1
|
Rupani H, Busse WW, Howarth PH, Bardin PG, Adcock IM, Konno S, Jackson DJ. Therapeutic relevance of eosinophilic inflammation and airway viral interactions in severe asthma. Allergy 2024; 79:2589-2604. [PMID: 39087443 DOI: 10.1111/all.16242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 06/21/2024] [Accepted: 07/08/2024] [Indexed: 08/02/2024]
Abstract
The role of eosinophils in airway inflammation and asthma pathogenesis is well established, with raised eosinophil counts in blood and sputum associated with increased disease severity and risk of asthma exacerbation. Conversely, there is also preliminary evidence suggesting antiviral properties of eosinophils in the airways. These dual roles for eosinophils are particularly pertinent as respiratory virus infections contribute to asthma exacerbations. Biologic therapies targeting key molecules implicated in eosinophil-associated pathologies have been approved in patients with severe asthma and, therefore, the effects of depleting eosinophils in a clinical setting are of considerable interest. This review discusses the pathological and antiviral roles of eosinophils in asthma and exacerbations. We also highlight the significant reduction in asthma exacerbations seen with biologic therapies, even at the height of the respiratory virus season. Furthermore, we discuss the implications of these findings in relation to the role of eosinophils in inflammation and antiviral responses to respiratory virus infection in asthma.
Collapse
Affiliation(s)
- Hitasha Rupani
- Department of Respiratory Medicine, University Hospital Southampton NHS Foundation Trust, Southampton, Hampshire, UK
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, Hampshire, UK
| | - William W Busse
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Peter H Howarth
- Global Medical, Global Specialty and Primary Care, GSK, Brentford, Middlesex, UK
| | - Philip G Bardin
- Monash Lung Sleep Allergy and Immunology, Monash University and Medical Centre and Hudson Institute, Melbourne, Victoria, Australia
| | - Ian M Adcock
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Satoshi Konno
- Department of Respiratory Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - David J Jackson
- Guy's Severe Asthma Centre, Guy's and St Thomas' Hospitals, London, UK
- School of Immunology and Microbial Sciences, King's College London, London, UK
| |
Collapse
|
2
|
Noh SS, Shin HJ. Role of Virus-Induced EGFR Trafficking in Proviral Functions. Biomolecules 2023; 13:1766. [PMID: 38136637 PMCID: PMC10741569 DOI: 10.3390/biom13121766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 11/30/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Since its discovery in the early 1980s, the epidermal growth factor receptor (EGFR) has emerged as a pivotal and multifaceted player in elucidating the intricate mechanisms underlying various human diseases and their associations with cell survival, proliferation, and cellular homeostasis. Recent advancements in research have underscored the profound and multifaceted role of EGFR in viral infections, highlighting its involvement in viral entry, replication, and the subversion of host immune responses. In this regard, the importance of EGFR trafficking has also been highlighted in recent studies. The dynamic relocation of EGFR to diverse intracellular organelles, including endosomes, lysosomes, mitochondria, and even the nucleus, is a central feature of its functionality in diverse contexts. This dynamic intracellular trafficking is not merely a passive process but an orchestrated symphony, facilitating EGFR involvement in various cellular pathways and interactions with viral components. Furthermore, EGFR, which is initially anchored on the plasma membrane, serves as a linchpin orchestrating viral entry processes, a crucial early step in the viral life cycle. The role of EGFR in this context is highly context-dependent and varies among viruses. Here, we present a comprehensive summary of the current state of knowledge regarding the intricate interactions between EGFR and viruses. These interactions are fundamental for successful propagation of a wide array of viral species and affect viral pathogenesis and host responses. Understanding EGFR significance in both normal cellular processes and viral infections may not only help develop innovative antiviral therapies but also provide a deeper understanding of the intricate roles of EGFR signaling in infectious diseases.
Collapse
Affiliation(s)
- Se Sil Noh
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea;
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Hye Jin Shin
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea;
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon 35015, Republic of Korea
- Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| |
Collapse
|
3
|
Curren B, Ahmed T, Howard DR, Ashik Ullah M, Sebina I, Rashid RB, Al Amin Sikder M, Namubiru P, Bissell A, Ngo S, Jackson DJ, Toussaint M, Edwards MR, Johnston SL, McSorley HJ, Phipps S. IL-33-induced neutrophilic inflammation and NETosis underlie rhinovirus-triggered exacerbations of asthma. Mucosal Immunol 2023; 16:671-684. [PMID: 37506849 DOI: 10.1016/j.mucimm.2023.07.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 06/04/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023]
Abstract
Rhinovirus-induced neutrophil extracellular traps (NETs) contribute to acute asthma exacerbations; however, the molecular factors that trigger NETosis in this context remain ill-defined. Here, we sought to implicate a role for IL-33, an epithelial cell-derived alarmin rapidly released in response to infection. In mice with chronic experimental asthma (CEA), but not naïve controls, rhinovirus inoculation induced an early (1 day post infection; dpi) inflammatory response dominated by neutrophils, neutrophil-associated cytokines (IL-1α, IL-1β, CXCL1), and NETosis, followed by a later, type-2 inflammatory phase (3-7 dpi), characterised by eosinophils, elevated IL-4 levels, and goblet cell hyperplasia. Notably, both phases were ablated by HpARI (Heligmosomoides polygyrus Alarmin Release Inhibitor), which blocks IL-33 release and signalling. Instillation of exogenous IL-33 recapitulated the rhinovirus-induced early phase, including the increased presence of NETs in the airway mucosa, in a PAD4-dependent manner. Ex vivo IL-33-stimulated neutrophils from mice with CEA, but not naïve mice, underwent NETosis and produced greater amounts of IL-1α/β, IL-4, and IL-5. In nasal samples from rhinovirus-infected people with asthma, but not healthy controls, IL-33 levels correlated with neutrophil elastase and dsDNA. Our findings suggest that IL-33 blockade ameliorates the severity of an asthma exacerbation by attenuating neutrophil recruitment and the downstream generation of NETs.
Collapse
Affiliation(s)
- Bodie Curren
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; School of Biomedical Sciences, The University of Queensland, Queensland 4072, Australia
| | - Tufael Ahmed
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; School of Biomedical Sciences, Queensland University of Technology, Queensland 4000, Australia
| | - Daniel R Howard
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; School of Biomedical Sciences, The University of Queensland, Queensland 4072, Australia
| | - Md Ashik Ullah
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
| | - Ismail Sebina
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; School of Biomedical Sciences, The University of Queensland, Queensland 4072, Australia; School of Biomedical Sciences, Queensland University of Technology, Queensland 4000, Australia
| | - Ridwan B Rashid
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; School of Biomedical Sciences, The University of Queensland, Queensland 4072, Australia
| | - Md Al Amin Sikder
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; School of Biomedical Sciences, The University of Queensland, Queensland 4072, Australia
| | - Patricia Namubiru
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; School of Biomedical Sciences, The University of Queensland, Queensland 4072, Australia
| | - Alec Bissell
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
| | - Sylvia Ngo
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
| | - David J Jackson
- School of Immunology & Microbial Sciences, King's College London, London, UK; National Heart and Lung Institute, Imperial College London, London, UK
| | - Marie Toussaint
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Michael R Edwards
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Henry J McSorley
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Simon Phipps
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; School of Biomedical Sciences, The University of Queensland, Queensland 4072, Australia; School of Biomedical Sciences, Queensland University of Technology, Queensland 4000, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, 4072 Queensland, Australia.
| |
Collapse
|
4
|
Numata M, Sajuthi S, Bochkov YA, Loeffler J, Everman J, Vladar EK, Cooney RA, Reinhardt RL, Liu AH, Seibold MA, Voelker DR. Anionic Pulmonary Surfactant Lipid Treatment Inhibits Rhinovirus A Infection of the Human Airway Epithelium. Viruses 2023; 15:747. [PMID: 36992456 PMCID: PMC10055697 DOI: 10.3390/v15030747] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/16/2023] Open
Abstract
Rhinoviruses (RVs) are major instigators of acute exacerbations of asthma, COPD, and other respiratory diseases. RVs are categorized into three species (RV-A, RV-B, and RV-C), which comprise more than 160 serotypes, making it difficult to develop an effective vaccine. Currently, no effective treatment for RV infection is available. Pulmonary surfactant is an extracellular complex of lipids and proteins that plays a central role in regulating innate immunity in the lung. The minor pulmonary surfactant lipids, palmitoyl-oleoyl-phosphatidylglycerol (POPG) and phosphatidylinositol (PI), are potent regulators of inflammatory processes and exert antiviral activity against respiratory syncytial virus (RSV) and influenza A viruses (IAV). In the current study, we examined the potencies of POPG and PI against rhinovirus A16 (RV-A16) in primary human airway epithelial cells (AECs) differentiated at an air-liquid interface (ALI). After AECs were infected with RV-A16, PI reduced the viral RNA copy number by 70% and downregulated (55-75%) the expression of antiviral (MDA5, IRF7, and IFN-lambda) and CXCL11 chemokine genes. In contrast, POPG only slightly decreased MDA5 (24%) and IRF7 (11%) gene expression but did not inhibit IFN-lambda gene expression or RV-A16 replication in AECs. However, both POPG and PI inhibited (50-80%) IL6 gene expression and protein secretion and CXCL11 protein secretion. PI treatment dramatically attenuated global gene expression changes induced by RV-A16 infection alone in AECs. The observed inhibitory effects were indirect and resulted mainly from the inhibition of virus replication. Cell-type enrichment analysis of viral-regulated genes opposed by PI treatment revealed the PI-inhibited viral induction of goblet cell metaplasia and the virus-induced downregulation of ciliated, club, and ionocyte cell types. Notably, the PI treatment also altered the ability of RV-A16 to regulate the expression of some phosphatidylinositol 4-kinase (PI4K); acyl-CoA-binding, domain-containing (ACBD); and low-density lipoprotein receptor (LDLR) genes that play critical roles in the formation and functioning of replication organelles (ROs) required for RV replication in host cells. These data suggest PI can be used as a potent, non-toxic, antiviral agent for RV infection prophylaxis and treatment.
Collapse
Affiliation(s)
- Mari Numata
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA
| | - Satria Sajuthi
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO 80206, USA
| | - Yury A. Bochkov
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Jessica Loeffler
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA
| | - Jamie Everman
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO 80206, USA
| | - Eszter K. Vladar
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Riley A. Cooney
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Richard Lee Reinhardt
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO 80206, USA
| | - Andrew H. Liu
- Section of Pediatric Pulmonary & Sleep Medicine, Children’s Hospital Colorado and University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Max A. Seibold
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO 80206, USA
- Department of Pediatrics, National Jewish Health, Denver, CO 80206, USA
| | - Dennis R. Voelker
- Department of Medicine, National Jewish Health, Denver, CO 80206, USA
| |
Collapse
|
5
|
Wang W, Sinha A, Lutter R, Yang J, Ascoli C, Sterk PJ, Nemsick NK, Perkins DL, Finn PW. Analysis of Exosomal MicroRNA Dynamics in Response to Rhinovirus Challenge in a Longitudinal Case-Control Study of Asthma. Viruses 2022; 14:v14112444. [PMID: 36366542 PMCID: PMC9695046 DOI: 10.3390/v14112444] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/19/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022] Open
Abstract
Asthma symptoms are often exacerbated by the common-cold-causing rhinovirus (RV). In this study, we characterized the temporal behavior of circulating exosomal microRNAs (ExoMiRNAs) in a longitudinal bi-phasic case-control study of mild asthmatics (n = 12) and matched non-atopic healthy controls (n = 12) inoculated with rhinovirus. We aimed to define clinical and immunologic characteristics associated with differentially expressed (DE) miRNAs. In total, 26 DE ExoMiRNAs, including hsa-let-7f-5p, hsa-let-7a-5p, hsa-miR-122-5p, hsa-miR-101-3p, and hsa-miR-126-3p, were identified between asthmatic and healthy subjects after inoculation with RV. Time series clustering identified a unique Cluster of Upregulated DE ExoMiRNAs with augmenting mean expression and a distinct Cluster of Downregulated DE ExoMiRNAs with mean expression decline in asthmatic subjects upon RV challenge. Notably, the Upregulated Cluster correlated with Th1 and interferon-induced cytokines/chemokines (IFN-γ and IFN-γ-inducible protein-10) and interleukin-10 (IL-10). Conversely, the Downregulated Cluster correlated with IL-13, a Th2 cytokine, pulmonary function measurements (FVC%, FEV1%, and PEF%), and inflammatory biomarkers (FeNO, eosinophil%, and neutrophil%). Key ExoMiRNA-target gene and anti-viral defense mechanisms of the Upregulated and Downregulated Clusters were identified by network and gene enrichment analyses. Our findings provide insight into the regulatory role of ExoMiRNAs in RV-induced asthma.
