151
|
Pathogenesis and prevention strategies of severe asthma exacerbations in children. Curr Opin Pulm Med 2016; 22:25-31. [PMID: 26574720 DOI: 10.1097/mcp.0000000000000223] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW Exacerbations of asthma in children are most frequently precipitated by respiratory infections with a seasonal pattern. However, management takes little account of the underlying infective or other precipitant abnormality. RECENT FINDINGS Interactions between environmental triggers, the airway microbiome and innate immune responses are key determinants of exacerbations. Elevated innate cytokines interleukin (IL)-33 and IL-25, and abnormal molecular responses in the interferon pathway are associated with rhinoviral infections. Exacerbations caused by fungal allergens also induce IL-33, highlighting this as an attractive therapeutic target. An equal contribution of bacterial and viral infection during exacerbations, particularly in preschool children, has become increasingly apparent, but some organisms may be protective. Investigation of mechanisms underlying infection-related exacerbations especially in preschool children is needed.Progressive loss of lung function from exacerbations is most pronounced in children aged 6-11 years, and low FEV1 is now recognized as a key predictor for the development of chronic obstructive pulmonary disease and premature death. Although prevention of exacerbations is critical, suboptimal patient education, prescription and adherence to maintenance therapy, and a lack of predictive biomarkers, remain key unaddressed issues in children. SUMMARY Precipitants and predictors of exacerbations, together with the child's age and clinical phenotype, need to be used to achieve individualized management in preference to the current uniform approach for all.
Collapse
|
152
|
Kumar RK, Herbert C, Foster PS. Mouse models of acute exacerbations of allergic asthma. Respirology 2016; 21:842-9. [PMID: 26922049 DOI: 10.1111/resp.12760] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 11/29/2015] [Accepted: 01/23/2016] [Indexed: 12/24/2022]
Abstract
Most of the healthcare costs associated with asthma relate to emergency department visits and hospitalizations because of acute exacerbations of underlying chronic disease. Development of appropriate animal models of acute exacerbations of asthma is a necessary prerequisite for understanding pathophysiological mechanisms and assessing potential novel therapeutic approaches. Most such models have been developed using mice. Relatively few mouse models attempt to simulate the acute-on-chronic disease that characterizes human asthma exacerbations. Instead, many reported models involve relatively short-term challenge with an antigen to which animals are sensitized, followed closely by an unrelated triggering agent, so are better described as models of potentiation of acute allergic inflammation. Triggers for experimental models of asthma exacerbations include (i) challenge with high levels of the sensitizing allergen (ii) infection by viruses or fungi, or challenge with components of these microorganisms (iii) exposure to environmental pollutants. In this review, we examine the strengths and weaknesses of published mouse models, their application for investigation of novel treatments and potential future developments.
Collapse
Affiliation(s)
- Rakesh K Kumar
- Department of Pathology, School of Medical Sciences, UNSW Australia, Sydney
| | - Cristan Herbert
- Department of Pathology, School of Medical Sciences, UNSW Australia, Sydney
| | - Paul S Foster
- Centre for Asthma and Respiratory Disease, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| |
Collapse
|
153
|
Mahmutovic Persson I, Akbarshahi H, Menzel M, Brandelius A, Uller L. Increased expression of upstream TH2-cytokines in a mouse model of viral-induced asthma exacerbation. J Transl Med 2016; 14:52. [PMID: 26879906 PMCID: PMC4754855 DOI: 10.1186/s12967-016-0808-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 02/04/2016] [Indexed: 12/17/2022] Open
Abstract
Background Exacerbations of asthma caused by respiratory viral infections are serious conditions in need of novel treatment. To this end animal models of asthma exacerbations are warranted. We have shown that dsRNA challenges or rhinoviral infection produce exacerbation effects in mice with ovalbumin (OVA)-induced allergic asthma. However, house dust mite (HDM) is a more human asthma-relevant allergen than OVA. We thus hypothesised that dsRNA challenges in mice with HDM-induced experimental asthma would produce important translational features of asthma exacerbations. Method Mouse airways were challenged locally with HDM or saline three times a week for three weeks to establish experimental asthma. Then daily local dsRNA challenges were given for three consecutive days to induce exacerbation. Bronchoalveolar lavage fluid (BALF) was analysed for inflammatory cells, total protein, the necrosis marker LDH and the alarmin ATP. Lung homogenates were analysed for mRNA expression (RT-qPCR) of TNF-α, CCL2, CCL5, IL-1β, IL-33, thymic stromal lymphopoietin (TSLP), and IL-25 as well as pattern recognition receptors (PRRs) RIG-I, MDA5 and TLR3. Lung tissue IL-33 was analysed with ELISA and PRRs were quantified by western blot. Immunohistochemistry indicated lung distribution of IL-33. Results HDM challenge alone caused sustained increase in BALF total protein, eosinophils, lymphocytes and neutrophils, and transient increase in lung tissue expression of TSLP, IL-33 and TNF-α. dsRNA-induced exacerbation markedly and dose-dependently exaggerated these effects. Further, BALF levels of LDH and ATP, and lung tissue expression of CCL2, CCL5, IL-1β, IL-25 and PRRs were increased exclusively at the exacerbations. Lung protein levels of IL-33 were transiently increased by HDM and further increased at exacerbation. Conclusion We demonstrate several novel aspects of HDM-induced experimental asthma and added exacerbation effects of dsRNA. General inflammatory parameters in BALF such as exuded proteins, mixed granulocytes, LDH and ATP were increased at the present exacerbations as they are in human asthma exacerbations. We suggest that this model of asthma exacerbation involving dsRNA challenges given to mice with established HDM-induced asthma has translational value and suggest that it may be particularly suited for in vivo studies involving pharmacological effects on exacerbation-induced expression of major upstream TH2-cytokines; IL-33, TSLP and IL-25, as well as PRRs. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-0808-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Irma Mahmutovic Persson
- Department Experimental Medical Science, Unit of Respiratory Immunopharmacology, Lund University, BMC D12, 221 84, Lund, Sweden.
| | - Hamid Akbarshahi
- Department Experimental Medical Science, Unit of Respiratory Immunopharmacology, Lund University, BMC D12, 221 84, Lund, Sweden.
| | - Mandy Menzel
- Department Experimental Medical Science, Unit of Respiratory Immunopharmacology, Lund University, BMC D12, 221 84, Lund, Sweden.
