS Trust, UK; Imperial Clinical Respiratory Research Unit (ICRRU), UK.
| | - Tanushree Tunstall
- Imperial College Healthcare NHS Trust, UK; Imperial Clinical Respiratory Research Unit (ICRRU), UK
| | - Maria-Belen Trujillo-Torralbo
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK; Imperial College Healthcare NHS Trust, UK
| | - Betty Shamji
- Novartis Institute for Biomedical Research, Horsham, UK
| | - Ajerico Del-Rosario
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK; Imperial College Healthcare NHS Trust, UK
| | - Jaideep Dhariwal
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK; Imperial College Healthcare NHS Trust, UK
| | - Paul D W Kirk
- MRC Biostatistics Unit, Cambridge Institute of Public Health, Cambridge, UK
| | | | - Jens Koopmann
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; Medimmune, Cambridge, UK
| | - Aurica Telcian
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Julia Aniscenko
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Leila Gogsadze
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Eteri Bakhsoliani
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Luminita Stanciu
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Nathan Bartlett
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Michael Edwards
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Ross Walton
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK
| | - Patrick Mallia
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK; Imperial College Healthcare NHS Trust, UK
| | - Toby M Hunt
- Hunt Developments (UK) Ltd, Midhurst, West Sussex, UK
| | - Trevor L Hunt
- Hunt Developments (UK) Ltd, Midhurst, West Sussex, UK
| | - Duncan G Hunt
- Hunt Developments (UK) Ltd, Midhurst, West Sussex, UK
| | - John Westwick
- Novartis Institute for Biomedical Research, Horsham, UK
| | | | - Onn Min Kon
- Imperial College Healthcare NHS Trust, UK; Imperial Clinical Respiratory Research Unit (ICRRU), UK
| | - David J Jackson
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK; Guy's and St Thomas' NHS Trust
| | - Sebastian L Johnston
- Airway Disease Infection Section, National Heart and Lung Institute (NHLI), Imperial College (IC), London, UK; MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, UK; Imperial College Healthcare NHS Trust, UK
| |
Collapse
|
215
|
Petrova NV, Emelyanova AG, Gorbunov EA, Edwards MR, Walton RP, Bartlett NW, Aniscenko J, Gogsadze L, Bakhsoliani E, Khaitov MR, Johnston SL, Tarasov SA, Epstein OI. Efficacy of novel antibody-based drugs against rhinovirus infection: In vitro and in vivo results. Antiviral Res 2017; 142:185-192. [PMID: 28356234 DOI: 10.1016/j.antiviral.2017.03.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 02/21/2017] [Accepted: 03/23/2017] [Indexed: 12/21/2022]
Abstract
Rhinoviruses (RVs) cause the common cold and are associated with exacerbations of chronic inflammatory respiratory diseases, especially asthma and chronic obstructive pulmonary disease (COPD). We have assessed the antiviral drugs Anaferon for Children (AC) and Ergoferon (containing AC as one of the active pharmaceutical ingredients) in in vitro and in vivo experimental models, in order to evaluate their anti-rhinoviral and immunomodulatory potential. HeLa cells were pretreated with AC, and levels of the interferon-stimulated gene (ISG), 2'-5'-oligoadenylate synthetase 1 (OAS1-A) and viral replication were analyzed. In a mouse model of RV-induced exacerbation of allergic airway inflammation we administered Ergoferon and analyzed its effect on type I (IFN-β), type II (IFN-γ) and type III (IFN-λ) IFNs induction, cell counts in bronchoalveolar lavage (BAL), cytokine (interleukin (IL)-4; IL-6) and chemokine (CXCL10/IP-10; CXCL1/KC) levels. It was shown that AC increased OAS1-А production and significantly decreased viral replication in vitro. Increased IFNs expression together with reduced neutrophils/lymphocytes recruitment and correlated IL-4/IL-6 declination was demonstrated for Ergoferon in vivo. However, there was no effect on examined chemokines. We conclude that AC and Ergoferon possess effects against RV infection and may have potential as novel therapies against RV-induced exacerbations of asthma.
Collapse
Affiliation(s)
- Nataliia V Petrova
- Research Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia.
| | | | | | - Michael R Edwards
- National Heart and Lung Institute, Imperial College, SW7 2AZ London, United Kingdom.
| | - Ross P Walton
- National Heart and Lung Institute, Imperial College, SW7 2AZ London, United Kingdom.
| | - Nathan W Bartlett
- National Heart and Lung Institute, Imperial College, SW7 2AZ London, United Kingdom.
| | - Julia Aniscenko
- National Heart and Lung Institute, Imperial College, SW7 2AZ London, United Kingdom.
| | - Leila Gogsadze
- National Heart and Lung Institute, Imperial College, SW7 2AZ London, United Kingdom.
| | - Eteri Bakhsoliani
- National Heart and Lung Institute, Imperial College, SW7 2AZ London, United Kingdom.
| | - Musa R Khaitov
- National Research Center "Institute of Immunology" FMBA Russia, 115478 Moscow, Russia.
| | - Sebastian L Johnston
- National Heart and Lung Institute, Imperial College, SW7 2AZ London, United Kingdom.
| | | | - Oleg I Epstein
- Research Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia.
| |
Collapse
|
216
|
Zheng R, Chen FH, Gao WX, Wang D, Yang QT, Wang K, Lai YY, Deng J, Jiang LJ, Sun YQ, Shi JB. The T H2-polarizing function of atopic interleukin 17 receptor B-positive dendritic cells up-regulated by lipopolysaccharide. Ann Allergy Asthma Immunol 2017; 118:474-482.e1. [PMID: 28132739 DOI: 10.1016/j.anai.2016.12.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Revised: 12/04/2016] [Accepted: 12/15/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Recent studies suggest that epithelial cell (EC)-derived cytokines contribute to allergic airway disease exacerbation. OBJECTIVE To confirm our hypothesis that atopic dendritic cells (DCs) are activated to up-regulate the receptors of cytokines that mainly derived from ECs and enhance TH2 responses. METHODS The expressions of interleukin 17 receptor B (IL-17RB) (IL-25 receptor), membrane-bound ST2 (IL-33 receptor), thymic stromal lymphopoietin receptor (TSLPR), granulocyte-macrophage colony-stimulating factor receptor (GM-CSFR), and several functional markers on CD1c+ monocyte-derived DCs (mo-DCs) were detected by flow cytometry. Lipopolysaccharide (LPS)-activated mo-DCs were cocultured with autologous CD4+ T cells, and cytokine production by these T cells was determined by intracellular flow cytometry. RESULTS LPS activated both nonatopic and atopic mo-DCs to express a higher level of GM-CSFR but only activated atopic mo-DCs to express increased IL-17RB, which was subsequently activated by IL-25 involved with signal transducer and activator of transcription 5 phosphorylation. In addition, LPS increased the expression of the OX40 ligand (OX40L) but decreased inducible costimulator ligand on atopic CD86+ mo-DCs. More importantly, IL-25 further up-regulated OX40L on atopic CD86+ mo-DCs. After coculturing with LPS-activated mo-DCs from atopic individuals, CD4+ T cells had enhanced inflammatory responses by increased production of IL-4, IL-5, IL-13, and interferon γ (IFN-γ). In contrast, further addition of IL-25 led CD4+ T cells to produce higher level of IL-4 but lower level of IFN-γ. CONCLUSION Atopic IL-17RB+ DCs can be up-regulated by LPS and promote a TH2-type response, implying that the IL-25/IL-17RB pathway may represent a potential molecular mechanism underlying the regulation of ECs on DCs in allergic airway disease.