Collapse
Affiliation(s)
- Wangfei Wang
- Richard and Loan Hill Department of Biomedical Engineering, College of Engineering and Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Anirban Sinha
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - René Lutter
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Experimental Immunology, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Jie Yang
- Department of Mathematics, Statistics, and Computer Science, College of Liberal Arts and Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Christian Ascoli
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Peter J. Sterk
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Nicole K. Nemsick
- Department of Molecular and Cellular Biology, College of Liberal Arts and Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - David L. Perkins
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Patricia W. Finn
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Correspondence:
| |
Collapse
|
6
|
Schwartz BS, Al-Sayouri SA, Pollak JS, Hirsch AG, Kern R, Tan B, Kato A, Schleimer RP, Peters AT. Strong and consistent associations of precedent chronic rhinosinusitis with risk of non-cystic fibrosis bronchiectasis. J Allergy Clin Immunol 2022; 150:701-708.e4. [PMID: 35314187 PMCID: PMC9463084 DOI: 10.1016/j.jaci.2022.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/20/2022] [Accepted: 03/03/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Chronic rhinosinusitis (CRS) and bronchiectasis commonly co-occur, but most prior studies were not designed to evaluate temporality and causality. OBJECTIVES In a sample representing the general population in 37 counties in Pennsylvania, and thus the full spectrum of sinonasal and relevant lung diseases, we aimed to evaluate the temporality and strength of associations of CRS with non-cystic fibrosis bronchiectasis. METHODS We completed case-control analyses for each of 3 primary bronchiectasis case finding methods. We used electronic health records to identify CRS and bronchiectasis with diagnoses, procedure orders, and/or specific text in sinus or chest computerized tomography scan radiology reports. The controls never had any indication of bronchiectasis and were frequency-matched to the 3 bronchiectasis groups on the basis of age, sex, and encounter year. There were 5,329 unique persons with bronchiectasis and 33,363 without bronchiectasis in the 3 analyses. Important co-occurring conditions were identified with diagnoses, medication orders, and encounter types. Logistic regression was used to evaluate associations (odds ratios [ORs] and 95% CIs) of CRS with bronchiectasis while adjusting for confounding variables. RESULTS In adjusted analyses, CRS was consistently and strongly associated with all 3 bronchiectasis definitions. The strongest associations for CRS (ORs and 95% CIs) were those that were based on the text of sinus computerized tomography scan reports; the associations were generally stronger for CRS without nasal polyps (eg, OR = 4.46 [95% CI = 2.09-9.51] for diagnosis-based bronchiectasis). On average, CRS was identified more than 6 years before bronchiectasis. CONCLUSION Precedent CRS was strongly and consistently associated with increased risk of bronchiectasis.
Collapse
Affiliation(s)
- Brian S Schwartz
- Department of Environmental Health and Engineering, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Md; Department of Population Health Sciences, Geisinger, Danville, Pa.
| | - Saba A Al-Sayouri
- Department of Environmental Health and Engineering, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Md
| | - Jonathan S Pollak
- Department of Environmental Health and Engineering, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Md
| | - Annemarie G Hirsch
- Department of Environmental Health and Engineering, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Md; Department of Population Health Sciences, Geisinger, Danville, Pa
| | - Robert Kern
- Department of Otolaryngology Head and Neck Surgery, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Bruce Tan
- Department of Otolaryngology Head and Neck Surgery, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Atsushi Kato
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Robert P Schleimer
- Department of Otolaryngology Head and Neck Surgery, Northwestern University Feinberg School of Medicine, Chicago, Ill; Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Ill
| | - Anju T Peters
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Ill
| |
Collapse
|
7
|
Gul A, Khan S, Arshad M, Anjum SI, Attaullah S, Ali I, Rauf A, Arshad A, Alghanem SM, Khan SN. Peripheral blood T cells response in human parainfluenza virus-associated lower respiratory tract infection in children. Saudi J Biol Sci 2020; 27:2847-2852. [PMID: 32994745 PMCID: PMC7499292 DOI: 10.1016/j.sjbs.2020.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/04/2020] [Accepted: 07/05/2020] [Indexed: 11/23/2022] Open
Abstract
Human Parainfluenza virus (HPIV) causes lower respiratory tract infections (LRTI) mostly in young children. Respiratory viral infections may decline T cells in circulation and display enhanced pathogenicity. This study is aimed to analyze T cells alterations due to HPIV in children with LRTIs. Children (N = 152) with bronchitis or pneumonia, admitted in tertiary care hospitals were included in the study. Respiratory samples (throat or nasopharyngeal swabs) were taken and HPIV genotypes (1-4) were analyzed through RT-PCR. Peripheral blood T cells, CD3+, CD4+, CD8+, and CD19+, were analyzed in confirmed HPIV positive and healthy control group children through flow cytometry. The positivity rate of HPIV was 24.34% and the most prevalent genotype was HPIV-3 (20.40%). HPIV-1 and HPIV-2 were detected in 0.66% and 02% children respectively. The T lymphocyte counts were observed significantly reduced in children infected with HPIV-3. CD4+ cell (1580 ± 97.87) counts did not change significantly but the lowest CD8+ T cell counts (518.5 ± 74.00) were recorded. Similarly, CD3+ and CD19 cell ratios were also reduced. The CD4/CD8 ratio was significantly higher (3.12 ± 0.59) in the study population as compared to the control group (2.18 ± 0.654). Changes in the count of CD8+ T cells were more pronounced in patients with bronchiolitis and pneumonia. It is concluded that CD8+ T cells show a reduced response to HPIV-3 in children with severe LRTIs suggesting a strong association of these cells with disease severity.
Collapse
Affiliation(s)
- Aisha Gul
- Department of Zoology, University of Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Sanaullah Khan
- Department of Zoology, University of Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Muhammad Arshad
- Department of Biological Sciences, International Islamic University, Islamabad Pakistan
| | - Syed Ishtiaq Anjum
- Department of Zoology Kohat University of Science & Technology, Kohat, Pakistan
| | - Sobia Attaullah
- Department of Zoology, Islamia College Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Ijaz Ali
- Department of Biosciences, COMSATS University, Islamabad, Pakistan
| | - Abdur Rauf
- Department of Zoology, University of Azad Jammu and Kashmir, Muzaffarabad, Pakistan
| | - Abida Arshad
- Department of Zoology, PMAS-Arid Agriculture University, Rawalpindi, Pakistan
| | - Suliman M. Alghanem
- Biology Department, Faculty of Science, Tabuk University, Tabuk 71491, Saudi Arabia
| | - Shahid Niaz Khan
- Department of Zoology Kohat University of Science & Technology, Kohat, Pakistan
| |
Collapse
|
8
|
Sabogal Piñeros YS, Dekker T, Smids B, Majoor CJ, Ravanetti L, Villetti G, Civelli M, Facchinetti F, Lutter R. Phosphodiesterase 4 inhibitors attenuate virus-induced activation of eosinophils from asthmatics without affecting virus binding. Pharmacol Res Perspect 2020; 8:e00557. [PMID: 32447834 PMCID: PMC7245579 DOI: 10.1002/prp2.557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 12/02/2019] [Accepted: 12/06/2019] [Indexed: 11/11/2022] Open
Abstract
Acute respiratory virus infections, such as influenza and RSV, are predominant causes of asthma exacerbations. Eosinophils act as a double-edged sword in exacerbations in that they are activated by viral infections but also can capture and inactivate respiratory viruses. Phosphodiesterase type 4 (PDE4) is abundantly expressed by eosinophils and has been implicated in their activation. This exploratory study aims to determine whether these opposing roles of eosinophils activation of eosinophils upon interaction with virus can be modulated by selective PDE4 inhibitors and whether eosinophils from healthy, moderate and severe asthmatic subjects respond differently. Eosinophils were purified by negative selection from blood and subsequently exposed to RSV or influenza. Prior to exposure to virus, eosinophils were treated with vehicle or selective PDE4 inhibitors CHF6001 and GSK256066. After 18 hours of exposure, influenza, but not RSV, increased CD69 and CD63 expression by eosinophils from each group, which were inhibited by PDE4 inhibitors. ECP release, although not stimulated by virus, was also attenuated by PDE4 inhibitors. Eosinophils showed an increased Nox2 activity upon virus exposure, which was less pronounced in eosinophils derived from mild and severe asthmatics and was counteracted by PDE4 inhibitors. PDE4 inhibitors had no effect on binding of virus by eosinophils from each group. Our data indicate that PDE4 inhibitors can attenuate eosinophil activation, without affecting virus binding. By attenuating virus-induced responses, PDE4 inhibitors may mitigate virus-induced asthma exacerbations.
Collapse
Affiliation(s)
- Yanaika Shari Sabogal Piñeros
- Department of Experimental ImmunologyAmsterdam Infection & Immunity InstituteAmsterdamThe Netherlands
- Department of Respiratory MedicineAmsterdam University Medical CentresUniversity of AmsterdamAmsterdamThe Netherlands
| | - Tamara Dekker
- Department of Experimental ImmunologyAmsterdam Infection & Immunity InstituteAmsterdamThe Netherlands
- Department of Respiratory MedicineAmsterdam University Medical CentresUniversity of AmsterdamAmsterdamThe Netherlands
| | - Barbara Smids
- Department of Experimental ImmunologyAmsterdam Infection & Immunity InstituteAmsterdamThe Netherlands
- Department of Respiratory MedicineAmsterdam University Medical CentresUniversity of AmsterdamAmsterdamThe Netherlands
| | - Christof J. Majoor
- Department of Respiratory MedicineAmsterdam University Medical CentresUniversity of AmsterdamAmsterdamThe Netherlands
| | - Lara Ravanetti
- Department of Experimental ImmunologyAmsterdam Infection & Immunity InstituteAmsterdamThe Netherlands
- Department of Respiratory MedicineAmsterdam University Medical CentresUniversity of AmsterdamAmsterdamThe Netherlands
| | - Gino Villetti
- Corporate Pre‐Clinical R&DChiesi Farmaceutici S.p.A.ParmaItaly
| | | | | | - René Lutter
- Department of Experimental ImmunologyAmsterdam Infection & Immunity InstituteAmsterdamThe Netherlands
- Department of Respiratory MedicineAmsterdam University Medical CentresUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
9
|
Stolz D, Papakonstantinou E, Grize L, Schilter D, Strobel W, Louis R, Schindler C, Hirsch HH, Tamm M. Time-course of upper respiratory tract viral infection and COPD exacerbation. Eur Respir J 2019; 54:13993003.00407-2019. [PMID: 31391222 DOI: 10.1183/13993003.00407-2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 06/28/2019] [Indexed: 11/05/2022]
Abstract
Viral respiratory tract infections have been implicated as the predominant risk factor for acute exacerbations of chronic obstructive pulmonary disease (AECOPD). We aimed to evaluate, longitudinally, the association between upper respiratory tract infections (URTI) caused by viruses and AECOPD.Detection of 18 viruses was performed in naso- and orοpharyngeal swabs from 450 COPD patients (Global Initiative for Chronic Obstructive Lung Disease stages 2-4) who were followed for a mean of 27 months. Swabs were taken during stable periods (n=1909), at URTI onset (n=391), 10 days after the URTI (n=356) and during an AECOPD (n=177) and tested using a multiplex nucleic acid amplification test.Evidence of at least one respiratory virus was significantly higher at URTI onset (52.7%), 10 days after the URTI (15.2%) and during an AECOPD (38.4%), compared with the stable period (5.3%, p<0.001). During stable visits, rhinovirus accounted for 54.2% of all viral infections, followed by coronavirus (20.5%). None of the viruses were identified in two consecutive stable visits. Patients with a viral infection at URTI onset did not have a higher incidence of exacerbation than patients without viral infection (p=0.993). Τhe incidence of any viral infection during an AECOPD was similar between URTI-related AECOPD and non-URTI-related AECOPD (p=0.359). Only 24% of the patients that had a URTI-related AECOPD had the same virus at URTI onset and during an AECOPD. Detection of parainfluenza 3 at URTI onset was associated with a higher risk of an AECOPD (p=0.003). Rhinovirus and coronavirus were the most frequently detected viruses during AECOPD visits, accounting for 35.7% and 25.9% of all viral infections, respectively.The prevalence of viral infection during the stable period of COPD was low. The risk of exacerbation following the onset of URTI symptoms depends on the particular virus associated with the event and was significant only for parainfluenza 3.
Collapse
Affiliation(s)
- Daiana Stolz
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital Basel, University of Basel, Basel, Switzerland .,Dept of Biomedicine, University of Basel, Basel, Switzerland
| | - Eleni Papakonstantinou
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital Basel, University of Basel, Basel, Switzerland.,Dept of Biomedicine, University of Basel, Basel, Switzerland
| | - Leticia Grize
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute, Basel, Switzerland
| | | | - Werner Strobel
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital Basel, University of Basel, Basel, Switzerland.,Dept of Biomedicine, University of Basel, Basel, Switzerland
| | - Renaud Louis
- Pneumology Dept, University of Liege, CHU Liege, Liege, Belgium
| | - Christian Schindler
- University of Basel, Basel, Switzerland.,Swiss Tropical and Public Health Institute, Basel, Switzerland
| | - Hans H Hirsch
- Dept of Biomedicine, University of Basel, Basel, Switzerland.,Both authors contributed equally
| | - Michael Tamm
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital Basel, University of Basel, Basel, Switzerland.,Dept of Biomedicine, University of Basel, Basel, Switzerland.,Both authors contributed equally
| |
Collapse
|
10
|
Abstract
Respiratory viral infections including human rhinovirus (RV) infection have been identified as the most important environmental trigger of exacerbations of chronic lung diseases. While well established as the most common viral infections associated with exacerbations of asthma and chronic obstructive pulmonary disease, RVs and other respiratory viruses are also now thought to be important in triggering exacerbations of cystic fibrosis and the interstitial lung diseases. Here, we summarize the epidemiological evidence the supports respiratory viruses including RV as triggers of exacerbations of chronic lung diseases. We propose that certain characteristics of RVs may explain why they are the most common trigger of exacerbations of chronic lung diseases. We further highlight the latest mechanistic evidence supporting how and why common respiratory viral infections may enhance and promote disease triggering exacerbation events, through their interactions with the host immune system, and may be affected by ongoing treatments. We also provide a commentary on how new treatments may better manage the disease burden associated with respiratory viral infections and the exacerbation events that they trigger.