| | - Angelica Brandelius
- Department Experimental Medical Science, Unit of Respiratory Immunopharmacology, Lund University, BMC D12, 221 84, Lund, Sweden.
| | - Lena Uller
- Department Experimental Medical Science, Unit of Respiratory Immunopharmacology, Lund University, BMC D12, 221 84, Lund, Sweden.
| |
Collapse
|
154
|
Vries MD, Bedke N, Smithers NP, Loxham M, Howarth PH, Nawijn MC, Davies DE. Inhibition of Pim1 kinase, new therapeutic approach in virus-induced asthma exacerbations. Eur Respir J 2016; 47:783-91. [PMID: 26869670 DOI: 10.1183/13993003.00171-2015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 12/09/2015] [Indexed: 01/01/2023]
Abstract
Therapeutic options to treat virus-induced asthma exacerbations are limited and urgently needed. Therefore, we tested Pim1 kinase as potential therapeutic target in human rhinovirus (HRV) infections. We hypothesised that inhibition of Pim1 kinase reduces HRV replication by augmenting the interferon-induced anti-viral response due to increased activity of the janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway.Air-liquid interface (ALI) cultures of primary bronchial epithelial cells (PBECs) from healthy individuals and moderate-to-severe asthmatic volunteers were infected with HRV-16 with or without a specific Pim1 inhibitor; viral replication and induction of anti-viral responses were measured using RT-qPCR. Viral titres were measured by 50% tissue culture infective dose and release of interferon-γ-induced protein 10 (IP-10) and RANTES protein assessed by ELISA. Phosphorylation of STAT-1 was determined using western blotting.Viral replication was reduced in ALI cultures of healthy and asthmatic PBECs treated with the Pim1 inhibitor. Using cultures from healthy donors, enhanced STAT-1 phosphorylation upon inhibition of Pim1 kinase activity resulted in increased mRNA expression of interferon-β, interleukin-29, IP-10 and RANTES 12 h after infection and increased protein levels of IP-10 and RANTES 24 h after infection.We have identified Pim1 kinase as novel target to reduce viral replication in ALI cultures of PBECs. This may open new avenues for therapeutic interventions in virus-induced asthma exacerbations.
Collapse
Affiliation(s)
- Maaike de Vries
- University of Groningen, University Medical Center Groningen, Experimental Pulmonology and Inflammation Research, Dept of Pathology and Medical Biology, Groningen, The Netherlands Brooke Laboratory, Academic Unit of Clinical and Experimental Sciences, University of Southampton, Faculty of Medicine, University Hospital Southampton, Southampton, UK University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - Nicole Bedke
- Brooke Laboratory, Academic Unit of Clinical and Experimental Sciences, University of Southampton, Faculty of Medicine, University Hospital Southampton, Southampton, UK
| | - Natalie P Smithers
- Brooke Laboratory, Academic Unit of Clinical and Experimental Sciences, University of Southampton, Faculty of Medicine, University Hospital Southampton, Southampton, UK
| | - Matthew Loxham
- Brooke Laboratory, Academic Unit of Clinical and Experimental Sciences, University of Southampton, Faculty of Medicine, University Hospital Southampton, Southampton, UK
| | - Peter H Howarth
- Brooke Laboratory, Academic Unit of Clinical and Experimental Sciences, University of Southampton, Faculty of Medicine, University Hospital Southampton, Southampton, UK National Institute for Health Research, Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton, UK
| | - Martijn C Nawijn
- University of Groningen, University Medical Center Groningen, Experimental Pulmonology and Inflammation Research, Dept of Pathology and Medical Biology, Groningen, The Netherlands University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - Donna E Davies
- Brooke Laboratory, Academic Unit of Clinical and Experimental Sciences, University of Southampton, Faculty of Medicine, University Hospital Southampton, Southampton, UK National Institute for Health Research, Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton, UK
| |
Collapse
|
155
|
You MJ, Kim WK. Vitamin D serum levels and risk of asthma in children. ALLERGY ASTHMA & RESPIRATORY DISEASE 2016. [DOI: 10.4168/aard.2016.4.1.44] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Min Jung You
- Department of Pediatrics, Inje University Sanggyae Paik Hospital, Seoul, Korea
| | - Woo Kyung Kim
- Department of Pediatrics, Inje University Seoul Paik Hospital, Seoul, Korea
- Allergy and Respiratory Research Laboratory, Inje University College of Medicine, Seoul, Korea
| |
Collapse
|
156
|
Luo G, Stone BL, Fassl B, Maloney CG, Gesteland PH, Yerram SR, Nkoy FL. Predicting asthma control deterioration in children. BMC Med Inform Decis Mak 2015; 15:84. [PMID: 26467091 PMCID: PMC4607145 DOI: 10.1186/s12911-015-0208-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 10/07/2015] [Indexed: 11/17/2022] Open
Abstract
Background Pediatric asthma affects 7.1 million American children incurring an annual total direct healthcare cost around 9.3 billion dollars. Asthma control in children is suboptimal, leading to frequent asthma exacerbations, excess costs, and decreased quality of life. Successful prediction of risk for asthma control deterioration at the individual patient level would enhance self-management and enable early interventions to reduce asthma exacerbations. We developed and tested the first set of models for predicting a child’s asthma control deterioration one week prior to occurrence. Methods We previously reported validation of the Asthma Symptom Tracker, a weekly asthma self-monitoring tool. Over a period of two years, we used this tool to collect a total of 2912 weekly assessments of asthma control on 210 children. We combined the asthma control data set with patient attributes and environmental variables to develop machine learning models to predict a child’s asthma control deterioration one week ahead. Results Our best model achieved an accuracy of 71.8 %, a sensitivity of 73.8 %, a specificity of 71.4 %, and an area under the receiver operating characteristic curve of 0.757. We also identified potential improvements to our models to stimulate future research on this topic. Conclusions Our best model successfully predicted a child’s asthma control level one week ahead. With adequate accuracy, the model could be integrated into electronic asthma self-monitoring systems to provide real-time decision support and personalized early warnings of potential asthma control deteriorations.