Collapse
Affiliation(s)
- Rui Zheng
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Feng-Hong Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wen-Xiang Gao
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dan Wang
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qin-Tai Yang
- Department of Otorhinolaryngology-Head and Neck Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Kai Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Yin-Yan Lai
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jie Deng
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Li-Jie Jiang
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yue-Qi Sun
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jian-Bo Shi
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
217
|
Beyond the Paradigm of Asthma as an Inflammatory Disease. A Summary of the 2015 Aspen Lung Conference. Ann Am Thorac Soc 2017; 13 Suppl 1:S91-4. [PMID: 27027960 DOI: 10.1513/annalsats.201510-677mg] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
218
|
Airway Epithelial Orchestration of Innate Immune Function in Response to Virus Infection. A Focus on Asthma. Ann Am Thorac Soc 2017; 13 Suppl 1:S55-63. [PMID: 27027954 DOI: 10.1513/annalsats.201507-421mg] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Asthma is a very common respiratory condition with a worldwide prevalence predicted to increase. There are significant differences in airway epithelial responses in asthma that are of particular interest during exacerbations. Preventing exacerbations is a primary aim when treating asthma because they often necessitate unscheduled healthcare visits and hospitalizations and are a significant cause of morbidity and mortality. The most common cause of asthma exacerbations is a respiratory virus infection, of which the most likely type is rhinovirus infection. This article focuses on the role played by the epithelium in orchestrating the innate immune responses to respiratory virus infection. Recent studies show impaired bronchial epithelial cell innate antiviral immune responses, as well as augmentation of a pro-Th2 response characterized by the epithelial-derived cytokines IL-25 and IL-33, crucial in maintaining the Th2 cytokine response to virus infection in asthma. A better understanding of the mechanisms of these abnormal immune responses has the potential to lead to the development of novel therapeutic targets for virus-induced exacerbations. The aim of this article is to highlight current knowledge regarding the role of viruses and immune modulation in the asthmatic epithelium and to discuss exciting areas for future research and novel treatments.
Collapse
|
219
|
Badloe FMS, Janmohamed SR, Ring J, Gutermuth J. Microbial Triggers in Autoimmunity, Severe Allergy, and Autoallergy. BIRKHÄUSER ADVANCES IN INFECTIOUS DISEASES 2017:11-26. [DOI: 10.1007/978-3-319-69968-4_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
220
|
Wittekindt OH. Tight junctions in pulmonary epithelia during lung inflammation. Pflugers Arch 2017; 469:135-147. [PMID: 27921210 PMCID: PMC5203840 DOI: 10.1007/s00424-016-1917-3] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 11/23/2016] [Accepted: 11/27/2016] [Indexed: 12/31/2022]
Abstract
Inflammatory lung diseases like asthma bronchiale, chronic obstructive pulmonary disease and allergic airway inflammation are widespread public diseases that constitute an enormous burden to the health systems. Mainly classified as inflammatory diseases, the treatment focuses on strategies interfering with local inflammatory responses by the immune system. Inflammatory lung diseases predispose patients to severe lung failures like alveolar oedema, respiratory distress syndrome and acute lung injury. These life-threatening syndromes are caused by increased permeability of the alveolar and airway epithelium and exudate formation. However, the mechanism underlying epithelium barrier breakdown in the lung during inflammation is elusive. This review emphasises the role of the tight junction of the airway epithelium as the predominating structure conferring epithelial tightness and preventing exudate formation and the impact of inflammatory perturbations on their function.
Collapse
Affiliation(s)
- Oliver H Wittekindt
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany.
| |
Collapse
|
221
|
Rhinovirus-induced asthma exacerbations and risk populations. Curr Opin Allergy Clin Immunol 2016; 16:179-85. [PMID: 26836624 DOI: 10.1097/aci.0000000000000245] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW This article discusses recent findings into the mechanisms that determine how viruses trigger asthma exacerbations. RECENT FINDINGS Substantial progress has been made in our understanding of the pathogenesis of virus-induced asthma exacerbations. This includes new insights into the role of bacteria, the regulation of interferon responses, and the discovery of innate immune pathways that link viral infections with allergic inflammation. Progress has also been made in elucidating the genetic risk factors for asthma exacerbations, most notably the contribution of the ORMDL3/GSDMB locus on 17q, the mechanisms underlying the farming effect, and the discovery that CDHR3 binds to rhinovirus species C. SUMMARY Asthma exacerbations are heterogeneous conditions that involve the complex interplay between environmental exposures and innate and adaptive immune function in genetically predisposed individuals. Recent insights into the interrelationships between these factors provide new opportunities for therapeutic intervention.
Collapse
|
222
|
Interactions of Respiratory Viruses and the Nasal Microbiota during the First Year of Life in Healthy Infants. mSphere 2016; 1:mSphere00312-16. [PMID: 27904883 PMCID: PMC5120172 DOI: 10.1128/msphere.00312-16] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 10/29/2016] [Indexed: 11/20/2022] Open
Abstract
Respiratory viral infections are very frequent in infancy and of importance in acute and chronic disease development. Infections with human rhinovirus (HRV) are, e.g., associated with the later development of asthma. We found that only symptomatic HRV infections were associated with acute changes in the nasal microbiota, mainly characterized by a loss of microbial diversity. Infants with more frequent symptomatic HRV infections had a lower bacterial diversity at the end of the first year of life. Whether the interaction between viruses and the microbiota is one pathway contributing to asthma development will be assessed in the follow-ups of these children. Independent of that, measurements of microbial diversity might represent a potential marker for risk of later lung disease or monitoring of early life interventions. Traditional culture techniques have shown that increased bacterial colonization is associated with viral colonization; however, the influence of viral colonization on the whole microbiota composition is less clear. We thus aimed to understand the interaction of viral infections and the nasal microbiota in early life to appraise their roles in disease development. Thirty-two healthy, unselected infants were included in this prospective longitudinal cohort study within the first year of life. Biweekly nasal swabs (n = 559) were taken, and the microbiota was analyzed by 16S rRNA pyrosequencing, and 10 different viruses and 2 atypical bacteria were characterized by real-time PCR (combination of seven duplex samples). In contrast to asymptomatic human rhinovirus (HRV) colonization, symptomatic HRV infections were associated with lower alpha diversity (Shannon diversity index [SDI]), higher bacterial density (PCR concentration), and a difference in beta diversities (Jaccard and Bray-Curtis index) of the microbiota. In addition, infants with more frequent HRV infections had a lower SDI at the end of the study period. Overall, changes in the microbiota associated with symptomatic HRV infections were characterized by a loss of microbial diversity. The interaction between HRV infections and the nasal microbiota in early life might be of importance for later disease development and indicate a potential approach for future interventions. IMPORTANCE Respiratory viral infections are very frequent in infancy and of importance in acute and chronic disease development. Infections with human rhinovirus (HRV) are, e.g., associated with the later development of asthma. We found that only symptomatic HRV infections were associated with acute changes in the nasal microbiota, mainly characterized by a loss of microbial diversity. Infants with more frequent symptomatic HRV infections had a lower bacterial diversity at the end of the first year of life. Whether the interaction between viruses and the microbiota is one pathway contributing to asthma development will be assessed in the follow-ups of these children. Independent of that, measurements of microbial diversity might represent a potential marker for risk of later lung disease or monitoring of early life interventions.
Collapse
|
223
|
Troy NM, Bosco A. Respiratory viral infections and host responses; insights from genomics. Respir Res 2016; 17:156. [PMID: 27871304 PMCID: PMC5117516 DOI: 10.1186/s12931-016-0474-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 11/10/2016] [Indexed: 01/23/2023] Open
Abstract
Respiratory viral infections are a leading cause of disease and mortality. The severity of these illnesses can vary markedly from mild or asymptomatic upper airway infections to severe wheezing, bronchiolitis or pneumonia. In this article, we review the viral sensing pathways and organizing principles that govern the innate immune response to infection. Then, we reconstruct the molecular networks that differentiate symptomatic from asymptomatic respiratory viral infections, and identify the underlying molecular drivers of these networks. Finally, we discuss unique aspects of the biology and pathogenesis of infections with respiratory syncytial virus, rhinovirus and influenza, drawing on insights from genomics.