Collapse
|
11
|
Girkin J, Maltby S, Singanayagam A, Bartlett N, Mallia P. In vivo experimental models of infection and disease. RHINOVIRUS INFECTIONS 2019. [PMCID: PMC7149593 DOI: 10.1016/b978-0-12-816417-4.00008-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Human and animal models continue to play a crucial role in research to understand host immunity to rhinovirus (RV) and identify disease mechanisms. Human models have provided direct evidence that RV infection is capable of exacerbating chronic respiratory diseases and identified immunological processes that correlate with clinical disease outcomes. Mice are the most commonly used nonhuman experimental RV infection model. Although semipermissive, under defined experimental conditions sufficient replication occurs to induce host immune responses that recapitulate immunity and disease during human infection. The capacity to use genetically modified mouse strains and drug interventions has shown the mouse model to be an invaluable research tool defining causal relationships between host immunity and disease and supporting development of new treatments. Used in combination the insights achieved from human and animal experimental infection models provide complementary insights into RV biology and yield novel therapeutic options to reduce the burden of RV-induced disease.
Collapse
|
12
|
Ağaç D, Gill MA, Farrar JD. Adrenergic Signaling at the Interface of Allergic Asthma and Viral Infections. Front Immunol 2018; 9:736. [PMID: 29696025 PMCID: PMC5904268 DOI: 10.3389/fimmu.2018.00736] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 03/26/2018] [Indexed: 12/16/2022] Open
Abstract
Upper respiratory viral infections are a major etiologic instigator of allergic asthma, and they drive severe exacerbations of allergic inflammation in the lower airways of asthma sufferers. Rhinovirus (RV), in particular, is the main viral instigator of these pathologies. Asthma exacerbations due to RV infections are the most frequent reasons for hospitalization and account for the majority of morbidity and mortality in asthma patients. In both critical care and disease control, long- and short-acting β2-agonists are the first line of therapeutic intervention, which are used to restore airway function by promoting smooth muscle cell relaxation in bronchioles. While prophylactic use of β2-agonists reduces the frequency and pathology of exacerbations, their role in modulating the inflammatory response is only now being appreciated. Adrenergic signaling is a component of the sympathetic nervous system, and the natural ligands, epinephrine and norepinephrine (NE), regulate a multitude of autonomic functions including regulation of both the innate and adaptive immune response. NE is the primary neurotransmitter released by post-ganglionic sympathetic neurons that innervate most all peripheral tissues including lung and secondary lymphoid organs. Thus, the adrenergic signaling pathways are in direct contact with both the central and peripheral immune compartments. We present a perspective on how the adrenergic signaling pathway controls immune function and how β2-agonists may influence inflammation in the context of virus-induced asthma exacerbations.
Collapse
Affiliation(s)
- Didem Ağaç
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Michelle A Gill
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - J David Farrar
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
13
|
Looi K, Buckley AG, Rigby PJ, Garratt LW, Iosifidis T, Zosky GR, Larcombe AN, Lannigan FJ, Ling KM, Martinovich KM, Kicic-Starcevich E, Shaw NC, Sutanto EN, Knight DA, Kicic A, Stick SM. Effects of human rhinovirus on epithelial barrier integrity and function in children with asthma. Clin Exp Allergy 2018; 48:513-524. [PMID: 29350877 DOI: 10.1111/cea.13097] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/13/2017] [Accepted: 11/21/2017] [Indexed: 11/29/2022]
Abstract
BACKGROUND Bronchial epithelial tight junctions (TJ) have been extensively assessed in healthy airway epithelium. However, no studies have yet assessed the effect of human rhinovirus (HRV) infection on the expression and resultant barrier function in epithelial tight junctions (TJ) in childhood asthma. OBJECTIVES To investigate the impact of HRV infection on airway epithelial TJ expression and barrier function in airway epithelial cells (AECs) of children with and without asthma. Furthermore, to test the hypothesis that barrier integrity and function is compromised to a greater extent by HRV in AECs from asthmatic children. METHODS Primary AECs were obtained from children with and without asthma, differentiated into air-liquid interface (ALI) cultures and infected with rhinovirus. Expression of claudin-1, occludin and zonula occluden-1 (ZO-1) was assessed via qPCR, immunocytochemistry (ICC), in-cell western (ICW) and confocal microscopy. Barrier function was assessed by transepithelial electrical resistance (TER; RT ) and permeability to fluorescent dextran. RESULTS Basal TJ gene expression of claudin-1 and occludin was significantly upregulated in asthmatic children compared to non-asthmatics; however, no difference was seen with ZO-1. Interestingly, claudin-1, occludin and ZO-1 protein expression was significantly reduced in AEC of asthmatic children compared to non-asthmatic controls suggesting possible post-transcriptional inherent differences. HRV infection resulted in a transient dissociation of TJ and airway barrier integrity in non-asthmatic children. Although similar dissociation of TJ was observed in asthmatic children, a significant and sustained reduction in TJ expression concurrent with both a significant decrease in TER and an increase in permeability in asthmatic children was observed. CONCLUSION This study demonstrates novel intrinsic differences in TJ gene and protein expression between AEC of children with and without asthma. Furthermore, it correlates directly the relationship between HRV infection and the resultant dissociation of epithelial TJ that causes a continued altered barrier function in children with asthma.
Collapse
Affiliation(s)
- K Looi
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - A G Buckley
- Centre for Microscopy, Characterisation and Analysis (CMCA), University of Western Australia, Crawley, WA, Australia
| | - P J Rigby
- Centre for Microscopy, Characterisation and Analysis (CMCA), University of Western Australia, Crawley, WA, Australia
| | - L W Garratt
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - T Iosifidis
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Nedlands, WA, Australia
| | - G R Zosky
- School of Medicine, Faculty of Health, University of Tasmania, Hohart, TAS, Australia
| | - A N Larcombe
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Occupation and Environment, School of Public Health, Curtin University, Perth, WA, Australia
| | - F J Lannigan
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,School of Medicine, Notre Dame University, Fremantle, WA, Australia
| | - K-M Ling
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - K M Martinovich
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - E Kicic-Starcevich
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia
| | - N C Shaw
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - E N Sutanto
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia
| | - D A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Priority Research Centre for Asthma and Respiratory Disease, Hunter Medical Research Institute, Newcastle, NSW, Australia.,Department of Anaesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - A Kicic
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Nedlands, WA, Australia.,Occupation and Environment, School of Public Health, Curtin University, Perth, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia
| | - S M Stick
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Nedlands, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia
| |
Collapse
|
14
|
Nixon J, Newbold P, Mustelin T, Anderson GP, Kolbeck R. Monoclonal antibody therapy for the treatment of asthma and chronic obstructive pulmonary disease with eosinophilic inflammation. Pharmacol Ther 2016; 169:57-77. [PMID: 27773786 DOI: 10.1016/j.pharmthera.2016.10.016] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Eosinophils have been linked with asthma for more than a century, but their role has been unclear. This review discusses the roles of eosinophils in asthma and chronic obstructive pulmonary disease (COPD) and describes therapeutic antibodies that affect eosinophilia. The aims of pharmacologic treatments for pulmonary conditions are to reduce symptoms, slow decline or improve lung function, and reduce the frequency and severity of exacerbations. Inhaled corticosteroids (ICS) are important in managing symptoms and exacerbations in asthma and COPD. However, control with these agents is often suboptimal, especially for patients with severe disease. Recently, new biologics that target eosinophilic inflammation, used as adjunctive therapy to corticosteroids, have proven beneficial and support a pivotal role for eosinophils in the pathology of asthma. Nucala® (mepolizumab; anti-interleukin [IL]-5) and Cinquair® (reslizumab; anti-IL-5), the second and third biologics approved, respectively, for the treatment of asthma, exemplifies these new treatment options. Emerging evidence suggests that eosinophils may contribute to exacerbations and possibly to lung function decline for a subset of patients with COPD. Here we describe the pharmacology of therapeutic antibodies inhibiting IL-5 or targeting the IL-5 receptor, as well as other cytokines contributing to eosinophilic inflammation. We discuss their roles as adjuncts to conventional therapeutic approaches, especially ICS therapy, when disease is suboptimally controlled. These agents have achieved a place in the therapeutic armamentarium for asthma and COPD and will deepen our understanding of the pathogenic role of eosinophils.
Collapse
Affiliation(s)
| | | | | | - Gary P Anderson
- Lung Health Research Centre, University of Melbourne, Melbourne, Victoria, Australia
| | | |
Collapse
|
15
|
Muehling LM, Mai DT, Kwok WW, Heymann PW, Pomés A, Woodfolk JA. Circulating Memory CD4+ T Cells Target Conserved Epitopes of Rhinovirus Capsid Proteins and Respond Rapidly to Experimental Infection in Humans. THE JOURNAL OF IMMUNOLOGY 2016; 197:3214-3224. [PMID: 27591323 DOI: 10.4049/jimmunol.1600663] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 08/09/2016] [Indexed: 01/15/2023]
Abstract
Rhinovirus (RV) is a major cause of common cold and an important trigger of acute episodes of chronic lung diseases. Antigenic variation across the numerous RV strains results in frequent infections and a lack of durable cross-protection. Because the nature of human CD4+ T cells that target RV is largely unknown, T cell epitopes of RV capsid proteins were analyzed, and cognate T cells were characterized in healthy subjects and those infected by intranasal challenge. Peptide epitopes of the RV-A16 capsid proteins VP1 and VP2 were identified by peptide/MHC class II tetramer-guided epitope mapping, validated by direct ex vivo enumeration, and interrogated using a variety of in silico methods. Among noninfected subjects, those circulating RV-A16-specific CD4+ T cells detected at the highest frequencies targeted 10 unique epitopes that bound to diverse HLA-DR molecules. T cell epitopes localized to conserved molecular regions of biological significance to the virus were enriched for HLA class I and II binding motifs, and constituted both species-specific (RV-A) and pan-species (RV-A, -B, and -C) varieties. Circulating epitope-specific T cells comprised both memory Th1 and T follicular helper cells, and were rapidly expanded and activated after intranasal challenge with RV-A16. Cross-reactivity was evidenced by identification of a common *0401-restricted epitope for RV-A16 and RV-A39 by tetramer-guided epitope mapping and the ability for RV-A16-specific Th1 cells to proliferate in response to their RV-A39 peptide counterpart. The preferential persistence of high-frequency RV-specific memory Th1 cells that recognize a limited set of conserved epitopes likely arises from iterative priming by previous exposures to different RV strains.
Collapse
Affiliation(s)
- Lyndsey M Muehling
- Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908
| | - Duy T Mai
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101
| | - William W Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, WA 98101
| | - Peter W Heymann
- Department of Pediatrics, University of Virginia Health System, Charlottesville, VA 22908; and
| | - Anna Pomés
- Indoor Biotechnologies Inc., Charlottesville, VA 22903
| | - Judith A Woodfolk
- Department of Medicine, University of Virginia Health System, Charlottesville, VA 22908;
| |
Collapse
|
16
|
Kobayashi T, Soma T, Noguchi T, Nakagome K, Nakamoto H, Kita H, Nagata M. ATP drives eosinophil effector responses through P2 purinergic receptors. Allergol Int 2015; 64 Suppl:S30-6. [PMID: 26344078 DOI: 10.1016/j.alit.2015.04.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/11/2015] [Accepted: 04/20/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Eosinophils recognize various stimuli, such as cytokines, chemokines, immunoglobulins, complement, and external pathogens, resulting in their accumulation in mucosal tissues and the progression of inflammation. Eosinophils are also involved in innate Th2-type immune responses mediated through endogenous danger signals, including IL-33, uric acid (UA), or ATP, in non-sensitized mice exposed to environmental allergens. However, the mechanism involved in eosinophil responses to these danger signals remains insufficiently understood. METHODS We examined migration, adhesion, superoxide production and degranulation of human eosinophils. Isolated eosinophils were incubated with monosodium urate (MSU) crystals and ATPγS, a non-hydrolysable ATP analogue. To determine the involvement of P2 or P2Y2 receptors in eosinophil responses to UA and ATP, eosinophils were preincubated with a pan-P2 receptor inhibitor, oxidized ATP (oATP), or anti-P2Y2 antibody before incubation with MSU crystals or ATPγS. RESULTS MSU crystals induced adhesion of eosinophils to recombinant human (rh)-ICAM-1 and induced production of superoxide. oATP abolished eosinophil responses to MSU crystals, suggesting involvement of endogenous ATP and its receptors. Furthermore, exogenous ATP, as ATPγS, induced migration of eosinophils through a model basement membrane, adhesion to rh-ICAM-1, superoxide generation, and degranulation of eosinophil-derived neurotoxin (EDN). oATP and anti-P2Y2 significantly reduced these eosinophil responses. CONCLUSIONS ATP serves as an essential mediator of functional responses in human eosinophils. Eosinophil responses to ATP may be implicated in airway inflammation in patients with asthma.