Collapse
Affiliation(s)
- Gang Luo
- Department of Biomedical Informatics, University of Utah, Suite 140, 421 Wakara Way, Salt Lake City, UT, 84108, USA.
| | - Bryan L Stone
- Department of Pediatrics, University of Utah, 100 N Mario Capecchi Drive, Salt Lake City, UT, 84113, USA
| | - Bernhard Fassl
- Department of Pediatrics, University of Utah, 100 N Mario Capecchi Drive, Salt Lake City, UT, 84113, USA
| | - Christopher G Maloney
- Department of Pediatrics, University of Utah, 100 N Mario Capecchi Drive, Salt Lake City, UT, 84113, USA
| | - Per H Gesteland
- Department of Pediatrics, University of Utah, 100 N Mario Capecchi Drive, Salt Lake City, UT, 84113, USA
| | - Sashidhar R Yerram
- Department of Biomedical Informatics, University of Utah, Suite 140, 421 Wakara Way, Salt Lake City, UT, 84108, USA
| | - Flory L Nkoy
- Department of Pediatrics, University of Utah, 100 N Mario Capecchi Drive, Salt Lake City, UT, 84113, USA
| |
Collapse
|
157
|
Herbert C, Sebesfi M, Zeng QX, Oliver BG, Foster PS, Kumar RK. Using multiple online databases to help identify microRNAs regulating the airway epithelial cell response to a virus-like stimulus. Respirology 2015; 20:1206-12. [PMID: 26289417 DOI: 10.1111/resp.12606] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 04/24/2015] [Accepted: 06/05/2015] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND OBJECTIVE Exacerbations of allergic asthma are often triggered by respiratory viral infections. We have previously shown that in a T-helper type 2 (Th2)-biased cytokine environment, mouse and human airway epithelial cells (AEC) exhibit increased expression of pro-inflammatory and anti-viral genes in response to synthetic double-stranded ribonucleic acid (dsRNA), a virus-like stimulus. This implies coordinated regulation of gene expression, suggesting possible involvement of microRNA. To investigate this, we developed a novel approach to identifying candidate microRNA using online databases, then confirmed their expression by quantitative real-time polymerase chain reaction (qRT-PCR). METHODS Using a list of genes of interest, defined on the basis of the previous study as being up-regulated in a Th2 environment, we searched mouse and human microRNA databases for possible regulatory microRNA, and selected 10 candidates that were conserved across species or predicted by more than one human database. Expression of these microRNA was tested by qRT-PCR, in primary human AEC pre-treated with Th2 cytokines and exposed to dsRNA. RESULTS Expression of hsa-miR-139-5p, miR-423-5p and miR-542-3p was significantly decreased in Th2 pre-treated AEC, and miR-135a-5p exhibited a trend towards decreased expression. Further database searches confirmed that these microRNA regulated additional pro-inflammatory and anti-viral response genes for which expression had previously been shown to be up-regulated, confirming the validity of this approach. CONCLUSIONS Our study demonstrates the value of using multiple online databases to identify candidate regulatory microRNA and provides the first evidence that in an allergic environment, microRNA may be important in altering the pro-inflammatory and anti-viral responses of human AEC during exacerbations of asthma.
Collapse
Affiliation(s)
- Cristan Herbert
- Department of Pathology, School of Medical Sciences, UNSW Australia, Sydney, New South Wales, Australia
| | - Michelle Sebesfi
- Department of Pathology, School of Medical Sciences, UNSW Australia, Sydney, New South Wales, Australia
| | - Qing-Xiang Zeng
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia.,Otorhinolaryngology Hospital, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Brian G Oliver
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia.,School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Paul S Foster
- Centre for Asthma and Respiratory Disease, University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Rakesh K Kumar
- Department of Pathology, School of Medical Sciences, UNSW Australia, Sydney, New South Wales, Australia
| |
Collapse
|
158
|
Prasetyo AA, Desyardi MN, Tanamas J, Suradi, Reviono, Harsini, Kageyama S, Chikumi H, Shimizu E. Respiratory viruses and torque teno virus in adults with acute respiratory infections. Intervirology 2015; 58:57-68. [PMID: 25890989 PMCID: PMC7179541 DOI: 10.1159/000369211] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Objective To define the molecular epidemiology of respiratory viral infections in adult patients. Methods Nasal and throat swabs were collected from all adult patients with influenza-like illness (ILI), acute respiratory infection (ARI), or severe ARI (SARI) admitted to a tertiary hospital in Surakarta, Indonesia, between March 2010 and April 2011 and analyzed for 19 respiratory viruses and for torque teno virus (TTV) and human gyrovirus (HGyV). Results Respiratory viruses were detected in 61.3% of the subjects, most of whom had ARI (90.8%, OR = 11.39), were hospitalized (96.9%, OR = 22.31), had asthma exacerbation (90.9%, OR = 8.67), and/or had pneumonia (80%, OR = 4.0). Human rhinovirus (HRV) A43 predominated. Influenza A H3N2, human metapneumovirus (HMPV) subtypes A1 and A2, the influenza B virus, human adenovirus B, and human coronavirus OC43 were also detected. All respiratory viruses were detected in the transition month between the rainy and dry seasons. No mixed respiratory virus infection was found. Coinfections of the influenza A H3N2 virus with TTV, HMPV with TTV, HRV with TTV, and human parainfluenza virus-3 with TTV were found in 4.7, 2.8, 19.8, and 0.9% of the samples, respectively. Conclusions This study highlights the need to perform routine detection of respiratory viruses in adults hospitalized with ARI, asthma exacerbation, and/or pneumonia.