Collapse
Affiliation(s)
- Niamh M Troy
- Telethon Kids Institute, The University of Western Australia, Subiaco, Australia
| | - Anthony Bosco
- Telethon Kids Institute, The University of Western Australia, Subiaco, Australia.
| |
Collapse
|
224
|
Song DJ. Rhinovirus and childhood asthma: an update. KOREAN JOURNAL OF PEDIATRICS 2016; 59:432-439. [PMID: 27895690 PMCID: PMC5118502 DOI: 10.3345/kjp.2016.59.11.432] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 10/18/2015] [Accepted: 10/23/2015] [Indexed: 01/26/2023]
Abstract
Asthma is recognized as a complex disease resulting from interactions between multiple genetic and environmental factors. Accumulating evidence suggests that respiratory viral infections in early life constitute a major environmental risk factor for the development of childhood asthma. Respiratory viral infections have also been recognized as the most common cause of asthma exacerbation. The advent of molecular diagnostics to detect respiratory viruses has provided new insights into the role of human rhinovirus (HRV) infections in the pathogenesis of asthma. However, it is still unclear whether HRV infections cause asthma or if wheezing with HRV infection is simply a predictor of childhood asthma. Recent clinical and experimental studies have identified plausible pathways by which HRV infection could cause asthma, particularly in a susceptible host, and exacerbate disease. Airway epithelial cells, the primary site of infection and replication of HRV, play a key role in these processes. Details regarding the role of genetic factors, including ORMDL3, are beginning to emerge. This review discusses recent clinical and experimental evidence for the role of HRV infection in the development and exacerbation of childhood asthma and the potential underlying mechanisms that have been proposed.
Collapse
Affiliation(s)
- Dae Jin Song
- Department of Pediatrics, Korea University College of Medicine, Seoul, Korea.; Environmental Health Center for Childhood Asthma, Korea University Anam Hospital, Seoul, Korea
| |
Collapse
|
225
|
Głobińska A, Pawełczyk M, Piechota-Polańczyk A, Olszewska-Ziąber A, Moskwa S, Mikołajczyk A, Jabłońska A, Zakrzewski PK, Brauncajs M, Jarzębska M, Taka S, Papadopoulos NG, Kowalski ML. Impaired virus replication and decreased innate immune responses to viral infections in nasal epithelial cells from patients with allergic rhinitis. Clin Exp Immunol 2016; 187:100-112. [PMID: 27667736 DOI: 10.1111/cei.12869] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2016] [Indexed: 12/17/2022] Open
Abstract
The aim of this study was to assess the immune response to parainfluenza virus type 3 (PIV3), rhinovirus 1B (RV1B) and intracellular Toll-like receptors (TLR) agonists in nasal epithelial cells (NECs) from patients with allergic rhinitis and healthy controls. NECs were obtained from eight patients with allergic rhinitis (AR) and 11 non-atopic healthy controls (HC) by nasal scraping, grown to confluence and exposed to PIV3, RV1B infection or TLR-3 and TLR-7/8 agonists. Interferon (IFN)-λ1, IFN-α, IFN-β and regulated on activation, normal T expressed and secreted (RANTES) release into the cell culture supernatants was assessed at 8, 24 and 48 h upon infection or 8 and 24 h after stimulation with poly(I:C) and R848. mRNA levels of IFNs, RANTES, interferon regulatory transcription factor (IRF)3, IRF7 and viral gene copy number were determined using real-time polymerase chain reaction (RT-PCR). PIV3 but not RV1B replication 48 h after infection was significantly lower (P < 0·01) in NECs from AR patients compared to HC. PIV3 infection induced significantly less IFN-λ1 (both protein and mRNA) in NECs from AR compared to HC. IFN-β mRNA expression and RANTES protein release and mRNA expression tended to be smaller in AR compared HC cells in response to both viruses. Stimulation with TLR-3 agonist [poly (I:C)] induced similar IFN-λ1 and RANTES generation in AR and HC subjects. Viral infections in NECs induced IRF7 expression, which correlated with IFN and RANTES expression. These data suggest that virus proliferation rates and the immune response profile are different in nasal epithelial cells from patients with allergic rhinitis compared to healthy individuals.
Collapse
Affiliation(s)
- A Głobińska
- Department of Immunology, Rheumatology and Allergy, Medical University of Łódź, Łódź, Poland.,Healthy Ageing Research Center, Medical University of Łódź, Łódź, Poland
| | - M Pawełczyk
- Department of Immunology, Rheumatology and Allergy, Medical University of Łódź, Łódź, Poland.,Healthy Ageing Research Center, Medical University of Łódź, Łódź, Poland
| | - A Piechota-Polańczyk
- Department of Immunology, Rheumatology and Allergy, Medical University of Łódź, Łódź, Poland.,Healthy Ageing Research Center, Medical University of Łódź, Łódź, Poland
| | - A Olszewska-Ziąber
- Department of Immunology, Rheumatology and Allergy, Medical University of Łódź, Łódź, Poland.,Healthy Ageing Research Center, Medical University of Łódź, Łódź, Poland
| | - S Moskwa
- Department of Immunology, Rheumatology and Allergy, Medical University of Łódź, Łódź, Poland.,Healthy Ageing Research Center, Medical University of Łódź, Łódź, Poland.,Microbiology and Laboratory Medical Immunology Department, Medical University of Łódź, Łódź, Poland
| | - A Mikołajczyk
- Department of Immunology, Rheumatology and Allergy, Medical University of Łódź, Łódź, Poland.,Healthy Ageing Research Center, Medical University of Łódź, Łódź, Poland
| | - A Jabłońska
- Department of Immunology, Rheumatology and Allergy, Medical University of Łódź, Łódź, Poland.,Healthy Ageing Research Center, Medical University of Łódź, Łódź, Poland
| | - P K Zakrzewski
- Department of Immunology, Rheumatology and Allergy, Medical University of Łódź, Łódź, Poland.,Healthy Ageing Research Center, Medical University of Łódź, Łódź, Poland
| | - M Brauncajs
- Microbiology and Laboratory Medical Immunology Department, Medical University of Łódź, Łódź, Poland
| | - M Jarzębska
- Department of Immunology, Rheumatology and Allergy, Medical University of Łódź, Łódź, Poland.,Healthy Ageing Research Center, Medical University of Łódź, Łódź, Poland
| | - S Taka
- Allergy Department, Second Paediatric Clinic, University of Athens, Athens, Greece
| | - N G Papadopoulos
- Allergy Department, Second Paediatric Clinic, University of Athens, Athens, Greece
| | - M L Kowalski
- Department of Immunology, Rheumatology and Allergy, Medical University of Łódź, Łódź, Poland.,Healthy Ageing Research Center, Medical University of Łódź, Łódź, Poland
| |
Collapse
|
226
|
Lan F, Zhang N, Gevaert E, Zhang L, Bachert C. Viruses and bacteria in Th2-biased allergic airway disease. Allergy 2016; 71:1381-92. [PMID: 27188632 DOI: 10.1111/all.12934] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2016] [Indexed: 01/24/2023]
Abstract
Allergic airway diseases are typically characterized by a type 2-biased inflammation. Multiple distinct viruses and bacteria have been detected in the airways. Recently, it has been confirmed that the microbiome of allergic individuals differs from that of healthy subjects, showing a close relationship with the type 2 response in allergic airway disease. In this review, we summarize the recent findings on the prevalence of viruses and bacteria in type 2-biased airway diseases and on the mechanisms employed by viruses and bacteria in propagating type 2 responses. The understanding of the microbial composition and postinfectious immune programming is critical for the reconstruction of the normal microflora and immune status in allergic airway diseases.