Collapse
|
17
|
Rohde G, Message SD, Haas JJ, Kebadze T, Parker H, Laza-Stanca V, Khaitov MR, Kon OM, Stanciu LA, Mallia P, Edwards MR, Johnston SL. CXC chemokines and antimicrobial peptides in rhinovirus-induced experimental asthma exacerbations. Clin Exp Allergy 2015; 44:930-9. [PMID: 24673807 PMCID: PMC4403958 DOI: 10.1111/cea.12313] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 01/28/2014] [Accepted: 02/10/2014] [Indexed: 12/28/2022]
Abstract
RATIONALE Rhinoviruses (RVs) are the major triggers of asthma exacerbations. We have shown previously that lower respiratory tract symptoms, airflow obstruction, and neutrophilic airway inflammation were increased in experimental RV-induced asthma exacerbations. OBJECTIVES We hypothesized that neutrophil-related CXC chemokines and antimicrobial peptides are increased and related to clinical, virologic, and pathologic outcomes in RV-induced exacerbations of asthma. METHODS Protein levels of antimicrobial peptides (SLPI, HNP 1-3, elafin, and LL-37) and neutrophil chemokines (CXCL1/GRO-α, CXCL2/GRO-β, CXCL5/ENA-78, CXCL6/GCP-2, CXCL7/NAP-2, and CXCL8/IL-8) were determined in bronchoalveolar lavage (BAL) fluid of 10 asthmatics and 15 normal controls taken before, at day four during and 6 weeks post-experimental infection. RESULTS BAL HNP 1-3 and Elafin were higher, CXCL7/NAP-2 was lower in asthmatics compared with controls at day 4 (P = 0.035, P = 0.048, and P = 0.025, respectively). BAL HNP 1-3 and CXCL8/IL-8 were increased during infection (P = 0.003 and P = 0.011, respectively). There was a trend to increased BAL neutrophils at day 4 compared with baseline (P = 0.076). BAL HNP 1-3 was positively correlated with BAL neutrophil numbers at day 4. There were no correlations between clinical parameters and HNP1-3 or IL-8 levels. CONCLUSIONS We propose that RV infection in asthma leads to increased release of CXCL8/IL-8, attracting neutrophils into the airways where they release HNP 1-3, which further enhances airway neutrophilia. Strategies to inhibit CXCL8/IL-8 may be useful in treatment of virus-induced asthma exacerbations.
Collapse
Affiliation(s)
- G Rohde
- Department of Respiratory Medicine, National Heart and Lung Institute, MRC and Asthma UK Centre in Allergic Mechanisms of Asthma & Centre for Respiratory Infection, Imperial College London, London, UK; Department of Respiratory Medicine, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Del Vecchio AM, Branigan PJ, Barnathan ES, Flavin SK, Silkoff PE, Turner RB. Utility of animal and in vivo experimental infection of humans with rhinoviruses in the development of therapeutic agents for viral exacerbations of asthma and chronic obstructive pulmonary disease. Pulm Pharmacol Ther 2015; 30:32-43. [PMID: 25445932 PMCID: PMC7110859 DOI: 10.1016/j.pupt.2014.10.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 10/24/2014] [Accepted: 10/29/2014] [Indexed: 12/16/2022]
Abstract
There is an association with acute viral infection of the respiratory tract and exacerbations of asthma and chronic obstructive pulmonary disease (COPD). Although these exacerbations are associated with several types of viruses, human rhinoviruses (HRVs) are associated with the vast majority of disease exacerbations. Due to the lack of an animal species that is naturally permissive for HRVs to use as a facile model system, and the limitations associated with animal models of asthma and COPD, studies of controlled experimental infection of humans with HRVs have been used and conducted safely for decades. This review discusses how these experimental infection studies with HRVs have provided a means of understanding the pathophysiology underlying virus-induced exacerbations of asthma and COPD with the goal of developing agents for their prevention and treatment.
Collapse
Affiliation(s)
- Alfred M Del Vecchio
- Janssen Research and Development, Immunology Clinical Research and Development, Welsh and McKean Roads, Spring House, PA 19477, USA
| | - Patrick J Branigan
- Janssen Research and Development, Immunology Clinical Research and Development, Welsh and McKean Roads, Spring House, PA 19477, USA
| | - Elliot S Barnathan
- Janssen Research and Development, Immunology Clinical Research and Development, Welsh and McKean Roads, Spring House, PA 19477, USA
| | - Susan K Flavin
- Janssen Research and Development, Immunology Clinical Research and Development, Welsh and McKean Roads, Spring House, PA 19477, USA
| | - Philip E Silkoff
- Janssen Research and Development, Immunology Clinical Research and Development, Welsh and McKean Roads, Spring House, PA 19477, USA.
| | - Ronald B Turner
- University of Virginia, School of Medicine, Charlottesville, VA 22908, USA
| |
Collapse
|
19
|
Rhinovirus-induced VP1-specific Antibodies are Group-specific and Associated With Severity of Respiratory Symptoms. EBioMedicine 2014; 2:64-70. [PMID: 26137535 PMCID: PMC4484518 DOI: 10.1016/j.ebiom.2014.11.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 11/14/2014] [Accepted: 11/17/2014] [Indexed: 01/07/2023] Open
Abstract
Background Rhinoviruses (RVs) are a major cause of common colds and induce exacerbations of asthma and chronic inflammatory lung diseases. Methods We expressed and purified recombinant RV coat proteins VP1-4, non-structural proteins as well as N-terminal fragments of VP1 from four RV strains (RV14, 16, 89, C) covering the three known RV groups (RV-A, RV-B and RV-C) and measured specific IgG-subclass-, IgA- and IgM-responses by ELISA in subjects with different severities of asthma or without asthma before and after experimental infection with RV16. Findings Before infection subjects showed IgG1 > IgA > IgM > IgG3 cross-reactivity with N-terminal fragments from the representative VP1 proteins of the three RV groups. Antibody levels were higher in the asthmatic group as compared to the non-asthmatic subjects. Six weeks after infection with RV16, IgG1 antibodies showed a group-specific increase towards the N-terminal VP1 fragment, but not towards other capsid and non-structural proteins, which was highest in subjects with severe upper and lower respiratory symptoms. Interpretation Our results demonstrate that increases of antibodies towards the VP1 N-terminus are group-specific and associated with severity of respiratory symptoms and suggest that it may be possible to develop serological tests for identifying causative RV groups. Increases of rhinovirus-specific antibodies are surrogate markers for severity of rhinovirus-induced respiratory symptoms. Serological tests based on recombinant rhinovirus coat protein fragments identify the culprit rhinovirus strain. Identification of the most relevant RV strains by serology should allow the rational design of RV vaccines.
Collapse
Key Words
- Antibody response
- Asthma
- COPD, Chronic obstructive pulmonary disease
- ELISA, Enzyme-linked immunosorbent assay
- HRP, Horseradish peroxidase
- HSA, Human serum albumin
- ICAM-1, Intercellular adhesion molecule 1
- ICS, Inhaled corticosteroids
- LDL-R, Low density lipoprotein receptor
- MALDI–TOF, Matrix-assisted laser desorption/ionization–time-of-flight mass spectrometry
- MBP, Maltose binding protein
- O.D, Optical density
- PEF, Peak expiratory flow
- RV, Rhinovirus
- Recombinant rhinovirus coat protein
- Rhinovirus
- SABA, Short-acting β2 agonists
- Serological test
- TCID50, Tissue culture 50% infective dose
Collapse
|
20
|
Oliver BGG, Robinson P, Peters M, Black J. Viral infections and asthma: an inflammatory interface? Eur Respir J 2014; 44:1666-81. [PMID: 25234802 DOI: 10.1183/09031936.00047714] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Asthma is a chronic inflammatory disease of the airways in which the majority of patients respond to treatment with corticosteroids and β₂-adrenoceptor agonists. Acute exacerbations of asthma substantially contribute to disease morbidity, mortality and healthcare costs, and are not restricted to patients who are not compliant with their treatment regimens. Given that respiratory viral infections are the principal cause of asthma exacerbations, this review article will explore the relationship between viral infections and asthma, and will put forward hypotheses as to why virus-induced exacerbations occur. Potential mechanisms that may explain why current therapeutics do not fully inhibit virus-induced exacerbations, for example, β₂-adrenergic desensitisation and corticosteroid insensitivity, are explored, as well as which aspects of virus-induced inflammation are likely to be attenuated by current therapy.
Collapse
Affiliation(s)
- Brian G G Oliver
- School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, Australia Woolcock Institute of Medical Research, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Paul Robinson
- Woolcock Institute of Medical Research, Sydney Medical School, The University of Sydney, Sydney, Australia Dept of Respiratory Medicine, The Children's Hospital at Westmead, Sydney, Australia The Children's Hospital at Westmead Clinical School, The University of Sydney, Sydney, Australia
| | - Mathew Peters
- Australian School of Advanced Medicine, Macquarie University, Sydney, Australia Dept of Thoracic Medicine, Concord General Hospital, Concord, Australia
| | - Judy Black
- Woolcock Institute of Medical Research, Sydney Medical School, The University of Sydney, Sydney, Australia
| |
Collapse
|
21
|
Parker LC, Stokes CA, Sabroe I. Rhinoviral infection and asthma: the detection and management of rhinoviruses by airway epithelial cells. Clin Exp Allergy 2014; 44:20-8. [PMID: 24355017 DOI: 10.1111/cea.12182] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 07/02/2013] [Accepted: 07/22/2013] [Indexed: 11/28/2022]
Abstract
Human rhinoviruses (HRV) have been linked to the development of childhood asthma and recurrent acute asthma exacerbations throughout life, and contribute considerably to the healthcare and economic burden of this disease. However, the ability of HRV infections to trigger exacerbations, and the link between allergic status and HRV responsiveness, remains incompletely understood. Whilst the receptors on human airway cells that detect and are utilized by most HRV group A and B, but not C serotypes are known, how endosomal pattern recognition receptors (PRRs) detect HRV replication products that are generated within the cytoplasm remains somewhat of an enigma. In this article, we explore a role for autophagy, a cellular homeostatic process that allows the cell to encapsulate its own cytosolic constituents, as the crucial mechanism controlling this process and regulating the innate immune response of airway epithelial cells to viral infection. We will also briefly describe some of the recent insights into the immune responses of the airway to HRV, focusing on neutrophilic inflammation that is a potentially unwanted feature of the acute response to viral infection, and the roles of IL-1 and Pellinos in the regulation of responses to HRV.
Collapse
Affiliation(s)
- L C Parker
- Academic Unit of Respiratory Medicine, Department of Infection and Immunity, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, UK
| | | | | |
Collapse
|
22
|
Kwon JM, Shim JW, Kim DS, Jung HL, Park MS, Shim JY. Prevalence of respiratory viral infection in children hospitalized for acute lower respiratory tract diseases, and association of rhinovirus and influenza virus with asthma exacerbations. KOREAN JOURNAL OF PEDIATRICS 2014; 57:29-34. [PMID: 24578714 PMCID: PMC3935110 DOI: 10.3345/kjp.2014.57.1.29] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Revised: 02/19/2013] [Accepted: 04/14/2013] [Indexed: 11/27/2022]
Abstract
PURPOSE In this study, we aimed to investigate the prevalence of year-round respiratory viral infection in children with lower respiratory tract infection (LRTI) and the relationship between respiratory viral infection and allergen sensitization in exacerbating asthma. METHODS We investigated the sources for acute LRTIs in children admitted to our hospital from May 2010 to April 2011. The 6 most common respiratory viruses were isolated from nasopharyngeal aspirate using multiplex reverse transcription-polymerase chain reaction in 309 children; respiratory syncytial virus (RSV), adenovirus (AV), parainfluenza virus (PIV), influenza virus (IFV), human metapneumovirus (hMPV), rhinovirus (RV). Atopic sensitization was defined if more than 1 serum specific Immunoglobulin E level measured using UniCAP (Pharmacia) was over 0.35 IU/mL. RESULTS RSV was the most common pathogen of bronchiolitis in hospitalized children through the year. RV or IFV infection was more prevalent in asthma exacerbations compared to other LRTIs. AV and hMPV were more likely to cause pneumonia. RV and IFV were associated with asthma exacerbations in children with atopic sensitization, but not in nonatopic children. CONCLUSION RV and IFV are associated with hospitalization for asthma exacerbation in children with atopic sensitization.
Collapse
Affiliation(s)
- Jang-Mi Kwon
- Department of Pediatrics, Kangbuk Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jae Won Shim
- Department of Pediatrics, Kangbuk Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Deok Soo Kim
- Department of Pediatrics, Kangbuk Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hye Lim Jung
- Department of Pediatrics, Kangbuk Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Moon Soo Park
- Department of Pediatrics, Kangbuk Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jung Yeon Shim
- Department of Pediatrics, Kangbuk Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
23
|
Javadyan A, Papadopoulos NG, Kafetzis DA. Rhinovirus infections and adenoidal hypertrophy: do they interact with atopy in children? Expert Rev Anti Infect Ther 2014; 1:223-9. [PMID: 15482117 DOI: 10.1586/14787210.1.2.223] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Modern diagnostic methods allow the evaluation of the connection between rhinoviruses and atopy. Recent studies suggest that rhinoviruses are present in the adenoids at higher titers than in other specific sites, after inoculation of nasal mucosa or conjunctiva in volunteers. Therefore, it is possible that they might be responsible for specific local changes, while such changes may be influenced by atopy. This review focuses on the interactions between rhinoviral infection, the host's immune status and adenoidal disease.