Collapse
Affiliation(s)
- Afiono Agung Prasetyo
- Department of Microbiology, Faculty of Medicine, Sebelas Maret University, Jl. Ir. Sutami No. 36A, Surakarta 57126 (Indonesia). afie.agp.la @ gmail.com
| | | | | | | | | | | | | | | | | |
Collapse
|
159
|
Cheng G, Smith AM, Levin L, Epstein T, Ryan PH, LeMasters GK, Khurana Hershey GK, Reponen T, Villareal M, Lockey J, Bernstein DI. Duration of day care attendance during infancy predicts asthma at the age of seven: the Cincinnati Childhood Allergy and Air Pollution Study. Clin Exp Allergy 2015; 44:1274-81. [PMID: 25179746 DOI: 10.1111/cea.12397] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 06/11/2014] [Accepted: 07/28/2014] [Indexed: 12/25/2022]
Abstract
BACKGROUND Studies vary with respect to the reported effects of day care attendance on childhood asthma. OBJECTIVES To evaluate the independent and combined effects of day care attendance and respiratory infections on the development of asthma at the age of seven in a prospective birth cohort. METHOD At the age of seven, the study sample included 589 children with complete data of 762 enrolled at birth. Day care hours and number of respiratory infections were reported in follow-up questionnaires through age four. At 7 years of age, asthma was diagnosed in 95 children (16%), based on predefined symptoms criteria confirmed by either asthma FEV1 reversibility after bronchodilator or a positive methacholine test (PC20 ≤ 4 mg/mL). Logistic regression was used to investigate the relationships between asthma at the age of seven, cumulative hours of day care attendance and reported respiratory infections at ages 1-4. RESULTS In the univariate analyses, day care attendance at 12 months was associated with an increased risk of asthma [odds ratio (OR) = 1.8, 95% confidence interval (CI) = 1.1-3.0]. Both upper and lower respiratory infections at 12 months also increased the likelihood of asthma [OR = 2.4 (1.4-4.1); OR = 2.3 (1.5-3.7), respectively]. In the final multivariate logistic model, cumulative hours of day care attendance and number of lower respiratory infections at 12 months were associated with asthma [OR = 1.2 (1.1-1.5); OR = 1.4 (1.2-1.7), respectively]. However, a threshold of greater than 37.5 hours per week of day care attendance was associated with a lower risk of asthma [OR = 0.6 (0.4-0.9)]. CONCLUSION Depending on duration of attendance, day care during infancy can either increase or reduce risk of asthma at the age of seven.
Collapse
Affiliation(s)
- G Cheng
- Division of Immunology, Allergy and Rheumatology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
160
|
Lacroix C, Laconi S, Angius F, Coluccia A, Silvestri R, Pompei R, Neyts J, Leyssen P. In vitro characterisation of a pleconaril/pirodavir-like compound with potent activity against rhinoviruses. Virol J 2015; 12:106. [PMID: 26169596 PMCID: PMC4501209 DOI: 10.1186/s12985-015-0330-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 06/24/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Rhinovirus infections do not only cause common colds, but may also trigger severe exacerbations of asthma and chronic obstructive pulmonary disease (COPD). Even though rhinoviruses have been the focus of extensive drug development efforts in the past, an anti-rhinoviral drug still has to make it to the market. In the past, the viral capsid protein VP1 has been shown to be an important target for the development of antiviral molecules. Furthermore, many different chemical scaffolds appear to possess the properties that are required to inhibit virus replication by this mechanism of action. I-6602, an analogue of the rhinovirus inhibitor pirodavir, was previously identified as a potent inhibitor of rhinovirus infection. Here, we describe the antiviral activity of its analogue ca603, a molecule with a modified linker structure, and corroborate its mechanism of action as a capsid binder. FINDINGS The molecule ca603 shows antiviral activity against a panel of rhino-and enteroviruses. Cross-resistance is observed against viruses with mutations that render them resistant to the inhibitory effect of the capsid binder pleconaril and thermostability assays demonstrate that the compound binds and stabilizes the viral capsid. Binding of the molecule to the VP1 protein is corroborated by in silico modeling. CONCLUSIONS It is confirmed that ca603 inhibits rhinovirus replication by interaction with the VP1 protein and, by this, allows to further expand the chemical diversity of capsid-binding molecules.
Collapse
Affiliation(s)
- Céline Lacroix
- Department of Microbiology and Immunology, Laboratory for Virology and Chemotherapy, KU Leuven, Rega Institute for Medical Research, B-3000, Leuven, Belgium.
| | - Samuela Laconi
- Department of Biomedical Sciences, University of Cagliari, I-09124, Cagliari, Italy.
| | - Fabrizio Angius
- Department of Biomedical Sciences, University of Cagliari, I-09124, Cagliari, Italy.
| | - Antonio Coluccia
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, Piazzale Aldo Moro 5, I-00185, Rome, Italy.
| | - Romano Silvestri
- Istituto Pasteur-Fondazione Cenci Bolognetti, Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, Piazzale Aldo Moro 5, I-00185, Rome, Italy.
| | - Raffaello Pompei
- Department of Biomedical Sciences, University of Cagliari, I-09124, Cagliari, Italy.
| | - Johan Neyts
- Department of Microbiology and Immunology, Laboratory for Virology and Chemotherapy, KU Leuven, Rega Institute for Medical Research, B-3000, Leuven, Belgium.
| | - Pieter Leyssen
- Department of Microbiology and Immunology, Laboratory for Virology and Chemotherapy, KU Leuven, Rega Institute for Medical Research, B-3000, Leuven, Belgium.
| |
Collapse
|
161
|
Abstract
The 2nd Cross Company Respiratory Symposium (CCRS), held in Horsham, U.K. in 2012, brought together representatives from across the pharmaceutical industry with expert academics, in the common interest of improving the design and translational predictiveness of in vivo models of respiratory disease. Organized by the respiratory representatives of the European Federation of Pharmaceutical Industries and Federations (EFPIA) group of companies involved in the EU-funded project (U-BIOPRED), the aim of the symposium was to identify state-of-the-art improvements in the utility and design of models of respiratory disease, with a view to improving their translational potential and reducing wasteful animal usage. The respiratory research and development community is responding to the challenge of improving translation in several ways: greater collaboration and open sharing of data, careful selection of the species, complexity and chronicity of the models, improved practices in preclinical research, continued refinement in models of respiratory diseases and their sub-types, greater understanding of the biology underlying human respiratory diseases and their sub-types, and finally greater use of human (and especially disease-relevant) cells, tissues and explants. The present review highlights these initiatives, combining lessons from the symposium and papers published in Clinical Science arising from the symposium, with critiques of the models currently used in the settings of asthma, idiopathic pulmonary fibrosis and COPD. The ultimate hope is that this will contribute to a more rational, efficient and sustainable development of a range of new treatments for respiratory diseases that continue to cause substantial morbidity and mortality across the world.