Collapse
Affiliation(s)
- F. Lan
- Upper Airways Research Laboratory; ENT Department; Ghent University; Gent Belgium
- Department of Otolaryngology Head and Neck Surgery; Beijing Tongren Hospital; Capital Medical University; Beijing China
| | - N. Zhang
- Upper Airways Research Laboratory; ENT Department; Ghent University; Gent Belgium
| | - E. Gevaert
- Upper Airways Research Laboratory; ENT Department; Ghent University; Gent Belgium
| | - L. Zhang
- Department of Otolaryngology Head and Neck Surgery; Beijing Tongren Hospital; Capital Medical University; Beijing China
| | - C. Bachert
- Upper Airways Research Laboratory; ENT Department; Ghent University; Gent Belgium
- Division of ENT Diseases; Clintec; Karolinska Institute; Stockholm Sweden
| |
Collapse
|
227
|
Glanville N, Peel TJ, Schröder A, Aniscenko J, Walton RP, Finotto S, Johnston SL. Tbet Deficiency Causes T Helper Cell Dependent Airways Eosinophilia and Mucus Hypersecretion in Response to Rhinovirus Infection. PLoS Pathog 2016; 12:e1005913. [PMID: 27683080 PMCID: PMC5040449 DOI: 10.1371/journal.ppat.1005913] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/04/2016] [Indexed: 11/18/2022] Open
Abstract
Current understanding of adaptive immune, particularly T cell, responses to human rhinoviruses (RV) is limited. Memory T cells are thought to be of a primarily T helper 1 type, but both T helper 1 and T helper 2 memory cells have been described, and heightened T helper 2/ lessened T helper 1 responses have been associated with increased RV-induced asthma exacerbation severity. We examined the contribution of T helper 1 cells to RV-induced airways inflammation using mice deficient in the transcription factor T-Box Expressed In T Cells (Tbet), a critical controller of T helper 1 cell differentiation. Using flow cytometry we showed that Tbet deficient mice lacked the T helper 1 response of wild type mice and instead developed mixed T helper 2/T helper 17 responses to RV infection, evidenced by increased numbers of GATA binding protein 3 (GATA-3) and RAR-related orphan receptor gamma t (RORγt), and interleukin-13 and interleukin-17A expressing CD4+ T cells in the lung. Forkhead box P3 (FOXP3) and interleukin-10 expressing T cell numbers were unaffected. Tbet deficient mice also displayed deficiencies in lung Natural Killer, Natural Killer T cell and γδT cell responses, and serum neutralising antibody responses. Tbet deficient mice exhibited pronounced airways eosinophilia and mucus production in response to RV infection that, by utilising a CD4+ cell depleting antibody, were found to be T helper cell dependent. RV induction of T helper 2 and T helper 17 responses may therefore have an important role in directly driving features of allergic airways disease such as eosinophilia and mucus hypersecretion during asthma exacerbations.
Collapse
Affiliation(s)
- Nicholas Glanville
- Airway Disease Infection Section, National Heart and Lung Institute, MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
| | - Tamlyn J. Peel
- Airway Disease Infection Section, National Heart and Lung Institute, MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
| | - Armin Schröder
- Laboratory of Cellular and Molecular Lung Immunology, Department of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Julia Aniscenko
- Airway Disease Infection Section, National Heart and Lung Institute, MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
| | - Ross P. Walton
- Airway Disease Infection Section, National Heart and Lung Institute, MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
| | - Susetta Finotto
- Laboratory of Cellular and Molecular Lung Immunology, Department of Molecular Pneumology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sebastian L. Johnston
- Airway Disease Infection Section, National Heart and Lung Institute, MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
228
|
Abstract
Chronic airway diseases are a significant cause of morbidity and mortality worldwide, and their prevalence is predicted to increase in the future. Respiratory viruses are the most common cause of acute pulmonary infection, and there is clear evidence of their role in acute exacerbations of inflammatory airway diseases such as asthma and chronic obstructive pulmonary disease. Studies have reported impaired host responses to virus infection in these diseases, and a better understanding of the mechanisms of these abnormal immune responses has the potential to lead to the development of novel therapeutic targets for virus-induced exacerbations. The aim of this article is to review the current knowledge regarding the role of viruses and immune modulation in acute exacerbations of chronic pulmonary diseases and to discuss exciting areas for future research and novel treatments.
Collapse
|
229
|
Jackson DJ, Gern JE, Lemanske RF. The contributions of allergic sensitization and respiratory pathogens to asthma inception. J Allergy Clin Immunol 2016; 137:659-65; quiz 666. [PMID: 26947979 PMCID: PMC4782609 DOI: 10.1016/j.jaci.2016.01.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 01/06/2016] [Accepted: 01/13/2016] [Indexed: 01/10/2023]
Abstract
Of the chronic diseases affecting grade-school children, asthma is the most common and accounts for the greatest number of school days missed. Moreover, it can influence family dynamics and function in other ways, and unfortunately, it can also be associated with mortality, particularly in the inner-city environments of the United States. Thus understanding factors that lead to its development in early life is essential in developing strategies aimed at primary prevention. Two risk factors that have been identified by a number of investigators include the development of allergic sensitization and wheezing respiratory tract illnesses caused by viruses and bacteria, either alone or in combination. Both of these factors appear to exert their influences within the first few years of life, such that asthma becomes established before the child enters grade school at age 5 to 6 years. Therefore, because both allergic sensitization and viral and bacterial illnesses can occur in children who do not have asthma, it is paramount to identify genetic and environmental factors that activate, interact with, and/or direct the immune system and components of the respiratory tract along pathways that allow asthma to become established and expressed clinically.
Collapse
Affiliation(s)
- Daniel J Jackson
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wis.
| | - James E Gern
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wis; Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Robert F Lemanske
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wis; Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| |
Collapse
|
230
|
Biologics and the lung: TSLP and other epithelial cell-derived cytokines in asthma. Pharmacol Ther 2016; 169:104-112. [PMID: 27365223 DOI: 10.1016/j.pharmthera.2016.06.009] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 06/13/2016] [Indexed: 12/21/2022]
Abstract
Asthma is a chronic airway inflammatory disorder with characteristic symptoms of dyspnea, wheeze, chest tightness and cough, and physiological abnormalities of variable airway obstruction, airway hyperresponsiveness, and in some patients with chronic long standing disease reduced lung function. The physiological abnormalities are due to chronic airway inflammation and underlying structural changes to the airway wall. The interaction between the airway epithelium and the environment is crucial to the pathobiology of asthma. Several recent discoveries have highlighted a crucial role of airway epithelial derived cytokines such as interleukin (IL)-25, IL-33 and thymic stromal lymphopoietin (TSLP). These cytokines are collectively known as epithelial "alarmins", which act solely or in concert to activate and potentiate the innate and humoral arms of the immune system in the presence of actual or perceive damage. Understanding the role of alarmins and how they are activated and released may allow the development of novel new therapeutics to treat asthma. This review describes the interactions between inhaled air, the pulmonary microbiome, airway epithelial cell layer and the alarmins, IL-25, IL-33 and TSLP. There is already compelling evidence for a role of TSLP in the airway responses to environmental allergens in allergic asthmatics, as well as in maintaining airway eosinophilic inflammation in these subjects. Further work is required to develop human monoclonal antibodies (hMabs) directed against IL-25 and IL-33 or their receptors, to help understand their role in the initiation and/or persistence of asthma.
Collapse
|
231
|
Sy CB, Siracusa MC. The Therapeutic Potential of Targeting Cytokine Alarmins to Treat Allergic Airway Inflammation. Front Physiol 2016; 7:214. [PMID: 27378934 PMCID: PMC4906008 DOI: 10.3389/fphys.2016.00214] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 05/23/2016] [Indexed: 01/12/2023] Open
Abstract
Asthma is a heterogeneous disorder that results in recurrent attacks of breathlessness, coughing, and wheezing that affects millions of people worldwide. Although the precise causes of asthma are unclear, studies suggest that a combination of genetic predisposition and environmental exposure to various allergens and pathogens contribute to its development. Currently, the most common treatment to control asthma is a dual combination of β2-adrenergic receptor agonists and corticosteroids. However, studies have shown that some patients do not respond well to these medications, while others experience significant side effects. It is reported that the majority of asthmas are associated with T helper type 2 (TH2) responses. In these patients, allergen challenge initiates the influx of TH2 cells in the airways leading to an increased production of TH2-associated cytokines and the promotion of allergy-induced asthma. Therefore, biologics that target this pathway may provide an alternative method to treat the allergic airway inflammation associated with asthma. As of now, only two biologics (omalizumab and mepolizumab), which target immunoglobulin E and interleukin-5, respectively, are FDA-approved and being prescribed to asthmatics. However, recent studies have reported that targeting other components of the TH2 response also show great promise. In this review, we will briefly describe the immunologic mechanisms underlying allergic asthma. Furthermore, we will discuss the current therapeutic strategies used to treat asthma including their limitations. Finally, we will highlight the benefits of using biologics to treat asthma-associated allergic airway inflammation with an emphasis on the potential of targeting cytokine alarmins, especially thymic stromal lymphopoietin.