Collapse
Affiliation(s)
- Artem Javadyan
- University of Athens, 'P and A Kiriakou' Children's Hospital, 13 Livadias St., GR-11527 Athens, Greece
| | | | | |
Collapse
|
24
|
Kim E, Lee H, Kim HS, Won S, Lee EK, Kim HS, Bang K, Chun YH, Yoon JS, Kim HH, Kim JT, Lee JS. The effect of rhinovirus on airway inflammation in a murine asthma model. KOREAN JOURNAL OF PEDIATRICS 2013; 56:482-9. [PMID: 24348661 PMCID: PMC3859881 DOI: 10.3345/kjp.2013.56.11.482] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 07/20/2013] [Accepted: 08/21/2013] [Indexed: 01/20/2023]
Abstract
PURPOSE The aim of the present study was to investigate the differences in lower airway inflammatory immune responses, including cellular responses and responses in terms of inflammatory mediators in bronchoalveolar lavage fluid (BALF) and the airway, to rhinovirus (RV) infection on asthma exacerbation by comparing a control and a murine asthma model, with or without RV infection. METHODS BALB/c mice were intraperitoneally injected with a crude extract of Dermatophagoides farinae (Df) or phosphate buffered saline (PBS) and were subsequently intranasally treated with a crude extract of Df or PBS. Airway responsiveness and cell infiltration, differential cell counts in BALF, and cytokine and chemokine concentrations in BALF were measured 24 hours after intranasal RV1B infection. RESULTS RV infection increased the enhanced pause (Penh) in both the Df sensitized and challenged mice (Df mice) and PBS-treated mice (PBS mice) (P<0.05). Airway eosinophil infiltration increased in Df mice after RV infection (P<0.05). The levels of interleukin (IL) 13, tumor necrosis factor alpha, and regulated on activation, normal T cells expressed and secreted (RANTES) increased in response to RV infection in Df mice, but not in PBS mice (P<0.05). The level of IL-10 significantly decreased following RV infection in Df mice (P<0.05). CONCLUSION Our findings suggest that the augmented induction of proinflammatory cytokines, Th2 cytokines, and chemokines that mediate an eosinophil response and the decreased induction of regulatory cytokines after RV infection may be important manifestations leading to airway inflammation with eosinophil infiltration and changes in airway responsiveness in the asthma model.
Collapse
Affiliation(s)
- Eugene Kim
- Department of Pediatrics, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Huisu Lee
- Department of Pediatrics, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Hyun Sook Kim
- Department of Pediatrics, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Sulmui Won
- Department of Pediatrics, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Eu Kyoung Lee
- Department of Pediatrics, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Hwan Soo Kim
- Department of Pediatrics, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Kyongwon Bang
- Department of Pediatrics, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Yoon Hong Chun
- Department of Pediatrics, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Jong-Seo Yoon
- Department of Pediatrics, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Hyun Hee Kim
- Department of Pediatrics, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Jin Tack Kim
- Department of Pediatrics, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Joon Sung Lee
- Department of Pediatrics, The Catholic University of Korea College of Medicine, Seoul, Korea
| |
Collapse
|
25
|
Kim JH, Moon BJ, Gong CH, Kim NH, Jang YJ. Detection of respiratory viruses in adult patients with perennial allergic rhinitis. Ann Allergy Asthma Immunol 2013; 111:508-11. [PMID: 24267360 PMCID: PMC7111117 DOI: 10.1016/j.anai.2013.08.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 08/16/2013] [Accepted: 08/25/2013] [Indexed: 11/18/2022]
Abstract
Background The symptoms of allergic rhinitis may be worsened by a viral respiratory infection. However, there are few data on the presence of respiratory virus in patients with allergic rhinitis. Objective To evaluate whether patients with allergic rhinitis have an increased frequency of respiratory virus detection in a prospective case–control study. Methods Fifty-eight adult patients diagnosed with perennial allergic rhinitis were evaluated from September 2011 through June 2012. A control group of 61 adult patients without allergy was included. Multiplex polymerase chain reaction was used to detect respiratory viruses in nasal lavage samples. Results Respiratory viruses were detected in 25 of 58 patients (43.1%) with perennial allergic rhinitis, but in only 15 of 61 control patients (24.6%). In virus-positive samples, multiple viruses were detected in 9 of 25 patients (36.0%) with perennial allergic rhinitis but in only 2 of 15 control patients (12.5%). Rhinovirus was the most common virus in patients without allergy and those with allergic rhinitis. There were significant differences in the detection rates of overall and multiple respiratory viruses and rhinovirus between the 2 groups (P < .05). However, in patients with allergic rhinitis, there was no statistically significant association between the detection of respiratory viruses and symptom scores. Conclusion This study shows that there is a high prevalence of respiratory viruses, especially rhinovirus, in patients with allergic rhinitis. Subsequent studies are needed to determine the clinical significance of highly prevalent respiratory viruses in patients with allergic rhinitis.
Collapse
Affiliation(s)
- Ji Heui Kim
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | | | | | | | | |
Collapse
|
26
|
Ilarraza R, Wu Y, Skappak CD, Ajamian F, Proud D, Adamko DJ. Rhinovirus has the unique ability to directly activate human T cells in vitro. J Allergy Clin Immunol 2013; 131:395-404. [PMID: 23374267 DOI: 10.1016/j.jaci.2012.11.041] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 10/22/2012] [Accepted: 11/30/2012] [Indexed: 12/24/2022]
Abstract
BACKGROUND Rhinovirus infection is a leading cause of exacerbation of airway diseases. We hypothesize that airway viruses activate inflammatory cells, inducing airway dysfunction. We have previously shown that airway viruses can induce eosinophil degranulation when cocultured with T cells and monocyte-derived dendritic cells (moDCs). These findings suggested that antigen presentation was important for T-cell activation. OBJECTIVE Given the clinical importance of rhinovirus, we sought to determine whether it had any unique abilities to activate inflammatory cells compared with another common virus, such as respiratory syncytial virus (RSV). METHODS We cocultured combinations of human leukocytes (T cells, moDCs, and eosinophils) with each virus. Using assays of BrdU incorporation, flow cytometry, and ELISA, we measured T-cell activation, rhinovirus expression, T-cell death, and eosinophil cysteinyl leukotriene release. RESULTS In contrast to RSV, rhinovirus induced T-cell activation without the involvement of moDCs. Without moDCs, rhinovirus induced T-cell proliferation of both CD4 and CD8(+) cells, cytokine production, and ultimately, eosinophil stimulation. Although chloroquine inhibited RSV-induced activation of T cells through moDCs, rhinovirus was not inhibited; UV inactivation did block the rhinovirus effect. We also found that T cells could be infected by rhinovirus in vitro and within human nasal explant tissue. Although Toll-like receptors did not appear to be involved in T-cell activation, antagonists of Jun N-terminal kinase and nuclear factor κB did inhibit T-cell responses to rhinovirus. CONCLUSION Rhinovirus has the unique ability to bypass antigen presentation and directly infect and activate human T cells. This could explain the strong association of rhinovirus with exacerbation of airway diseases.
Collapse
Affiliation(s)
- Ramses Ilarraza
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | |
Collapse
|
27
|
Lewis TC, Henderson TA, Carpenter AR, Ramirez IA, McHenry CL, Goldsmith AM, Ren X, Mentz GB, Mukherjee B, Robins TG, Joiner TA, Mohammad LS, Nguyen ER, Burns MA, Burke DT, Hershenson MB. Nasal cytokine responses to natural colds in asthmatic children. Clin Exp Allergy 2013. [PMID: 23181789 PMCID: PMC4219353 DOI: 10.1111/cea.12005] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background The mechanisms by which viruses induce asthma exacerbations are not well understood. Objective We characterized fluctuations in nasal aspirate cytokines during naturally occurring respiratory viral infections in children with asthma. Methods Sixteen children underwent home collections of nasal aspirates when they were without cold symptoms and again during self‐reported respiratory illnesses. The presence of viral infection was ascertained by multiplex PCR. Cytokines were measured using multiplex immune assay. mRNA expression for selected markers of viral infection was measured using RT‐PCR. A cumulative respiratory symptom score was calculated for each day of measurement. Generalized estimated equations were used to evaluate associations between viral infection and marker elevation, and between marker elevation and symptom score. Results The 16 patients completed a total of 37 weeks of assessment (15 ‘well’ weeks; 22 self‐assessed ‘sick’ weeks). Viral infections were detected in 3 of the ‘well’ weeks and 17 of the ‘sick’ weeks (10 rhinovirus, three coronavirus, two influenza A, two influenza B, two respiratory syncytial virus, one parainfluenza). Compared to virus‐negative well weeks, nasal aspirate IFN‐γ, CXCL8/IL‐8, CXCL10/IP‐10, CCL5/RANTES, CCL11/eotaxin‐1, CCL2/MCP‐1, CCL4/MIP‐1β, CCL7/MCP‐3, and CCL20/MIP3α protein levels increased during virus‐positive sick weeks. Only a subset of cytokines (IFN‐γ, CXCL8, CCL2, CCL4, CCL5, and CCL20) correlated with self‐reported respiratory tract symptoms. While many aspirates were dilute and showed no mRNA signal, viral infection significantly increased the number of samples that were positive for IFN‐λ1, IFN‐λ2/3, TLR3, RIG‐I, and IRF7 mRNA. Conclusions and clinical relevance We conclude that in children with asthma, naturally occurring viral infections apparently induce a robust innate immune response including expression of specific chemokines, IFNs, and IFN‐responsive genes.
Collapse
Affiliation(s)
- T C Lewis
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Córdova Martínez A, Del Villar Sordo V. Infecciones en el deportista de elite: gastroenteritis aguda infecciosa e infecciones respiratorias víricas comunes. Medicine (Baltimore) 2013; 8:4587-4595. [PMID: 32287920 PMCID: PMC7144317 DOI: 10.1016/s0304-5412(02)70853-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/30/2022] Open
Affiliation(s)
- A Córdova Martínez
- Departamento de Fisiología. Escuela Universitaria de Fisioterapia de Soria. Universidad de Valladolid. Equipo ciclista Kelme
| | - V Del Villar Sordo
- Departamento de Medicina. Escuela Universitaria de Fisioterapia de Soria. Universidad de Valladolid
- Servicio de Medicina Interna. Hospital del Insalud de Soria. Soria
| |
Collapse
|
29
|
Campbell-Harding G, Sawkins H, Bedke N, Holgate ST, Davies DE, Andrews AL. The innate antiviral response upregulates IL-13 receptor α2 in bronchial fibroblasts. J Allergy Clin Immunol 2013; 131:849-55. [PMID: 23069489 DOI: 10.1016/j.jaci.2012.08.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 08/13/2012] [Accepted: 08/22/2012] [Indexed: 02/08/2023]
Abstract
BACKGROUND IL-13 is key mediator of allergic inflammation in asthmatic patients. We have previously shown that the decoy receptor IL-13 receptor (IL-13R) α2 attenuates responses of fibroblasts to IL-13. Because the expression of IL-13Rα2 can be regulated by IFN-γ, a type II interferon, we hypothesized that innate antiviral responses characterized by type I interferon expression can also induce IL-13Rα2 expression. OBJECTIVE We sought to induce an innate antiviral response in primary fibroblasts using exposure to double-stranded RNA (dsRNA) and to examine the expression and function of IL-13Rα2. METHODS Primary human fibroblasts were cultured from endobronchial biopsy specimens obtained from healthy or asthmatic volunteers and challenged with dsRNA. Upregulation of IL-13Rα2 mRNA was measured by using real-time quantitative PCR, and cell-surface IL-13Rα2 protein expression was measured by using fluorescence-activated cell sorting. Eotaxin release was determined by means of ELISA. RESULTS Direct treatment with IFN-β led to an upregulation of IL-13Rα2. Exposure to dsRNA rapidly induced IFN-β mRNA in fibroblasts, and this was followed by significant induction of IL-13Rα2 mRNA and cell-surface protein expression, which was dependent on de novo protein synthesis. A neutralizing antibody to the IFN-α/β receptor blocked cell-surface expression of IL-13Rα2 in the presence of dsRNA. Pretreatment of fibroblasts with dsRNA led to attenuation of IL-13-stimulated eotaxin production. However, the presence of an IL-13Rα2 neutralizing antibody restored IL-13-stimulated eotaxin production in dsRNA-treated cells. CONCLUSION IFN-β induces IL-13Rα2 expression, leading to a consequential suppression of responsiveness to IL-13. These data suggest cross-talk between TH1 and TH2 pathways and point to an immunomodulatory role for IL-13Rα2 in human bronchial fibroblasts during viral infection.