Collapse
|
162
|
Eosinophil-Derived Neurotoxin (EDN/RNase 2) and the Mouse Eosinophil-Associated RNases (mEars): Expanding Roles in Promoting Host Defense. Int J Mol Sci 2015; 16:15442-55. [PMID: 26184157 PMCID: PMC4519907 DOI: 10.3390/ijms160715442] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 06/18/2015] [Accepted: 06/30/2015] [Indexed: 12/30/2022] Open
Abstract
The eosinophil-derived neurotoxin (EDN/RNase2) and its divergent orthologs, the mouse eosinophil-associated RNases (mEars), are prominent secretory proteins of eosinophilic leukocytes and are all members of the larger family of RNase A-type ribonucleases. While EDN has broad antiviral activity, targeting RNA viruses via mechanisms that may require enzymatic activity, more recent studies have elucidated how these RNases may generate host defense via roles in promoting leukocyte activation, maturation, and chemotaxis. This review provides an update on recent discoveries, and highlights the versatility of this family in promoting innate immunity.
Collapse
|
163
|
Ota K, Kawaguchi M, Fujita J, Kokubu F, Huang SK, Morishima Y, Matsukura S, Kurokawa M, Ishii Y, Satoh H, Sakamoto T, Hizawa N. Synthetic double-stranded RNA induces interleukin-32 in bronchial epithelial cells. Exp Lung Res 2015; 41:335-43. [DOI: 10.3109/01902148.2015.1033569] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
164
|
|
165
|
Gillissen A, Paparoupa M. Inflammation and infections in asthma. THE CLINICAL RESPIRATORY JOURNAL 2015; 9:257-69. [PMID: 24725460 PMCID: PMC7162380 DOI: 10.1111/crj.12135] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 04/26/2014] [Accepted: 04/04/2014] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Asthma is driven by an inflammatory response against normally harmless environmental inorganic and organic compounds in the respiratory tract. Immune responses to airborne pathogens such as viruses and bacteria may reduce the allergic responses but are also known to trigger asthma attacks and eventually lead to severe disease condition. OBJECTIVE To investigate the role of respiratory pathogens concerning the induction or protection against acute or chronic asthma manifestations. METHODS We included 131 articles for the final review according to their relevance with the subject. RESULTS There is apparently contradictory interaction of respiratory germs in the airways of asthmatics which may be protective on one angle but deleterious on the other. CONCLUSION The relationship between inflammation and remodeling and the pathogenic role of viral and bacterial infection in the airways of asthmatic patients is still highly debatable and incompletely understood.
Collapse
Affiliation(s)
- Adrian Gillissen
- Department of Pulmonary MedicineGeneral Hospital KasselKasselGermany
| | - Maria Paparoupa
- Department of Pulmonary MedicineGeneral Hospital KasselKasselGermany
| |
Collapse
|
166
|
Bernard A, Lacroix C, Cabiddu MG, Neyts J, Leyssen P, Pompei R. Exploration of the anti-enterovirus activity of a series of pleconaril/pirodavir-like compounds. Antivir Chem Chemother 2015; 24:56-61. [PMID: 26071135 DOI: 10.1177/2040206615589035] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND The Enterovirus genus of the Picornaviridae is represented by several viral pathogens that are associated with human disease, namely Poliovirus 1, Enterovirus 71 and Rhinoviruses. Enterovirus 71 has been associated with encephalitis, while Rhinoviruses are a major cause of asthma exacerbations and chronic obstructive pulmonary disease. Based on the structure of both pleconaril and pirodavir, we previously synthesized some original compounds as potential inhibitors of Rhinovirus replication. METHODS These compounds were explored for in vitro antiviral potential on other human pathogenic Enteroviruses, namely Enterovirus 71 on rhabdo-myosarcoma cells, Coxsackievirus B3 on Vero cells, Poliovirus 1 and Echovirus 11 on BGM cells. RESULTS Activity was confirmed for compound against Rhinovirus 14. Furthermore, few compounds showed a cell-protective effect on Enterovirus 71, presented a marked improvement as compared to the reference drug pleconaril for inhibitory activity on both Enterovirus 71 and Poliovirus 1. The most striking observation was the clear cell protective effect for the set of analogues in a virus-cell-based assay for Echovirus 11 with an effective concentration (EC50) as low as 0.3 µM (Selectivity index or SI = 483), and selectivity indexes greater than 857 (EC50 = 0.6 µM) and 1524 (EC50 = 0.33 µM). CONCLUSION Some of the evaluated compounds showed potent and selective antiviral activity against several enterovirus species, such as Enterovirus 71 (EV-A), Echovirus 11 (EV-B), and Poliovirus 1 (EV-C). This could be used as a starting point for the development of other pleconaril/pirodavir-like enterovirus inhibitors with broad-spectrum activity and improved effects as compared to the reference drugs.
Collapse
Affiliation(s)
- Angela Bernard
- Department of Chemical and Geological Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Monserrato (CA), Italy
| | - Céline Lacroix
- Department of Microbiology and Immunology, Laboratory for Virology and Chemotherapy, KU Leuven - University of Leuven, Rega Institute for Medical Research, Leuven, Belgium
| | - Maria G Cabiddu
- Department of Chemical and Geological Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Monserrato (CA), Italy
| | - Johan Neyts
- Department of Microbiology and Immunology, Laboratory for Virology and Chemotherapy, KU Leuven - University of Leuven, Rega Institute for Medical Research, Leuven, Belgium
| | - Pieter Leyssen
- Department of Microbiology and Immunology, Laboratory for Virology and Chemotherapy, KU Leuven - University of Leuven, Rega Institute for Medical Research, Leuven, Belgium
| | - Raffaello Pompei
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| |
Collapse
|
167
|
Affiliation(s)
- Ian Pavord
- Respiratory Medicine Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Andrew Bush
- Paediatric Respiratory Medicine and Paediatrics, Royal Brompton Hospital and Imperial College, London, UK
| |
Collapse
|
168
|
Kim SH, Lim KH, Park HK, Lee SY, Kim SH, Kang HR, Park HW, Chang YS, Cho SH. Reduced IRF7 response to rhinovirus unrelated with DNA methylation in peripheral mononuclear cells of adult asthmatics. Asia Pac Allergy 2015; 5:114-22. [PMID: 25938076 PMCID: PMC4415177 DOI: 10.5415/apallergy.2015.5.2.114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Accepted: 04/12/2015] [Indexed: 02/04/2023] Open
Abstract
Background Human rhinoviruses are the major cause of asthma exacerbation in both children and adults. Recently, impaired antiviral interferon (IFN) response in asthmatics has been indicated as a primary reason of the susceptibility to respiratory virus, but the mechanism of defective IFN production is little understood to date. The expression of IFN regulatory factor 7 (IRF7), a transcriptional factor for virus-induced type I IFN production is known to be regulated epigenetically by DNA methylation. Objective We aimed to investigate the expression of IFN-α, IFN-β, and IRF7 in response to rhinovirus infection in the adult asthmatics and evaluate DNA methylation status of IRF7 gene promotor. Methods Twenty symptomatic adult asthmatics and 10 healthy subjects were enrolled and peripheral blood was collected from each subject. Peripheral blood mononuclear cells (PBMCs) were isolated, cultured, and ex vivo stimulated with rhinovirus-16. The mRNA expressions of IFN-α, IFN-β, and IRF7 were analyzed using real time quantitative polymerase chain reaction. Genomic DNA was isolated from untreated PBMCs and the methylation status of IRF7 gene promotor was investigated using bisulfite pyrosequencing. Results The mean age of asthmatics was 45.4 ± 15.7 years and 40% was male, which were not different with those of control group. Asthmatics showed significantly decreased mRNA expressions (relative expression to beta-actin) of IFN-α and IFN-β compared with normal control. The mRNA expression of IRF7 in the asthmatics was also significantly lower than those in the normal control. No significant difference of DNA methylation was observed between asthmatics and controls in all analyzed positions of IRF7 promotor CpG loci. Conclusion The mRNA expression of type I IFN in response to rhinovirus was impaired in the PBMCs of adult asthmatics. The mRNA expression of IRF7, transcriptional factor inducing type I IFN was also reduced, but not caused by altered DNA methylation in the IRF7 gene promotor.