Collapse
Affiliation(s)
- Chandler B Sy
- Department of Medicine, Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey Newark, NJ, USA
| | - Mark C Siracusa
- Department of Medicine, Center for Immunity and Inflammation, New Jersey Medical School, Rutgers-The State University of New Jersey Newark, NJ, USA
| |
Collapse
|
232
|
Smits HH, van der Vlugt LE, von Mutius E, Hiemstra PS. Childhood allergies and asthma: New insights on environmental exposures and local immunity at the lung barrier. Curr Opin Immunol 2016; 42:41-47. [PMID: 27254380 DOI: 10.1016/j.coi.2016.05.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 05/13/2016] [Accepted: 05/16/2016] [Indexed: 12/11/2022]
Abstract
While certain bacteria and respiratory viruses promote local inflammation and disease onset, a more diverse colonization of the different species in the (gut) microbiome may be linked to more regulatory responses and protection against asthma and allergies. These processes are also influenced in part by food intake, both targeting the composition of the gut microbiome and influencing the immune system via metabolites. Early life environmental microbial exposure also contributes to protection against asthma and allergy and is linked with an early activation of the innate immune system and the development of regulatory immune responses. Although greater mechanistic insight is needed, it is tempting to speculate that part of the environmental effect can be explained by modulation of the microbiome composition at mucosal surfaces, epithelial barrier function and/or local immunity. A review of the latest studies is provided.
Collapse
Affiliation(s)
- Hermelijn H Smits
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Luciën Epm van der Vlugt
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands; Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Erika von Mutius
- Dr von Hauner Children's Hospital, Ludwig Maximilians University of Munich, Munich, Germany; Comprehensive Pneumology Centre Munich (CPC-M), Member of the German Center for Lung Research, Germany
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
233
|
Abstract
Viral exacerbations continue to represent the major burden in terms of morbidity, mortality and health care costs associated with asthma. Those at greatest risk for acute asthma are those with more severe airways disease and poor asthma control. It is this group with established asthma in whom acute exacerbations triggered by virus infections remain a serious cause of increased morbidity. A range of novel therapies are emerging to treat asthma and in particular target this group with poor disease control, and in most cases their efficacy is now being judged by their ability to reduce the frequency of acute exacerbations. Critical for the development of new treatment approaches is an improved understanding of virus-host interaction in the context of the asthmatic airway. This requires research into the virology of the disease in physiological models in conjunction with detailed phenotypic characterisation of asthma patients to identify targets amenable to therapeutic intervention.
Collapse
Affiliation(s)
- Hock Tay
- a Hunter Medical Research Institute , Newcastle , Australia.,b Priority Research Centre for Healthy Lungs , The University of Newcastle , Australia
| | - Peter A B Wark
- a Hunter Medical Research Institute , Newcastle , Australia.,b Priority Research Centre for Healthy Lungs , The University of Newcastle , Australia.,c Centre of Excellence in Severe Asthma , The University of Newcastle , Australia.,d Department of Respiratory and Sleep Medicine , John Hunter Hospital , Newcastle , Australia
| | - Nathan W Bartlett
- a Hunter Medical Research Institute , Newcastle , Australia.,b Priority Research Centre for Healthy Lungs , The University of Newcastle , Australia.,e National Heart and Lung Institute , Imperial College London , London , UK
| |
Collapse
|
234
|
Abstract
Recent discoveries have led to the identification of a novel group of immune cells, the innate lymphoid cells (ILCs). The members of this group are divided into three subpopulations: ILC1s, ILC2s, and ILC3s. ILC2s produce Th2 cytokines, IL-4, IL-5, and IL-13, upon activation by epithelial cell-derived cytokines, lipid mediators (cysteinyl leukotrienes and prostaglandin D2), and TNF family member TL1A and promote structural and immune cell responses in the airways after antigen exposure. In addition, ILC2 function is also influenced by inducible T cell costimulator (ICOS)/ICOS-ligand (ICOS-L) interactions via direct contact between immune cells. The most common airway antigens are allergens and viruses which are highly linked to the induction of airway diseases with underlying type 2 inflammation including asthma and allergic rhinitis. Based on recent findings linking ILC2s and airway Th2 responses, there is intensive investigation into the role of ILC2s in human disease with the hope of a better understanding of the pathophysiology and the discovery of novel potential therapeutic targets. This review summarizes the recent advances made in elucidating ILC2 involvement in human Th2 airway disease.
Collapse
|
235
|
Kim HY, Umetsu DT, Dekruyff RH. Innate lymphoid cells in asthma: Will they take your breath away? Eur J Immunol 2016; 46:795-806. [PMID: 26891006 DOI: 10.1002/eji.201444557] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 02/08/2016] [Accepted: 02/12/2016] [Indexed: 02/06/2023]
Abstract
Asthma is a complex and heterogeneous disease that is characterized by airway hyper-reactivity (AHR) and airway inflammation. Although asthma was long thought to be driven by allergen-reactive TH 2 cells, it has recently become clear that the pathogenesis of asthma is more complicated and associated with multiple pathways and cell types. A very exciting recent development was the discovery of innate lymphoid cells (ILCs) as key players in the pathogenesis of asthma. ILCs do not express antigen receptors but react promptly to "danger signals" from inflamed tissue and produce an array of cytokines that direct the ensuing immune response. The roles of ILCs may differ in distinct asthma phenotypes. ILC2s may be critical for initiation of adaptive immune responses in inhaled allergen-driven AHR, but may also function independently of adaptive immunity, mediating influenza-induced AHR. ILC2s also contribute to resolution of lung inflammation through their production of amphiregulin. Obesity-induced asthma is associated with expansion of IL-17A-producing ILC3s in the lungs. Furthermore, ILCs may also contribute to steroid-resistant asthma. Although the precise roles of ILCs in different types of asthma are still under investigation, it is clear that inhibition of ILC function represents a potential target that could provide novel treatments for asthma.
Collapse
Affiliation(s)
- Hye Young Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dale T Umetsu
- Genentech, One DNA Way, South San Francisco, CA, USA
| | | |
Collapse
|
236
|
Abstract
PURPOSE OF REVIEW The Th2 pathway starts with the binding of IL-4 to the IL-4 receptor followed by the phosphorylation of signal transducer and activator of transcription 6 and the activation of GATA3. The most important question relates to the sources of IL-4 and IL-4 related inflammation. Which cells other than Th2 cells are responsible for airway inflammation in asthma? RECENT FINDINGS Accumulating data indicate that basophils contribute to endothelium-related IL-4-dependent inflammation. There is also a dendritic cell-related alternative for the induction of Th2 cells via Notch signalling. GATA3 deoxyribozyme improves asthma that is not clearly related to T-cells. The innate immune response in allergy is linked to mast cells, basophils, and the innate lymphoid cell type 2 (ILC2). ILC2s respond to IL-25, IL-33, thymic stromal lymphopoietin, and leukotrienes by producing IL-4, IL-5, and IL-13. In addition to all this inflammatory-cell-driven asthma, increasing evidence has emerged relating to smooth muscle cell activation, the endothelial and epithelial barrier functions, and improvements in the barrier function. The elevation of intracellular cyclic adenosine monophosphate because of the use of phosphodiesterase inhibitors adds to the prevention of epithelial-endothelial leakage, supports airway smooth muscle relaxation, and is immunosuppressive. CONCLUSION AND SUMMARY IL-4 is the predominant Th2 cell cytokine. Many more cells, including eosinophils, basophils, mast cells, and ILC2, contribute to the production of IL-4 in the airways. Epithelial cells and endothelial cells lose barrier function in the context of allergic airway inflammation, and this could be at least partially remedied by increasing the intracellular cyclic adenosine monophosphate levels through phosphodiesterase inhibition.