Collapse
Affiliation(s)
- Gemma Campbell-Harding
- Academic Unit of Clinical and Experimental Sciences and the Southampton NIHR Respiratory Biomedical Research Unit, University of Southampton Faculty of Medicine, Sir Henry Wellcome Laboratories, Southampton General Hospital, Southampton, United Kingdom
| | | | | | | | | | | |
Collapse
|
30
|
Adler FR, Kim PS. Models of contrasting strategies of rhinovirus immune manipulation. J Theor Biol 2013; 327:1-10. [PMID: 23485454 DOI: 10.1016/j.jtbi.2013.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 01/22/2013] [Accepted: 02/15/2013] [Indexed: 01/09/2023]
Abstract
Rhinoviruses, consisting of well over one hundred serotypes that cause a plurality of common colds, are completely cleared by the host immune system after causing minimal cell death, but often without inducing long-term immune memory. We develop mathematical models of two kinds of rhinoviruses, the major group and minor group, that use different receptors to enter target cells. Roughly the 90 serotypes in the major group bind to ICAM-1, a molecule that is upregulated on antigen-presenting cells, and alter the timing, location and type of the immune response. The 12 members of the minor group do not so modulate the response. Our model predicts similar virus dynamics for the major and minor groups but with quite different underlying mechanisms. Over a range of key parameters that quantify immune manipulation, disease outcomes lie within a triangle in the plane describing damage and memory, of which the major and minor group form two corners. This model of infection by a highly adapted and low virulence virus provides a starting point for understanding the development of asthma and other pathologies.
Collapse
Affiliation(s)
- Frederick R Adler
- Department of Mathematics, 155 South 1400 East, University of Utah, Salt Lake City, UT 84112, United States.
| | | |
Collapse
|
31
|
Virus infection-induced bronchial asthma exacerbation. Pulm Med 2012; 2012:834826. [PMID: 22966430 PMCID: PMC3432542 DOI: 10.1155/2012/834826] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 05/01/2012] [Accepted: 06/22/2012] [Indexed: 12/04/2022] Open
Abstract
Infection with respiratory viruses, including rhinoviruses, influenza virus, and respiratory syncytial virus, exacerbates asthma, which is associated with processes such as airway inflammation, airway hyperresponsiveness, and mucus hypersecretion. In patients with viral infections and with infection-induced asthma exacerbation, inflammatory mediators and substances, including interleukins (ILs), leukotrienes and histamine, have been identified in the airway secretions, serum, plasma, and urine. Viral infections induce an accumulation of inflammatory cells in the airway mucosa and submucosa, including neutrophils, lymphocytes and eosinophils. Viral infections also enhance the production of inflammatory mediators and substances in airway epithelial cells, mast cells, and other inflammatory cells, such as IL-1, IL-6, IL-8, GM-CSF, RANTES, histamine, and intercellular adhesion molecule-1. Viral infections affect the barrier function of the airway epithelial cells and vascular endothelial cells. Recent reports have demonstrated augmented viral production mediated by an impaired interferon response in the airway epithelial cells of asthma patients. Several drugs used for the treatment of bronchial asthma reduce viral and pro-inflammatory cytokine release from airway epithelial cells infected with viruses. Here, I review the literature on the pathogenesis of the viral infection-induced exacerbation of asthma and on the modulation of viral infection-induced airway inflammation.
Collapse
|
32
|
Singanayagam A, Joshi PV, Mallia P, Johnston SL. Viruses exacerbating chronic pulmonary disease: the role of immune modulation. BMC Med 2012; 10:27. [PMID: 22420941 PMCID: PMC3353868 DOI: 10.1186/1741-7015-10-27] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 03/15/2012] [Indexed: 12/30/2022] Open
Abstract
Chronic pulmonary diseases are a major cause of morbidity and mortality and their impact is expected to increase in the future. Respiratory viruses are the most common cause of acute respiratory infections and it is increasingly recognized that respiratory viruses are a major cause of acute exacerbations of chronic pulmonary diseases such as asthma, chronic obstructive pulmonary disease and cystic fibrosis. There is now increasing evidence that the host response to virus infection is dysregulated in these diseases and a better understanding of the mechanisms of abnormal immune responses has the potential to lead to the development of new therapies for virus-induced exacerbations. The aim of this article is to review the current knowledge regarding the role of viruses and immune modulation in chronic pulmonary diseases and discuss avenues for future research and therapeutic implications.
Collapse
Affiliation(s)
- Aran Singanayagam
- National Heart and Lung Institute, Imperial College London, Norfolk Place, London W2 1PG, UK
| | | | | | | |
Collapse
|
33
|
Koh YI, Shim JU, Wi J, Kwon YE. The role of natural killer T cells in the pathogenesis of acute exacerbation of human asthma. Int Arch Allergy Immunol 2012; 158:131-41. [PMID: 22269676 DOI: 10.1159/000330908] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2011] [Accepted: 07/15/2011] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Natural killer T (NKT) cells have been reported to play a crucial role in the pathogenesis of asthma in a mouse model of acute asthma. The present study aimed to investigate the role of NKT cells in the immune pathogenesis of acute exacerbation of human asthma. METHODS Blood and sputum were obtained at baseline and 8 h after a challenge in 20 asthmatics who underwent allergen bronchial provocation testing and during exacerbation and convalescence in 9 asthmatics who were admitted to hospital with an acute exacerbation after an upper respiratory tract infection. 6B11+ or Vα24+ NKT cells were measured with flow cytometry. Inflammatory cells, cytokines and chemokines were determined in sputum. RESULTS The number of blood NKT cells did not change after a positive allergen challenge compared to the baseline. However, blood CD4+Vα24+ NKT cells decreased during infection-associated asthma exacerbations compared to the convalescence measurements of the same patients (p < 0.05) or the baseline measurements of asthmatics who underwent allergen challenges (p < 0.01). The number of sputum NKT cells did not change after a positive allergen challenge or during infection-associated asthma exacerbations. Eosinophils and various cytokines and chemokines increased in sputum during infection-associated asthma exacerbations. Blood CD4+Vα24+ NKT cells were inversely related to sputum eosinophils (Spearman's correlation coefficient = -0.62; p < 0.01). CONCLUSION Blood NKT cells decreased during infection-associated asthma exacerbation and were inversely associated with eosinophilic airway inflammation, suggesting that blood NKT cells might be mobilized to the airways and lungs during asthma exacerbation in humans.
Collapse
Affiliation(s)
- Young-Il Koh
- Department of Allergy, Asthma and Clinical Immunology and Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju, Korea.
| | | | | | | |
Collapse
|
34
|
Takaku Y, Nakagome K, Kobayashi T, Hagiwara K, Kanazawa M, Nagata M. IFN-γ-inducible protein of 10 kDa upregulates the effector functions of eosinophils through β2 integrin and CXCR3. Respir Res 2011; 12:138. [PMID: 22004287 PMCID: PMC3215664 DOI: 10.1186/1465-9921-12-138] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2011] [Accepted: 10/17/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Eosinophils play an important role in the pathogenesis of bronchial asthma and its exacerbation. Recent reports suggest the involvement of IFN-γ-inducible protein of 10 kDa (IP-10) in virus-induced asthma exacerbation. The objective of this study was to examine whether CXCR3 ligands including IP-10 modify the effector functions of eosinophils. METHODS Eosinophils isolated from the blood of healthy donors were stimulated with CXCR3 ligands and their adhesion to rh-ICAM-1 was then measured using eosinophil peroxidase assays. The generation of eosinophil superoxide anion (O2-) was examined based on the superoxide dismutase-inhibitable reduction of cytochrome C. Eosinophil-derived neurotoxin (EDN) release was evaluated to determine whether CXCR3 ligands induced eosinophil degranulation. Cytokine and chemokine production by eosinophils was examined using a Bio-plex assay. RESULTS Eosinophil adhesion to ICAM-1 was significantly enhanced by IP-10, which also significantly induced eosinophil O2- generation in the presence of ICAM-1. Both the enhanced adhesion and O2- generation were inhibited by an anti-β2 integrin mAb or an anti-CXCR3 mAb. Other CXCR3 ligands, such as monokine induced by IFN-γ (Mig) and IFN-inducible T cell α chemoattractant (I-TAC), also induced eosinophil adhesion and O2- generation in the presence of ICAM-1. IP-10, but not Mig or I-TAC, increased the release of EDN. IP-10 increased the production of a number of cytokines and chemokines by eosinophils. CONCLUSIONS These findings suggest that CXCR3 ligands such as IP-10 can directly upregulate the effector functions of eosinophils. These effects might be involved in the activation and infiltration of eosinophils in the airway of asthma, especially in virus-induced asthma exacerbation.
Collapse
Affiliation(s)
- Yotaro Takaku
- Department of Respiratory Medicine, Saitama Medical University, 38 Morohongo, Moroyama-cho, Iruma-gun, Saitama, Japan
| | | | | | | | | | | |
Collapse
|
35
|
Kuo C, Lim S, King NJC, Bartlett NW, Walton RP, Zhu J, Glanville N, Aniscenko J, Johnston SL, Burgess JK, Black JL, Oliver BG. Rhinovirus infection induces expression of airway remodelling factors in vitro and in vivo. Respirology 2011; 16:367-77. [PMID: 21199160 DOI: 10.1111/j.1440-1843.2010.01918.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND OBJECTIVE A hallmark of asthma is airway remodelling, which includes increased deposition of extracellular matrix (ECM) protein. Viral infections may promote the development of asthma and are the most common causes of asthma exacerbations. We evaluated whether rhinovirus (RV) infection induces airway remodelling, as assessed by ECM deposition. METHODS Primary human bronchial epithelial cells and lung parenchymal fibroblasts were infected with RV-2 or RV-16, or treated with RV-16 RNA, imiquimod (Toll-like receptor (TLR) 7/8 agonist) or polyinosinic : polycytidylic acid (poly I : C) (activator of TLR 3, retinoic-acid-inducible protein I and melanoma-differentiated-associated gene 5). Changes in ECM proteins and their transcription were measured by ELISA and quantitative real-time PCR. In addition, gene expression for ECM proteins was assessed in a mouse model of RV infection. RESULTS RV infection increased deposition of the ECM protein, perlecan, by human bronchial epithelial cells, and collagen V and matrix-bound vascular endothelial growth factor were increased in both human bronchial epithelial cell and fibroblast cultures. Purified RV-16 RNA, poly I : C and imiquimod induced similar increases in ECM deposition to those observed with RV-infected fibroblasts. However, only poly I : C induced ECM deposition by bronchial epithelial cells, suggesting that RV-induced ECM deposition is mediated through TLR. Furthermore, gene expression for fibronectin and collagen I was increased in lung homogenates of mice infected with RV-1b. CONCLUSIONS RV infection and TLR ligands promote ECM deposition in isolated cell systems and RV induces ECM gene expression in vivo, thus demonstrating that RV has the potential to contribute to remodelling of the airways through induction of ECM deposition.
Collapse
Affiliation(s)
- Curtis Kuo
- Discipline of Pharmacology, The University of Sydney, Camperdown, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Bystrom J, Amin K, Bishop-Bailey D. Analysing the eosinophil cationic protein--a clue to the function of the eosinophil granulocyte. Respir Res 2011; 12:10. [PMID: 21235798 PMCID: PMC3030543 DOI: 10.1186/1465-9921-12-10] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Accepted: 01/14/2011] [Indexed: 02/06/2023] Open
Abstract
Eosinophil granulocytes reside in respiratory mucosa including lungs, in the gastro-intestinal tract, and in lymphocyte associated organs, the thymus, lymph nodes and the spleen. In parasitic infections, atopic diseases such as atopic dermatitis and asthma, the numbers of the circulating eosinophils are frequently elevated. In conditions such as Hypereosinophilic Syndrome (HES) circulating eosinophil levels are even further raised. Although, eosinophils were identified more than hundred years ago, their roles in homeostasis and in disease still remain unclear. The most prominent feature of the eosinophils are their large secondary granules, each containing four basic proteins, the best known being the eosinophil cationic protein (ECP). This protein has been developed as a marker for eosinophilic disease and quantified in biological fluids including serum, bronchoalveolar lavage and nasal secretions. Elevated ECP levels are found in T helper lymphocyte type 2 (atopic) diseases such as allergic asthma and allergic rhinitis but also occasionally in other diseases such as bacterial sinusitis. ECP is a ribonuclease which has been attributed with cytotoxic, neurotoxic, fibrosis promoting and immune-regulatory functions. ECP regulates mucosal and immune cells and may directly act against helminth, bacterial and viral infections. The levels of ECP measured in disease in combination with the catalogue of known functions of the protein and its polymorphisms presented here will build a foundation for further speculations of the role of ECP, and ultimately the role of the eosinophil.