Collapse
Affiliation(s)
- Sae-Hoon Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-899, Korea. ; Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 110-899, Korea. ; Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 463-707, Korea
| | - Kyung-Hwan Lim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-899, Korea. ; Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 110-899, Korea
| | - Han-Ki Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-899, Korea. ; Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 110-899, Korea
| | - Suh-Young Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-899, Korea. ; Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 110-899, Korea. ; Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 463-707, Korea
| | - Soon-Hee Kim
- Biomedical Research Institute, Seoul National University Bundang Hospital, Seongnam 463-707, Korea
| | - Hye-Ryun Kang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-899, Korea. ; Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 110-899, Korea
| | - Heung-Woo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-899, Korea. ; Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 110-899, Korea
| | - Yoon-Seok Chang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-899, Korea. ; Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 110-899, Korea. ; Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 463-707, Korea
| | - Sang-Heon Cho
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-899, Korea. ; Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 110-899, Korea
| |
Collapse
|
169
|
Tanaka S, Tamari M, Nakayama T, Ishii H, Hatsushika K, Hayashi A, Watanabe H, Kanai M, Osano M, Yonaga T, Tomita K, Fujieda S, Sakuma Y, Shiono O, Ishitoya J, Masuyama K, Hirota T. Synergistic suppression of Poly(I:C)-induced CCL3 by a corticosteroid and a long acting β2 agonist in nasal epithelial cells. Allergol Int 2015; 64:196-9. [PMID: 25838100 DOI: 10.1016/j.alit.2014.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 10/03/2014] [Accepted: 10/13/2014] [Indexed: 11/18/2022] Open
Affiliation(s)
- Shota Tanaka
- Laboratory for Respiratory and Allergic Diseases, Center for Integrative Medical Sciences, The Institute of Physical and Chemical Research (RIKEN), Kanagawa, Japan; Department of Otolaryngology-Head and Neck Surgery, University of Yamanashi, Faculty of Medicine, Yamanashi, Japan
| | - Mayumi Tamari
- Laboratory for Respiratory and Allergic Diseases, Center for Integrative Medical Sciences, The Institute of Physical and Chemical Research (RIKEN), Kanagawa, Japan.
| | - Tsuguhisa Nakayama
- Laboratory for Respiratory and Allergic Diseases, Center for Integrative Medical Sciences, The Institute of Physical and Chemical Research (RIKEN), Kanagawa, Japan; Department of Otorhinolaryngology, Jikei University School of Medicine, Tokyo, Japan
| | - Hiroki Ishii
- Department of Otolaryngology-Head and Neck Surgery, University of Yamanashi, Faculty of Medicine, Yamanashi, Japan
| | - Kyosuke Hatsushika
- Department of Otolaryngology-Head and Neck Surgery, University of Yamanashi, Faculty of Medicine, Yamanashi, Japan
| | - Akira Hayashi
- Department of Otolaryngology-Head and Neck Surgery, University of Yamanashi, Faculty of Medicine, Yamanashi, Japan
| | - Hiroyuki Watanabe
- Department of Otolaryngology-Head and Neck Surgery, University of Yamanashi, Faculty of Medicine, Yamanashi, Japan
| | - Mari Kanai
- Department of Otolaryngology-Head and Neck Surgery, University of Yamanashi, Faculty of Medicine, Yamanashi, Japan
| | - Masashi Osano
- Department of Otolaryngology-Head and Neck Surgery, University of Yamanashi, Faculty of Medicine, Yamanashi, Japan
| | - Takaaki Yonaga
- Department of Otolaryngology-Head and Neck Surgery, University of Yamanashi, Faculty of Medicine, Yamanashi, Japan
| | - Kaori Tomita
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Fukui, Faculty of Medicine, Fukui, Japan
| | - Shigeharu Fujieda
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Fukui, Faculty of Medicine, Fukui, Japan
| | - Yasunori Sakuma
- Department of Otorhinolaryngology, Yokohama City University Medical Center, Kanagawa, Japan
| | - Osamu Shiono
- Department of Otorhinolaryngology, Yokohama City University Medical Center, Kanagawa, Japan
| | - Junichi Ishitoya
- Department of Otorhinolaryngology, Yokohama City University Medical Center, Kanagawa, Japan
| | - Keisuke Masuyama
- Department of Otolaryngology-Head and Neck Surgery, University of Yamanashi, Faculty of Medicine, Yamanashi, Japan
| | - Tomomitsu Hirota
- Laboratory for Respiratory and Allergic Diseases, Center for Integrative Medical Sciences, The Institute of Physical and Chemical Research (RIKEN), Kanagawa, Japan
| |
Collapse
|
170
|
Annoni R, Silva LFF, Nussbaumer-Ochsner Y, van Schadewijk A, Mauad T, Hiemstra PS, Rabe KF. Increased expression of granzymes A and B in fatal asthma. Eur Respir J 2015; 45:1485-8. [DOI: 10.1183/09031936.00213814] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 01/11/2015] [Indexed: 11/05/2022]
|
171
|