Collapse
|
237
|
Pathogenesis and prevention strategies of severe asthma exacerbations in children. Curr Opin Pulm Med 2016; 22:25-31. [PMID: 26574720 DOI: 10.1097/mcp.0000000000000223] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW Exacerbations of asthma in children are most frequently precipitated by respiratory infections with a seasonal pattern. However, management takes little account of the underlying infective or other precipitant abnormality. RECENT FINDINGS Interactions between environmental triggers, the airway microbiome and innate immune responses are key determinants of exacerbations. Elevated innate cytokines interleukin (IL)-33 and IL-25, and abnormal molecular responses in the interferon pathway are associated with rhinoviral infections. Exacerbations caused by fungal allergens also induce IL-33, highlighting this as an attractive therapeutic target. An equal contribution of bacterial and viral infection during exacerbations, particularly in preschool children, has become increasingly apparent, but some organisms may be protective. Investigation of mechanisms underlying infection-related exacerbations especially in preschool children is needed.Progressive loss of lung function from exacerbations is most pronounced in children aged 6-11 years, and low FEV1 is now recognized as a key predictor for the development of chronic obstructive pulmonary disease and premature death. Although prevention of exacerbations is critical, suboptimal patient education, prescription and adherence to maintenance therapy, and a lack of predictive biomarkers, remain key unaddressed issues in children. SUMMARY Precipitants and predictors of exacerbations, together with the child's age and clinical phenotype, need to be used to achieve individualized management in preference to the current uniform approach for all.
Collapse
|
238
|
Mahmutovic Persson I, Akbarshahi H, Menzel M, Brandelius A, Uller L. Increased expression of upstream TH2-cytokines in a mouse model of viral-induced asthma exacerbation. J Transl Med 2016; 14:52. [PMID: 26879906 PMCID: PMC4754855 DOI: 10.1186/s12967-016-0808-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 02/04/2016] [Indexed: 12/17/2022] Open
Abstract
Background Exacerbations of asthma caused by respiratory viral infections are serious conditions in need of novel treatment. To this end animal models of asthma exacerbations are warranted. We have shown that dsRNA challenges or rhinoviral infection produce exacerbation effects in mice with ovalbumin (OVA)-induced allergic asthma. However, house dust mite (HDM) is a more human asthma-relevant allergen than OVA. We thus hypothesised that dsRNA challenges in mice with HDM-induced experimental asthma would produce important translational features of asthma exacerbations. Method Mouse airways were challenged locally with HDM or saline three times a week for three weeks to establish experimental asthma. Then daily local dsRNA challenges were given for three consecutive days to induce exacerbation. Bronchoalveolar lavage fluid (BALF) was analysed for inflammatory cells, total protein, the necrosis marker LDH and the alarmin ATP. Lung homogenates were analysed for mRNA expression (RT-qPCR) of TNF-α, CCL2, CCL5, IL-1β, IL-33, thymic stromal lymphopoietin (TSLP), and IL-25 as well as pattern recognition receptors (PRRs) RIG-I, MDA5 and TLR3. Lung tissue IL-33 was analysed with ELISA and PRRs were quantified by western blot. Immunohistochemistry indicated lung distribution of IL-33. Results HDM challenge alone caused sustained increase in BALF total protein, eosinophils, lymphocytes and neutrophils, and transient increase in lung tissue expression of TSLP, IL-33 and TNF-α. dsRNA-induced exacerbation markedly and dose-dependently exaggerated these effects. Further, BALF levels of LDH and ATP, and lung tissue expression of CCL2, CCL5, IL-1β, IL-25 and PRRs were increased exclusively at the exacerbations. Lung protein levels of IL-33 were transiently increased by HDM and further increased at exacerbation. Conclusion We demonstrate several novel aspects of HDM-induced experimental asthma and added exacerbation effects of dsRNA. General inflammatory parameters in BALF such as exuded proteins, mixed granulocytes, LDH and ATP were increased at the present exacerbations as they are in human asthma exacerbations. We suggest that this model of asthma exacerbation involving dsRNA challenges given to mice with established HDM-induced asthma has translational value and suggest that it may be particularly suited for in vivo studies involving pharmacological effects on exacerbation-induced expression of major upstream TH2-cytokines; IL-33, TSLP and IL-25, as well as PRRs. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-0808-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Irma Mahmutovic Persson
- Department Experimental Medical Science, Unit of Respiratory Immunopharmacology, Lund University, BMC D12, 221 84, Lund, Sweden.
| | - Hamid Akbarshahi
- Department Experimental Medical Science, Unit of Respiratory Immunopharmacology, Lund University, BMC D12, 221 84, Lund, Sweden.
| | - Mandy Menzel
- Department Experimental Medical Science, Unit of Respiratory Immunopharmacology, Lund University, BMC D12, 221 84, Lund, Sweden.
| | - Angelica Brandelius
- Department Experimental Medical Science, Unit of Respiratory Immunopharmacology, Lund University, BMC D12, 221 84, Lund, Sweden.
| | - Lena Uller
- Department Experimental Medical Science, Unit of Respiratory Immunopharmacology, Lund University, BMC D12, 221 84, Lund, Sweden.
| |
Collapse
|
239
|
Tung HY, Landers C, Li E, Porter P, Kheradmand F, Corry DB. Allergen-encoded signals that control allergic responses. Curr Opin Allergy Clin Immunol 2016; 16:51-8. [PMID: 26658015 PMCID: PMC4863991 DOI: 10.1097/aci.0000000000000233] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
PURPOSE OF REVIEW The purpose is to review the important recent advances made in how innate immune cells, microbes, and the environment contribute to the expression of allergic disease, emphasizing the allergen-related signals that drive allergic responses. RECENT FINDINGS The last few years have seen crucial advances in how innate immune cells such as innate lymphoid cells group 2 and airway epithelial cells and related molecular pathways through organismal proteinases and innate immune cytokines, such as thymic stromal lymphopoietin, IL-25, and IL-33 contribute to allergy and asthma. Simultaneously with these advances, important progress has been made in our understanding of how the environment, and especially pathogenic organisms, such as bacteria, viruses, helminths, and especially fungi derived from the natural and built environments, either promote or inhibit allergic inflammation and disease. Of specific interest are how lipopolysaccharide mediates its antiallergic effect through the ubiquitin modifying factor A20 and the antiallergic activity of both helminths and protozoa. SUMMARY Innate immune cells and molecular pathways, often activated by allergen-derived proteinases acting on airway epithelium and macrophages as well as additional unknown factors, are essential to the expression of allergic inflammation and disease. These findings suggest numerous future research opportunities and new opportunities for therapeutic intervention in allergic disease.
Collapse
Affiliation(s)
- Hui-Ying Tung
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
| | - Cameron Landers
- Translational Biology and Molecular Medicine Program, Baylor College of Medicine, Houston, Texas, USA
| | - Evan Li
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Paul Porter
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Farrah Kheradmand
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
- Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas, USA
- Michael E. DeBakey VA Center for Translational Research on Inflammatory Diseases, Houston, Texas, USA
| | - David B. Corry
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
- Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas, USA
- Michael E. DeBakey VA Center for Translational Research on Inflammatory Diseases, Houston, Texas, USA
| |
Collapse
|
240
|
Regulation of IL-4 Expression in Immunity and Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 941:31-77. [PMID: 27734408 DOI: 10.1007/978-94-024-0921-5_3] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
IL-4 was first identified as a T cell-derived growth factor for B cells. Studies over the past several decades have markedly expanded our understanding of its cellular sources and function. In addition to T cells, IL-4 is produced by innate lymphocytes, such as NTK cells, and myeloid cells, such as basophils and mast cells. It is a signature cytokine of type 2 immune response but also has a nonimmune function. Its expression is tightly regulated at several levels, including signaling pathways, transcription factors, epigenetic modifications, microRNA, and long noncoding RNA. This chapter will review in detail the molecular mechanism regulating the cell type-specific expression of IL-4 in physiological and pathological type 2 immune responses.