Collapse
Affiliation(s)
- Jonas Bystrom
- Translational Medicine and Therapeutics, William Harvey Research Institute, Bart's and the London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Kawa Amin
- Respiratory Medicine and Allergology, Department of Medical Science, Uppsala University Hospital, Uppsala, Sweden
- College of Medicine, Sulaimani University, Sulaimani, Iraq
| | - David Bishop-Bailey
- Translational Medicine and Therapeutics, William Harvey Research Institute, Bart's and the London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
37
|
Björnsson E, Lúdvíksdóttir D, Hedenström H, Eriksson BM, Högman M, Venge P, Janson C. Airway hyperresponsiveness, peak flow variability and inflammatory markers in non-asthmatic subjects with respiratory infections. CLINICAL RESPIRATORY JOURNAL 2010; 1:42-50. [PMID: 20298277 DOI: 10.1111/j.1752-699x.2007.00013.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE The aim of this study was to characterise non-asthmatic subjects with asthma-like symptoms during a common cold, particularly in relation to airway hyperresponsiveness (AHR). MATERIALS AND METHODS Subjects with acute respiratory infections and a group of controls (n = 20 + 20), age 20-65 years, underwent bronchial provocations with methacholine, adenosine and cold air. All were non-smokers and had no history of asthma or heart disease. Those with infection had asthma-like symptoms (>2). Measurements of exhaled nitric oxide (eNO), serum levels of eosinophil cationic protein (ECP), eosinophil peroxidase, myeloperoxidase and human neutrophil lipocalin were made at each provocation. A 17-day symptom and peak flow diary was calculated. RESULTS No differences between the two groups were found, regarding responsiveness to methacholine, adenosine or cold air challenge, as well as the inflammatory markers measured. In the infected group, the mean (standard deviation) ECP was higher in those with AHR to methacholine or cold air [15.7 (6.5) and 11.4 (4.2) microg/L, respectively; P < 0.05]; furthermore, eNO was higher in the infected group [116 (54) and 88 (52) nL/min, respectively; P = 0.055]. The infected group had, at all times, more symptoms and higher peak flow, with a decrease in the symptoms (P = 0.02) and a tendency to change in peak flow variation (P = 0.06). CONCLUSION AHR does not seem to be the main cause of asthma-like symptoms in adults with infectious wheezing. Peak flow variation and symptom prevalence during the post-infection period may imply airway pathology different from AHR.
Collapse
Affiliation(s)
- Eythór Björnsson
- Department of Allergy and Respiratory Medicine, Landspitali-University Hospital, Reykjavik, Iceland.
| | | | | | | | | | | | | |
Collapse
|
38
|
Nagarkar DR, Bowman ER, Schneider D, Wang Q, Shim J, Zhao Y, Linn MJ, McHenry CL, Gosangi B, Bentley JK, Tsai WC, Sajjan US, Lukacs NW, Hershenson MB. Rhinovirus infection of allergen-sensitized and -challenged mice induces eotaxin release from functionally polarized macrophages. THE JOURNAL OF IMMUNOLOGY 2010; 185:2525-35. [PMID: 20644177 DOI: 10.4049/jimmunol.1000286] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Human rhinovirus is responsible for the majority of virus-induced asthma exacerbations. To determine the immunologic mechanisms underlying rhinovirus (RV)-induced asthma exacerbations, we combined mouse models of allergic airways disease and human rhinovirus infection. We inoculated OVA-sensitized and challenged BALB/c mice with rhinovirus serotype 1B, a minor group strain capable of infecting mouse cells. Compared with sham-infected, OVA-treated mice, virus-infected mice showed increased lung infiltration with neutrophils, eosinophils and macrophages, airway cholinergic hyperresponsiveness, and increased lung expression of cytokines including eotaxin-1/CCL11, IL-4, IL-13, and IFN-gamma. Administration of anti-eotaxin-1 attenuated rhinovirus-induced airway eosinophilia and responsiveness. Immunohistochemical analysis showed eotaxin-1 in the lung macrophages of virus-infected, OVA-treated mice, and confocal fluorescence microscopy revealed colocalization of rhinovirus, eotaxin-1, and IL-4 in CD68-positive cells. RV inoculation of lung macrophages from OVA-treated, but not PBS-treated, mice induced expression of eotaxin-1, IL-4, and IL-13 ex vivo. Macrophages from OVA-treated mice showed increased expression of arginase-1, Ym-1, Mgl-2, and IL-10, indicating a shift in macrophage activation status. Depletion of macrophages from OVA-sensitized and -challenged mice reduced eosinophilic inflammation and airways responsiveness following RV infection. We conclude that augmented airway eosinophilic inflammation and hyperresponsiveness in RV-infected mice with allergic airways disease is directed in part by eotaxin-1. Airway macrophages from mice with allergic airways disease demonstrate a change in activation state characterized in part by altered eotaxin and IL-4 production in response to RV infection. These data provide a new paradigm to explain RV-induced asthma exacerbations.
Collapse
Affiliation(s)
- Deepti R Nagarkar
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Jackson DJ, Johnston SL. The role of viruses in acute exacerbations of asthma. J Allergy Clin Immunol 2010; 125:1178-87; quiz 1188-9. [PMID: 20513517 PMCID: PMC7172767 DOI: 10.1016/j.jaci.2010.04.021] [Citation(s) in RCA: 261] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2010] [Accepted: 04/21/2010] [Indexed: 01/17/2023]
Abstract
Viral respiratory infections are the most common cause of an acute asthma exacerbation in both children and adults and represent a significant global health burden. An increasing body of evidence supports the hypothesis that these infections cause a greater degree of morbidity in asthmatic subjects than in the healthy population, emphasizing a discrepancy in the antiviral response of asthmatics. In this review we discuss why such a discrepancy might exist, examining the role of the bronchial epithelium as well as the main inflammatory cells, mediators, and molecular pathways that are involved in the immune response. In addition, the potential impact of virus-induced asthma exacerbations on airway remodelling is reviewed and we explore which therapeutic options might be of benefit in preventing the deterioration of asthma control seen following viral infection.
Collapse
Key Words
- asthma
- acute exacerbation
- virus
- bal, bronchoalveolar lavage
- bec, bronchial epithelial cell
- fgf, fibroblast growth factor
- hrv, human rhinovirus
- icam-1, intercellular adhesion molecule 1
- ip-10, interferon-inducible protein 10
- irf, interferon regulatory factor
- nf-κb, nuclear factor kappa b
- prr, pattern-recognition receptor
- socs1, suppressor of cytokine signaling 1
- tlr, toll-like receptor
- vegf, vascular endothelial growth factor
Collapse
Affiliation(s)
- David J Jackson
- Department of Respiratory Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| | | |
Collapse
|
40
|
Respiratory tract infections and lung function in early life--"Cling together, swing together". Allergol Immunopathol (Madr) 2010; 38:107-9. [PMID: 20434824 DOI: 10.1016/j.aller.2010.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Accepted: 02/18/2010] [Indexed: 11/22/2022]
|
41
|
Nagarkar DR, Wang Q, Shim J, Zhao Y, Tsai WC, Lukacs NW, Sajjan U, Hershenson MB. CXCR2 is required for neutrophilic airway inflammation and hyperresponsiveness in a mouse model of human rhinovirus infection. THE JOURNAL OF IMMUNOLOGY 2009; 183:6698-707. [PMID: 19864593 DOI: 10.4049/jimmunol.0900298] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Human rhinovirus (RV) infection is responsible for the majority of virus-induced asthma exacerbations. Using a mouse model of human RV infection, we sought to determine the requirement of CXCR2, the receptor for ELR-positive CXC chemokines, for RV-induced airway neutrophilia and hyperresponsiveness. Wild-type and CXCR2(-/-) mice were inoculated intranasally with RV1B or sham HeLa cell supernatant. Following RV1B infection, CXCR2(-/-) mice showed reduced airway and lung neutrophils and cholinergic responsiveness compared with wild-type mice. Similar results were obtained in mice treated with neutralizing Ab to Ly6G, a neutrophil-depleting Ab. Lungs from RV-infected, CXCR2(-/-) mice showed significantly reduced production of TNF-alpha, MIP-2/CXCL2, and KC/CXCL1 and lower expression of MUC5B compared with RV-treated wild-type mice. The requirement of TNF-alpha for RV1B-induced airway responses was tested using TNFR1(-/-) mice. TNFR1(-/-) animals displayed reduced airway responsiveness to RV1B, even when exogenous MIP-2 was added to the airways. We conclude that CXCR2 is required for RV-induced neutrophilic airway inflammation and that neutrophil TNF-alpha release is required for airway hyperresponsiveness.
Collapse
Affiliation(s)
- Deepti R Nagarkar
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109-5688, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Wark PAB, Grissell T, Davies B, See H, Gibson PG. Diversity in the bronchial epithelial cell response to infection with different rhinovirus strains. Respirology 2009; 14:180-6. [PMID: 19207121 DOI: 10.1111/j.1440-1843.2009.01480.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND AND OBJECTIVE Infection with rhinovirus (RV) is the most common trigger for acute asthma and COPD. The aim of this study was to characterize the variability in the response of primary bronchial epithelial cells to infection with several strains of RV. METHODS RV strains, RV-43, RV-48 (major group RV), RV-47 (minor) and EV-68 (enterovirus), were cultured from subjects with acute asthma and compared with the laboratory RV strains, RV-16, RV-14 (major) and RV-1B (minor). Primary bronchial epithelial cells were obtained from healthy control and asthmatic subjects by endobronchial brushing. Response to infection was assessed by the release of IL-6, interferon (IFN)-gamma induced protein (IP)-10 and IFN-beta, as measured by ELISA. Viral replication was assessed by serial titration assays and cell viability by flow cytometry. RESULTS Major group RV strains and EV-68 all efficiently infected and replicated in epithelial cells causing little cell death. The clinical major group RV strains caused greater release of IL-6 and IP-10 compared with laboratory major group RV strains. Infection with minor group RV resulted in greater release of IP-10, IL-6 and IFN-beta that was associated with induction of apoptosis and less efficient viral replication. Asthmatic bronchial epithelial cells were less able to respond by releasing IFN-beta following infection with RV-1B. CONCLUSIONS Considerable diversity exists in the response to RV strains, especially between minor and major group RV. The impaired IFN-beta response in asthmatic bronchial epithelial cells may make them particularly susceptible to minor group RV.
Collapse
Affiliation(s)
- Peter A B Wark
- Centre for Asthma and Respiratory Disease, Hunter Medical Research Institute, University of Newcastle, Newcastle, UK.
| | | | | | | | | |
Collapse
|
43
|
[Infections and asthma]. Internist (Berl) 2009; 49:1302, 1304-6, 1308-9. [PMID: 18839125 DOI: 10.1007/s00108-008-2133-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Asthma is a chronic inflammatory disorder of the airways. Allergen-induced airway inflammation plays a central role in the pathogenesis of the disease whereas infections are known to be important triggers of acute exacerbations. Chronic or latent infections may also influence and enhance the local inflammation. The aim of this review is to give an overview of the different pathogens and to present the current understanding of the role of respiratory viruses and atypical bacteria in the pathogenesis of the disease.
Collapse
|
44
|
Message SD, Johnston SL. Infections. ASTHMA AND COPD 2009. [PMCID: PMC7149941 DOI: 10.1016/b978-0-12-374001-4.00037-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
This chapter reviews the epidemiological evidence implicating infectious pathogens as triggers and will discuss the mechanisms of interaction between the host–pathogen response and preexisting airway pathology that result in an exacerbation. Asthma is a multifaceted syndrome involving atopy, bronchial hyperreactivity, and IgE and non-IgE-mediated acute and chronic immune responses. The asthmatic airway is characterized by an infiltrate of eosinophils and of T-lymphocytes expressing the type 2 cytokines IL-4, IL-5, and IL-13. Trigger factors associated with acute exacerbations of asthma include exposure to environmental allergens, especially animals, molds, pollens and mites, cold, exercise, and drugs. The frequency of exacerbations is a major factor in the quality of life of patients with COPD. The typical clinical features of an exacerbation include increased dyspnea, wheezing, cough, sputum production, and worsened gas exchange. Although noninfectious causes of exacerbations such as allergy, air pollution, or inhaled irritants including cigarette smoke may be important, acute airway infections are the major precipitants. The infection and consequent host inflammatory response result in increased airway obstruction. The success of vaccination to prevent respiratory virus infections has been limited by significant variation within the major virus types causing disease. Currently much of the treatment of infective exacerbations of asthma and COPD is symptomatic, consisting of increased bronchodilators, either short-acting β 2—agonists in inhaled or intravenous form or anticholinergics or theophyllines, or supportive in the form of oxygen and in severe cases noninvasive or invasive ventilatory measures.
Collapse
|
45
|
Host immune responses to rhinovirus: mechanisms in asthma. J Allergy Clin Immunol 2008; 122:671-682. [PMID: 19014757 DOI: 10.1016/j.jaci.2008.08.013] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Revised: 08/15/2008] [Accepted: 08/18/2008] [Indexed: 11/22/2022]
Abstract
Viral respiratory infections can have a profound effect on many aspects of asthma including its inception, exacerbations, and, possibly, severity. Of the many viral respiratory infections that influence asthma, the common cold virus, rhinovirus, has emerged as the most frequent illness associated with exacerbations and other aspects of asthma. The mechanisms by which rhinovirus influences asthma are not fully established, but current evidence indicates that the immune response to this virus is critical in this process. Many airway cell types are involved in the immune response to rhinovirus, but most important are respiratory epithelial cells and possibly macrophages. Infection of epithelial cells generates a variety of proinflammatory mediators to attract inflammatory cells to the airway with a subsequent worsening of underlying disease. Furthermore, there is evidence that the epithelial airway antiviral response to rhinovirus may be defective in asthma. Therefore, understanding the immune response to rhinovirus is a key step in defining mechanisms of asthma, exacerbations, and, perhaps most importantly, improved treatment.