Jackson DJ, Makrinioti H, Rana BMJ, Shamji BWH, Trujillo-Torralbo MB, Footitt J, Jerico Del-Rosario, Telcian AG, Nikonova A, Zhu J, Aniscenko J, Gogsadze L, Bakhsoliani E, Traub S, Dhariwal J, Porter J, Hunt D, Hunt T, Hunt T, Stanciu LA, Khaitov M, Bartlett NW, Edwards MR, Kon OM, Mallia P, Papadopoulos NG, Akdis CA, Westwick J, Edwards MJ, Cousins DJ, Walton RP, Johnston SL. IL-33-dependent type 2 inflammation during rhinovirus-induced asthma exacerbations in vivo. Am J Respir Crit Care Med 2015; 190:1373-82. [PMID: 25350863 DOI: 10.1164/rccm.201406-1039oc] [Citation(s) in RCA: 463] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Rhinoviruses are the major cause of asthma exacerbations; however, its underlying mechanisms are poorly understood. We hypothesized that the epithelial cell-derived cytokine IL-33 plays a central role in exacerbation pathogenesis through augmentation of type 2 inflammation. OBJECTIVES To assess whether rhinovirus induces a type 2 inflammatory response in asthma in vivo and to define a role for IL-33 in this pathway. METHODS We used a human experimental model of rhinovirus infection and novel airway sampling techniques to measure IL-4, IL-5, IL-13, and IL-33 levels in the asthmatic and healthy airways during a rhinovirus infection. Additionally, we cultured human T cells and type 2 innate lymphoid cells (ILC2s) with the supernatants of rhinovirus-infected bronchial epithelial cells (BECs) to assess type 2 cytokine production in the presence or absence of IL-33 receptor blockade. MEASUREMENTS AND MAIN RESULTS IL-4, IL-5, IL-13, and IL-33 are all induced by rhinovirus in the asthmatic airway in vivo and relate to exacerbation severity. Further, induction of IL-33 correlates with viral load and IL-5 and IL-13 levels. Rhinovirus infection of human primary BECs induced IL-33, and culture of human T cells and ILC2s with supernatants of rhinovirus-infected BECs strongly induced type 2 cytokines. This induction was entirely dependent on IL-33. CONCLUSIONS IL-33 and type 2 cytokines are induced during a rhinovirus-induced asthma exacerbation in vivo. Virus-induced IL-33 and IL-33-responsive T cells and ILC2s are key mechanistic links between viral infection and exacerbation of asthma. IL-33 inhibition is a novel therapeutic approach for asthma exacerbations.
Collapse
Affiliation(s)
- David J Jackson
- 1 Airway Disease Infection Section, National Heart & Lung Institute, Imperial College London, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
172
|
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
|
173
|
Abstract
Patients with severe asthma or COPD have often a suboptimal symptom control due to inadequate treatment. A better understanding of pathogenetic mechanisms, phenotypes, endotypes and the new technologies available in the fields of molecular biology and immunogenetics have made it possible to synthesize specific monoclonal antibodies virtually able to interact with any target antigen, or to open a way for new therapeutic target options. At the moment, the only biologic drug available in clinical practice is omalizumab. To overcome the limits of omalizumab, the research has focused on new monoclonal antibodies presenting higher avidity for IgE (e.g. ligelizumab and lumiximab) and ability to interact also with low affinity IgE receptor (FcϵRII). At present, many new biological drugs with different mechanisms of action and targets are matter of research. It is very important to identify the asthmatic phenotype in order to select the most appropriate drug for the individual patient. The most promising agents are targeted against cytokines of Th2 pattern and related receptors, such as IL-2 (daclizumab) and IL-13 (lebrikizumab) or IL-5 in patients with hypereosinophilia (mepolizumab, reslizumab and benralizumab). Other interesting drugs have as a target TNF-α or its soluble receptor (infliximab, golimumab and etanercept) or IL-1 (canakinumab), a cytokine with an important systemic proinflammatory action. Finally, the discovery of increased levels of C5a in the airways of asthmatic patients has led to the synthesis of a specific monoclonal antibody (eculizumab). Further help should come from the identification of biomarkers that can guide in choosing the best treatment for the individual patient, such as IgE for omalizumab or periostin for lebrikizumab.
Collapse
|
174
|
Schwantes EA, Manthei DM, Denlinger LC, Evans MD, Gern JE, Jarjour NN, Mathur SK. Interferon gene expression in sputum cells correlates with the Asthma Index Score during virus-induced exacerbations. Clin Exp Allergy 2015; 44:813-21. [PMID: 24450586 PMCID: PMC4037351 DOI: 10.1111/cea.12269] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 11/20/2013] [Accepted: 01/01/2014] [Indexed: 01/17/2023]
Abstract
Background The majority of asthma exacerbations are related to viral respiratory infections. Some, but not all, previous studies have reported that low interferon responses in patients with asthma increase the risk for virus‐induced exacerbations. Objective We sought to determine the relationship between lower airway inflammatory biomarkers, specifically interferon gene expression, and the severity or presence of an exacerbation in asthmatics experiencing a naturally occurring viral infection. Methods Sputum samples were analysed from subjects in an asthma exacerbation study who experienced a confirmed viral infection. Subjects were monitored for daily symptoms, medication use and peak expiratory flow rate until baseline. Sputum samples were assessed for cell counts and gene expression. Results Interferon gamma expression was significantly greater in patients with asthma exacerbations compared to non‐exacerbating patients (P = 0.002). IFN‐α1, IFN‐β1 and IFN‐γ mRNA levels correlated with the peak Asthma Index (r = 0.58, P < 0.001; r = 0.57, P = 0.001; and r = 0.51, P = 0.004, respectively). Additionally, IL‐13, IL‐10 and eosinophil major basic protein mRNA levels were greater in patients with asthma exacerbations compared to non‐exacerbating patients (P = 0.03, P = 0.06 and P = 0.02, respectively), and IL‐13 mRNA correlated with the peak Asthma Index (P = 0.006). Conclusions Our findings indicate that asthma exacerbations are associated with increased rather than decreased expression of interferons early in the course of infection. These findings raise the possibility that excessive virus‐induced interferon production during acute infections can contribute to airway inflammation and exacerbations of asthma.