Collapse
|
241
|
Yao X, Sun Y, Wang W, Sun Y. Interleukin (IL)-25: Pleiotropic roles in asthma. Respirology 2015; 21:638-47. [PMID: 26699081 DOI: 10.1111/resp.12707] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 09/06/2015] [Accepted: 09/23/2015] [Indexed: 12/29/2022]
Abstract
IL-25, also named IL-17E, is a distinct member of the IL-17 cytokine family, which can promote and augment T helper type 2 (Th2) responses locally or systemically. Growing evidence from experimental and clinical studies indicates that the expression of IL-25 and its cognate receptor, IL-17RB/RA, is markedly upregulated in asthmatic conditions. It has also been found that IL-25 induces not only typical eosinophilic inflammation and airway hyperresponsiveness (AHR), but also airway remodelling, manifested by goblet cell hyperplasia, subepithelial collagen deposition and angiogenesis. This review will focus on the discovery, cellular origins and targets of IL-25, and try to update current animal and human studies elucidating the roles of IL-25 in asthma. We conclude that although IL-25 is a pleiotropic cytokine, it may only play its dominant role in a certain specific asthmatic endotype, named 'IL-25 high' phenotype. Thus, targeting IL-25 or its receptor might selectively benefit some subgroups with asthma. Furthermore, the major IL-25 producing as well as responsive cells in the changeable milieu of asthma should be assessed in the future.
Collapse
Affiliation(s)
- Xiujuan Yao
- Department of Respiratory Medicine, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yongchang Sun
- Department of Respiratory Medicine, Beijing Tongren Hospital, Capital Medical University, Beijing, China.,Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| | - Wei Wang
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ying Sun
- School of Basic Medical Sciences, Capital Medical University, Beijing, China.,King's College London, MRC and Asthma UK Centre in Allergic Mechanisms of Asthma, Division of Asthma, Allergy and Lung Biology, London, UK
| |
Collapse
|
242
|
Hams E, Bermingham R, Fallon PG. Macrophage and Innate Lymphoid Cell Interplay in the Genesis of Fibrosis. Front Immunol 2015; 6:597. [PMID: 26635811 PMCID: PMC4655423 DOI: 10.3389/fimmu.2015.00597] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 11/06/2015] [Indexed: 01/15/2023] Open
Abstract
Fibrosis is a characteristic pathological feature of an array of chronic diseases, where development of fibrosis in tissue can lead to marked alterations in the architecture of the affected organs. As a result of this process of sustained attrition to organs, many diseases that involve fibrosis are often progressive conditions and have a poor long-term prognosis. Inflammation is often a prelude to fibrosis, with innate and adaptive immunity involved in both the initiation and regulation of the fibrotic process. In this review, we will focus on the emerging roles of the newly described innate lymphoid cells (ILCs) in the generation of fibrotic disease with an examination of the potential interplay between ILC and macrophages and the adaptive immune system.
Collapse
Affiliation(s)
- Emily Hams
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin , Dublin , Ireland
| | - Rachel Bermingham
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin , Dublin , Ireland
| | - Padraic G Fallon
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin , Dublin , Ireland
| |
Collapse
|
243
|
Bredo G, Storie J, Shrestha Palikhe N, Davidson C, Adams A, Vliagoftis H, Cameron L. Interleukin-25 initiates Th2 differentiation of human CD4(+) T cells and influences expression of its own receptor. IMMUNITY INFLAMMATION AND DISEASE 2015; 3:455-68. [PMID: 26734466 PMCID: PMC4693727 DOI: 10.1002/iid3.87] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 08/20/2015] [Accepted: 09/05/2015] [Indexed: 12/15/2022]
Abstract
Human CRTh2+ Th2 cells express IL‐25 receptor (IL‐25R) and IL‐25 has been shown to potentiate production of Th2 cytokines. However, regulation of IL‐25R and whether it participates in Th2 differentiation of human cells have not been examined. We sought to characterize IL‐25R expression on CD4+ T cells and determine whether IL‐25 plays a role in Th2 differentiation. Naïve human CD4+ T cells were activated in the presence of IL‐25, IL‐4 (Th2 conditions) or both cytokines to assess their relative influence on Th2 differentiation. For experiments with differentiated Th2 cells, CRTh2‐expressing cells were isolated from differentiating cultures. IL‐25R, GATA3, CRTh2 and Th2 cytokine expression were assessed by flow cytometry, qRT‐PCR and ELISA. Expression of surface IL‐25R was induced early during Th2 differentiation (2 days). Addition of IL‐25 to naïve CD4+ T cells revealed that it induces expression of its own receptor, more strongly than IL‐4. IL‐25 also increased the proportions of IL‐4‐, GATA3‐ and CRTh2‐expressing cells and expression of IL‐5 and IL‐13. Activation of differentiated CRTh2+ Th2 cells through the TCR or by CRTh2 agonist increased surface expression of IL‐25R, though re‐expression of CRTh2 following TCR downregulation was impeded by IL‐25. These data suggest that IL‐25 may play various roles in Th2 mediated immunity. We establish here it regulates expression of its own receptor and can initiate Th2 differentiation, though not as strongly as IL‐4.
Collapse
Affiliation(s)
- Graeme Bredo
- Pulmonary Research Group, Department of Medicine University of Alberta Edmonton Alberta Canada
| | - Jessica Storie
- Pulmonary Research Group, Department of Medicine University of Alberta Edmonton Alberta Canada
| | - Nami Shrestha Palikhe
- Pulmonary Research Group, Department of Medicine University of Alberta Edmonton Alberta Canada
| | - Courtney Davidson
- Pulmonary Research Group, Department of Medicine University of Alberta Edmonton Alberta Canada
| | - Alexis Adams
- Pulmonary Research Group, Department of Medicine University of Alberta Edmonton Alberta Canada
| | - Harissios Vliagoftis
- Pulmonary Research Group, Department of Medicine University of Alberta Edmonton Alberta Canada
| | - Lisa Cameron
- Pulmonary Research Group, Department of MedicineUniversity of AlbertaEdmontonAlbertaCanada; Department of Pathology and Laboratory Medicine, Schulich School of Medicine & DentistryWestern UniversityLondonOntarioCanada
| |
Collapse
|
244
|
Claudio E, Tassi I, Wang H, Tang W, Ha HL, Siebenlist U. Cutting Edge: IL-25 Targets Dendritic Cells To Attract IL-9-Producing T Cells in Acute Allergic Lung Inflammation. THE JOURNAL OF IMMUNOLOGY 2015; 195:3525-9. [PMID: 26371249 DOI: 10.4049/jimmunol.1500436] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 08/18/2015] [Indexed: 12/17/2022]
Abstract
Asthma is a common inflammatory disease of airways that is often associated with type 2 responses triggered by allergens, such as house dust mites (HDMs). IL-25 is a key mucosal cytokine that may be produced by stressed epithelial cells; it rapidly activates type 2 innate lymphoid cells to produce IL-13 and IL-5. When administered directly into lungs, IL-25 induces acute inflammation. However, the mechanisms underlying IL-25-initiated inflammation and the roles of this cytokine in the context of HDM-induced allergic inflammation are not fully understood. We show in this article that lung-resident conventional dendritic cells were direct targets of IL-25. IL-25-stimulated dendritic cells rapidly induced mediators, such as the chemokine CCL17, which, in turn, attracted IL-9-producing T cells. Importantly, these mechanisms also operated during HDM-induced allergic lung inflammation.