Collapse
|
46
|
Targeting the NF-kappaB pathway in asthma and chronic obstructive pulmonary disease. Pharmacol Ther 2008; 121:1-13. [PMID: 18950657 PMCID: PMC7172981 DOI: 10.1016/j.pharmthera.2008.09.003] [Citation(s) in RCA: 302] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2008] [Accepted: 09/09/2008] [Indexed: 11/23/2022]
Abstract
Asthma and chronic obstructive pulmonary disease are inflammatory lung disorders responsible for significant morbidity and mortality worldwide. While the importance of allergic responses in asthma is well known, respiratory viral and bacterial infections and pollutants especially cigarette smoke are important factors in the pathogenesis of both diseases. Corticosteroid treatment remains the first preference of treatment in either disease, however these therapies are not always completely effective, and are associated with side effects and steroid resistance. Due to such limitations, development of new treatments represents a major goal for both the pharmaceutical industry and academic researchers. There are now excellent reasons to promote NF-kappaB signalling intermediates and Rel family proteins as potential therapeutic targets for both asthma and chronic obstructive pulmonary disease. This notion is supported by the fact that much of the underlying inflammation of both diseases independent of stimuli, is mediated at least in part, by NF-kappaB mediated signalling events in several cell types. Also, a range of inhibitors of NF-kappaB signalling intermediates are now available, including DNA oligonucleotides and DNA-peptide molecules that act as NF-kappaB decoy sequences, small molecule inhibitors such as IKK-beta inhibitors, and proteasome inhibitors affecting NF-kappaB signalling, that have either shown promise in animal models or have begun clinical trials in other disorders. This review will focus on the role of NF-kappaB in both diseases, will discuss its suitability as a target, and will highlight recent key studies that support the potential of NF-kappaB as a therapeutic target in these two important inflammatory lung diseases.
Collapse
|
47
|
Newcomb DC, Sajjan US, Nagarkar DR, Wang Q, Nanua S, Zhou Y, McHenry CL, Hennrick KT, Tsai WC, Bentley JK, Lukacs NW, Johnston SL, Hershenson MB. Human rhinovirus 1B exposure induces phosphatidylinositol 3-kinase-dependent airway inflammation in mice. Am J Respir Crit Care Med 2008; 177:1111-21. [PMID: 18276942 PMCID: PMC2383993 DOI: 10.1164/rccm.200708-1243oc] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2007] [Accepted: 02/11/2008] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Infection with rhinovirus (RV) triggers exacerbations of asthma and chronic obstructive lung disease. OBJECTIVES We sought to develop a mouse model of RV employing RV1B, a minor group serotype that binds to the low-density lipoprotein receptor. METHODS C57BL/6 mice were inoculated intranasally with RV1B, replication-deficient ultraviolet (UV)-irradiated RV1B, or RV39, a major group virus. MEASUREMENTS AND MAIN RESULTS Viral RNA was present in the lungs of RV1B-treated mice, but not in those exposed to UV-irradiated RV1B or RV39. Lung homogenates of RV-treated mice contained infectious RV 4 days after inoculation. RV1B exposure induced neutrophilic and lymphocytic airway inflammation, as well as increased lung expression of KC, macrophage-inflammatory protein-2, and IFN-alpha and IFN-beta. RV1B-exposed mice showed airway hyperresponsiveness 1 and 4 days after inoculation. UV-irradiated RV1B induced modest neutrophilic airway inflammation and hyperresponsiveness 1 day after exposure. Both RV1B and UV-irradiated RV1B, but not RV39, increased lung phosphorylation of Akt. Confocal immunofluorescence showed colocalization of RV1B and phospho-Akt in the airway epithelium. Finally, pretreatment with the phosphatidylinositol 3-kinase inhibitor LY294002 attenuated chemokine production and neutrophil infiltration. CONCLUSIONS We conclude that RV1B induces airway inflammation in vivo. Evidence is presented that viral replication occurs in vivo and is required for maximal responses. On the other hand, viral replication was not required for a subset of RV-induced responses, including neutrophilic inflammation, airway hyperresponsiveness, and Akt phosphorylation. Finally, phosphatidylinositol 3-kinase/Akt signaling is required for maximal RV1B-induced airway neutrophilic inflammation, likely via its essential role in virus internalization.
Collapse
Affiliation(s)
- Dawn C Newcomb
- Medical Sciences Research Building II, 1150 W. Medical Center Drive, Room 3570B, Ann Arbor, MI 48109-0688, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Davoine F, Cao M, Wu Y, Ajamian F, Ilarraza R, Kokaji AI, Moqbel R, Adamko DJ. Virus-induced eosinophil mediator release requires antigen-presenting and CD4+ T cells. J Allergy Clin Immunol 2008; 122:69-77, 77.e1-2. [PMID: 18472150 DOI: 10.1016/j.jaci.2008.03.028] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2007] [Revised: 03/03/2008] [Accepted: 03/18/2008] [Indexed: 01/05/2023]
Abstract
BACKGROUND The most frequent trigger of asthma exacerbation is infection with common airway viruses; however, the precise mechanism regulating such severe reactions is not understood. The presence of airway eosinophil products is a unique feature detected in asthmatic airways. Using an animal model, we previously demonstrated that T cells play an important role in regulating an eosinophil-dependant pathway of virus-induced airway hyperreactivity. We hypothesize that human eosinophils respond to viruses, although only after interaction with T cells. OBJECTIVES We sought to determine whether eosinophils can respond to airway viruses in vitro and determine the mechanism of response. METHODS An in vitro coculture model of human eosinophils, antigen-presenting cells, and T cells was used with parainfluenza virus, respiratory syncytial virus, or rhinovirus. We measured release of eosinophil peroxidase (EPO) in concert with T-cell proliferation, cytokine release, and changes in T-cell phenotype. RESULTS The viruses induced release of EPO when coincubated in the presence of antigen-presenting cells (dendritic cells or macrophages) and T cells. Virus-mediated release was associated with proliferation of CD3(+)CD4(+) T cells and release of cytokines. UV inactivation of the virus did not prevent virus-induced EPO release or T-cell proliferation. Proliferating CD4(+) T cells show increased expression of CD25 and CD45RO. CD8(+) T cells were not activated. CONCLUSION Virus-induced EPO release can occur in the context of antigen presentation to CD4(+) T cells.
Collapse
Affiliation(s)
- Francis Davoine
- Pulmonary Research Group, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Shiraishi Y, Asano K, Niimi K, Fukunaga K, Wakaki M, Kagyo J, Takihara T, Ueda S, Nakajima T, Oguma T, Suzuki Y, Shiomi T, Sayama K, Kagawa S, Ikeda E, Hirai H, Nagata K, Nakamura M, Miyasho T, Ishizaka A. Cyclooxygenase-2/prostaglandin D2/CRTH2 pathway mediates double-stranded RNA-induced enhancement of allergic airway inflammation. THE JOURNAL OF IMMUNOLOGY 2008; 180:541-9. [PMID: 18097056 DOI: 10.4049/jimmunol.180.1.541] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Respiratory RNA viruses responsible for the common cold often worsen airway inflammation and bronchial responsiveness, two characteristic features of human asthma. We studied the effects of dsRNA, a nucleotide synthesized during viral replication, on airway inflammation and bronchial hyperresponsiveness in murine models of asthma. Intratracheal instillation of poly I:C, a synthetic dsRNA, increased the airway eosinophilia and enhanced bronchial hyperresponsiveness to methacholine in OVA-sensitized, exposed rats. These changes were associated with induction of cyclooxygenase-2 (COX-2) expression and COX-2-dependent PGD2 synthesis in the lungs, particularly in alveolar macrophages. The direct intratracheal instillation of PGD2 enhanced the eosinophilic inflammation in OVA-exposed animals, whereas pretreatment with a dual antagonist against the PGD2 receptor-(CRTH2) and the thromboxane A2 receptor, but not with a thromboxane A2 receptor-specific antagonist, nearly completely eliminated the dsRNA-induced worsening of airway inflammation and bronchial hyperresponsiveness. CRTH2-deficient mice had the same degree of allergen-induced airway eosinophilia as wild-type mice, but they did not exhibit a dsRNA-induced increase in eosinophil accumulation. Our data demonstrate that COX-2-dependent production of PGD2 followed by eosinophil recruitment into the airways via a CRTH2 receptor are the major pathogenetic factors responsible for the dsRNA-induced enhancement of airway inflammation and responsiveness.
Collapse
Affiliation(s)
- Yoshiki Shiraishi
- Division of Pulmonary Medicine, Department of Medicine, Shinanomachi Research Park, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Hansbro NG, Horvat JC, Wark PA, Hansbro PM. Understanding the mechanisms of viral induced asthma: new therapeutic directions. Pharmacol Ther 2008; 117:313-53. [PMID: 18234348 PMCID: PMC7112677 DOI: 10.1016/j.pharmthera.2007.11.002] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2007] [Accepted: 11/19/2007] [Indexed: 12/12/2022]
Abstract
Asthma is a common and debilitating disease that has substantially increased in prevalence in Western Societies in the last 2 decades. Respiratory tract infections by respiratory syncytial virus (RSV) and rhinovirus (RV) are widely implicated as common causes of the induction and exacerbation of asthma. These infections in early life are associated with the induction of wheeze that may progress to the development of asthma. Infections may also promote airway inflammation and enhance T helper type 2 lymphocyte (Th2 cell) responses that result in exacerbations of established asthma. The mechanisms of how RSV and RV induce and exacerbate asthma are currently being elucidated by clinical studies, in vitro work with human cells and animal models of disease. This research has led to many potential therapeutic strategies and, although none are yet part of clinical practise, they show much promise for the prevention and treatment of viral disease and subsequent asthma.
Collapse
Key Words
- aad, allergic airways disease
- ahr, airway hyperresponsiveness
- apc, antigen-presenting cell
- asm, airway smooth muscle
- balf, broncho-alveolar lavage fluid
- bec, bronchoepithelial cell
- bfgf, basic fibroblast growth factor
- cam, cellular adhesion molecules
- ccr, cc chemokine receptor
- cgrp, calcitonin gene-related peptide
- crp, c reactive protein
- dsrna, double stranded rna
- ecp, eosinophil cationic protein
- ena-78, epithelial neutrophil-activating peptide-78
- fev1, forced expiratory volume
- fi, formalin-inactivated
- g-csf and gm-csf, granulocyte and granulocyte-macrophage colony stimulating factor
- ics, inhaled corticosteroid
- ifn, interferon, ifn
- il, interleukin
- ip-10, ifn-γ inducible protein-10
- laba, long acting beta agonist
- ldh, lactate dehydrogenase
- ldlpr, low density lipoprotein receptor
- lrt, lower respiratory tract
- lt, leukotriene
- mab, monoclonal antibody
- mcp, monocyte chemoattractant proteins
- mdc, myeloid dendritic cell
- mhc, major histocompatibility
- mip, macrophage inhibitory proteins
- mpv, metapneumovirus
- nf-kb, nuclear factor (nf)-kb
- nk cells, natural killer cells
- nk1, neurogenic receptor 1
- or, odds ratio
- paf, platelet-activating factor
- pbmc, peripheral blood mononuclear cell
- pdc, plasmacytoid dendritic cell
- pef, peak expiratory flow
- penh, enhanced pause
- pfu, plaque forming units
- pg, prostaglandin
- pkr, protein kinase r
- pvm, pneumonia virus of mice
- rad, reactive airway disease
- rantes, regulated on activation normal t cell expressed and secreted
- rr, relative risk
- rsv, respiratory syncytial virus
- rv, rhinovirus (rv)
- ssrna, single stranded rna
- tgf, transforming growth factor
- th, t helper lymphocytes
- tlr, toll-like receptors
- tnf, tumor necrosis factor
- urt, upper respiratory tract
- vegf, vascular endothelial growth factor
- vs, versus
- wbc, white blood cell
- respiratory syncytial virus
- rhinovirus
- induction
- exacerbation
- asthma
- allergy
- treatment
- prevention
Collapse
Affiliation(s)
- Nicole G. Hansbro
- Priority Research Centre for Asthma and Respiratory Disease, Faculty of Health, The University of Newcastle, New South Wales 2308, Australia
- Vaccines, Immunology/Infection, Viruses and Asthma Group, Hunter Medical Research Institute, Locked Bag 1 New Lambton, New South Wales 2305, Australia
| | - Jay C. Horvat
- Priority Research Centre for Asthma and Respiratory Disease, Faculty of Health, The University of Newcastle, New South Wales 2308, Australia
- Vaccines, Immunology/Infection, Viruses and Asthma Group, Hunter Medical Research Institute, Locked Bag 1 New Lambton, New South Wales 2305, Australia
| | - Peter A. Wark
- Priority Research Centre for Asthma and Respiratory Disease, Faculty of Health, The University of Newcastle, New South Wales 2308, Australia
- Vaccines, Immunology/Infection, Viruses and Asthma Group, Hunter Medical Research Institute, Locked Bag 1 New Lambton, New South Wales 2305, Australia
- Department of Respiratory & Sleep Medicine, John Hunter Hospital & Sleep Medicine, School of Medical Practice, University of Newcastle, Newcastle, Australia
| | - Philip M. Hansbro
- Priority Research Centre for Asthma and Respiratory Disease, Faculty of Health, The University of Newcastle, New South Wales 2308, Australia
- Vaccines, Immunology/Infection, Viruses and Asthma Group, Hunter Medical Research Institute, Locked Bag 1 New Lambton, New South Wales 2305, Australia
| |
Collapse
|