Collapse
Affiliation(s)
- Estefania Claudio
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1876
| | - Ilaria Tassi
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1876
| | - Hongshan Wang
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1876
| | - Wanhu Tang
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1876
| | - Hye-Lin Ha
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1876
| | - Ulrich Siebenlist
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1876
| |
Collapse
|
245
|
Halim TYF. Group 2 innate lymphoid cells in disease. Int Immunol 2015; 28:13-22. [PMID: 26306498 DOI: 10.1093/intimm/dxv050] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 08/18/2015] [Indexed: 12/11/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2) are now recognized as an important innate source of type-2 effector cytokines. Although initially associated with mucosal tissues, it is clear that ILC2 are present in diverse anatomical locations. The function of ILC2 at these sites is equally varied, and although ILC2 represent a relatively minor population, they are fundamentally important regulators of innate and adaptive immune processes. As such, there is much interest to understand the role of ILC2 in diseases with a type-2 inflammatory component. This review explores the known roles of ILC2 in disease, and the diseases that show associations or other strong evidence for the involvement of ILC2.
Collapse
Affiliation(s)
- Timotheus Y F Halim
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| |
Collapse
|
246
|
Bertrand P, Lay MK, Piedimonte G, Brockmann PE, Palavecino CE, Hernández J, León MA, Kalergis AM, Bueno SM. Elevated IL-3 and IL-12p40 levels in the lower airway of infants with RSV-induced bronchiolitis correlate with recurrent wheezing. Cytokine 2015; 76:417-423. [PMID: 26299549 DOI: 10.1016/j.cyto.2015.07.017] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 07/17/2015] [Accepted: 07/20/2015] [Indexed: 12/30/2022]
Abstract
Respiratory Syncytial Virus (RSV) is the first cause of hospitalization due to bronchiolitis in infants. RSV bronchiolitis has been linked to asthma and recurrent wheezing, however the mechanisms behind this association have not been elucidated. Here, we evaluated the cytokine and chemokine profiles in the airways in infants with RSV bronchiolitis. Nasopharyngeal Aspirates (NPA) and Bronchoalveolar Lavage Fluids (BALF) from infants hospitalized due to RSV bronchiolitis and healthy controls were analyzed for cytokine and chemokine production. We observed elevated levels of Th2 cytokines (IL-3, IL-4, IL-10 and IL-13), pro-inflammatory cytokines and chemokines (IL-1β, IL-6, TNF-β, MCP-1/CCL2, MIP-1α/CCL3 and IL-8/CXCL8) in BALF from infants with RSV bronchiolitis, as compared to controls. We found a direct correlation of IL-3 and IL-12p40 levels with the development of recurrent wheezing later in life. These results suggest that IL-3 and IL-12p40 could be considered as molecular predictors for recurrent wheezing due to RSV infection.
Collapse
Affiliation(s)
- Pablo Bertrand
- División de Pediatría, Unidad de Enfermedades Respiratorias Pediátricas, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Margarita K Lay
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Giovanni Piedimonte
- The Cleveland Clinic Pediatric Institute and Children's Hospital, Cleveland, OH, United States
| | - Pablo E Brockmann
- División de Pediatría, Unidad de Enfermedades Respiratorias Pediátricas, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Christian E Palavecino
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jury Hernández
- División de Pediatría, Unidad de Enfermedades Respiratorias Pediátricas, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Miguel A León
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Reumatología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile; INSERM U1064, Nantes, France
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Reumatología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
247
|
Wang JY. The sticky relationship between allergies and infections. Asia Pac Allergy 2015; 5:133-5. [PMID: 26240789 PMCID: PMC4521161 DOI: 10.5415/apallergy.2015.5.3.133] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 07/24/2015] [Indexed: 11/22/2022] Open
Affiliation(s)
- Jiu-Yao Wang
- Division of Allergy and Clinical Immunology, Department of Pediatrics, National Cheng Kung University College of Medicine, Tainan 704, Taiwan
| |
Collapse
|
248
|
Fainardi V, Saglani S. The need to differentiate between adults and children when treating severe asthma. Expert Rev Respir Med 2015; 9:419-28. [PMID: 26175269 DOI: 10.1586/17476348.2015.1068693] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Severe asthma at all ages is heterogeneous incorporating several phenotypes that are distinct in children and adults, however, there are also numerous similar features including the limitation that they may not remain stable longitudinally. Severe asthma in both children and adults is characterized by eosinophilic airway inflammation and evidence of airway remodeling. In adults, targeting eosinophilia with anti-IL-5 antibody therapy is very successful, resulting in the recommendation that sputum eosinophils should be used to guide treatment. In contrast, data for the efficacy of blocking IL-5 remain unavailable in children. However, its effectiveness is uncertain since many children with severe asthma have normal blood eosinophils and the dominance of Th2-mediated inflammation is controversial. Approaches that have revealed gene signatures and biomarkers such as periostin that are specific to adult disease now need to be adopted in children to identify effective pediatric specific therapeutics and minimize the extrapolation of adult therapeutics to children.
Collapse
Affiliation(s)
- Valentina Fainardi
- Leukocyte Biology and Respiratory Paediatrics, National Heart and Lung Institute, Imperial College London, London, UK
| | | |
Collapse
|
249
|
Hammond C, Kurten M, Kennedy JL. Rhinovirus and asthma: a storied history of incompatibility. Curr Allergy Asthma Rep 2015; 15:502. [PMID: 25612798 DOI: 10.1007/s11882-014-0502-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The human rhinovirus (HRV) is commonly associated with loss of asthma symptom control requiring escalation of care and emergency room visits in many patients. While the association is clear, the mechanisms behind HRV-induced asthma exacerbations remain uncertain. Immune dysregulation via aberrant immune responses, both deficient and exaggerated, have been proposed as mechanisms for HRV-induced exacerbations of asthma. Epithelium-derived innate immune cytokines that bias Th2 responses, including interleukin (IL)-25, IL-33, and thymic stromal lymphopoietin (TSLP), have also been implicated as a means to bridge allergic conditions with asthma exacerbations. In this review, we discuss the literature supporting these positions. We also discuss new and emerging biotherapeutics that may target virus-induced exacerbations of asthma.
Collapse
Affiliation(s)
- Catherine Hammond
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA,
| | | | | |
Collapse
|
250
|
Kabata H, Moro K, Koyasu S, Asano K. Group 2 innate lymphoid cells and asthma. Allergol Int 2015; 64:227-34. [PMID: 26117253 DOI: 10.1016/j.alit.2015.03.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 03/23/2015] [Accepted: 03/24/2015] [Indexed: 01/21/2023] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) are recently identified cell populations that produce type 2 cytokines such as IL-5 and IL-13 in response to epithelial cell-derived cytokines. Although ILC2s were initially reported to play a key role in the anti-helminth innate immunity, we now have greater interest in their role in asthma and other allergic diseases. In various asthma mouse models, ILC2s provoke eosinophilic inflammation accompanied by airway hyperresponsiveness independent of acquired immunity. Moreover, recent mouse studies show that ILC2s also promote acquired immunity and Th2 polarization, and various cytokines and lipid mediators influence the functions of ILC2s. Although ILC2s have also been identified in humans, studies on the role of human ILC2s in asthma are very limited. Thus far, human studies have shown that there is a slight difference in responsiveness and production of cytokines between mouse and human ILC2s, and it has been suggested that ILC2s are involved in allergic-type asthma and the exacerbation of asthma. In this review, we focus on mouse and human ILC2s, and discuss their role in asthma.
Collapse
Affiliation(s)
- Hiroki Kabata
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan; Department of Internal Medicine, Kawasaki Municipal Hospital, Kanagawa, Japan; Laboratory for Immune Cell Systems, RIKEN Center for Integrative Medical Sciences (IMS), Kanagawa, Japan
| | - Kazuyo Moro
- Laboratory for Immune Cell Systems, RIKEN Center for Integrative Medical Sciences (IMS), Kanagawa, Japan; Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, Tokyo, Japan; Division of Immunobiology, Department of Medical Life Science, Graduate School of Medical Life Science, Yokohama City University, Kanagawa, Japan
| | - Shigeo Koyasu
- Laboratory for Immune Cell Systems, RIKEN Center for Integrative Medical Sciences (IMS), Kanagawa, Japan; Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Koichiro Asano
- Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, Kanagawa, Japan.
| |
Collapse
|