Collapse
Affiliation(s)
- E A Schwantes
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | | | | | | | | | | |
Collapse
|
175
|
Kanchongkittiphon W, Mendell MJ, Gaffin JM, Wang G, Phipatanakul W. Indoor environmental exposures and exacerbation of asthma: an update to the 2000 review by the Institute of Medicine. ENVIRONMENTAL HEALTH PERSPECTIVES 2015; 123:6-20. [PMID: 25303775 PMCID: PMC4286274 DOI: 10.1289/ehp.1307922] [Citation(s) in RCA: 243] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 10/09/2014] [Indexed: 05/18/2023]
Abstract
BACKGROUND Previous research has found relationships between specific indoor environmental exposures and exacerbation of asthma. OBJECTIVES In this review we provide an updated summary of knowledge from the scientific literature on indoor exposures and exacerbation of asthma. METHODS Peer-reviewed articles published from 2000 to 2013 on indoor exposures and exacerbation of asthma were identified through PubMed, from reference lists, and from authors' files. Articles that focused on modifiable indoor exposures in relation to frequency or severity of exacerbation of asthma were selected for review. Research findings were reviewed and summarized with consideration of the strength of the evidence. RESULTS Sixty-nine eligible articles were included. Major changed conclusions include a causal relationship with exacerbation for indoor dampness or dampness-related agents (in children); associations with exacerbation for dampness or dampness-related agents (in adults), endotoxin, and environmental tobacco smoke (in preschool children); and limited or suggestive evidence for association with exacerbation for indoor culturable Penicillium or total fungi, nitrogen dioxide, rodents (nonoccupational), feather/down pillows (protective relative to synthetic bedding), and (regardless of specific sensitization) dust mite, cockroach, dog, and dampness-related agents. DISCUSSION This review, incorporating evidence reported since 2000, increases the strength of evidence linking many indoor factors to the exacerbation of asthma. Conclusions should be considered provisional until all available evidence is examined more thoroughly. CONCLUSION Multiple indoor exposures, especially dampness-related agents, merit increased attention to prevent exacerbation of asthma, possibly even in nonsensitized individuals. Additional research to establish causality and evaluate interventions is needed for these and other indoor exposures.
Collapse
|
176
|
Cheah EY, Mann TS, Burcham PC, Henry PJ. Influenza A infection attenuates relaxation responses of mouse tracheal smooth muscle evoked by acrolein. Biochem Pharmacol 2014; 93:519-26. [PMID: 25557294 DOI: 10.1016/j.bcp.2014.12.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 12/17/2014] [Accepted: 12/23/2014] [Indexed: 10/24/2022]
Abstract
The airway epithelium is an important source of relaxant mediators, and damage to the epithelium caused by respiratory tract viruses may contribute to airway hyperreactivity. The aim of this study was to determine whether influenza A-induced epithelial damage would modulate relaxation responses evoked by acrolein, a toxic and prevalent component of smoke. Male BALB/c mice were inoculated intranasally with influenza A/PR-8/34 (VIRUS-infected) or allantoic fluid (SHAM-infected). On day 4 post-inoculation, isometric tension recording studies were conducted on carbachol pre-contracted tracheal segments isolated from VIRUS and SHAM mice. Relaxant responses to acrolein (30 μM) were markedly smaller in VIRUS segments compared to SHAM segments (2 ± 1% relaxation vs. 28 ± 5%, n=14, p<0.01). Similarly, relaxation responses of VIRUS segments to the neuropeptide substance P (SP) were greatly attenuated (1 ± 1% vs. 47 ± 6% evoked by 1 nM SP, n=14, p<0.001). Consistent with epithelial damage, PGE2 release in response to both acrolein and SP were reduced in VIRUS segments (>35% reduction, n=6, p<0.01), as determined using ELISA. In contrast, exogenous PGE2 was 2.8-fold more potent in VIRUS relative to SHAM segments (-log EC50 7.82 ± 0.14 vs. 7.38 ± 0.05, n=7, p<0.01) whilst responses of VIRUS segments to the β-adrenoceptor agonist isoprenaline were similar to SHAM segments. In conclusion, relaxation responses evoked by acrolein were profoundly diminished in tracheal segments isolated from influenza A-infected mice. The mechanism through which influenza A infection attenuates this response appears to involve reduced production of PGE2 in response to SP due to epithelial cell loss, and may provide insight into the airway hyperreactivity observed with influenza A infection.
Collapse
Affiliation(s)
- Esther Y Cheah
- School of Medicine and Pharmacology, The University of Western Australia, Crawley, WA 6009, Australia
| | - Tracy S Mann
- School of Medicine and Pharmacology, The University of Western Australia, Crawley, WA 6009, Australia
| | - Philip C Burcham
- School of Medicine and Pharmacology, The University of Western Australia, Crawley, WA 6009, Australia
| | - Peter J Henry
- School of Medicine and Pharmacology, The University of Western Australia, Crawley, WA 6009, Australia.
| |
Collapse
|
177
|
Patel DA, You Y, Huang G, Byers DE, Kim HJ, Agapov E, Moore ML, Peebles RS, Castro M, Sumino K, Shifren A, Brody SL, Holtzman MJ. Interferon response and respiratory virus control are preserved in bronchial epithelial cells in asthma. J Allergy Clin Immunol 2014; 134:1402-1412.e7. [PMID: 25216987 PMCID: PMC4261010 DOI: 10.1016/j.jaci.2014.07.013] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 06/06/2014] [Accepted: 07/02/2014] [Indexed: 12/27/2022]
Abstract
BACKGROUND Some investigators find a deficiency in IFN production from airway epithelial cells infected with human rhinovirus in asthma, but whether this abnormality occurs with other respiratory viruses is uncertain. OBJECTIVE To assess the effect of influenza A virus (IAV) and respiratory syncytial virus (RSV) infection on IFN production and viral level in human bronchial epithelial cells (hBECs) from subjects with and without asthma. METHODS Primary-culture hBECs from subjects with mild to severe asthma (n = 11) and controls without asthma (hBECs; n = 7) were infected with live or ultraviolet-inactivated IAV (WS/33 strain), RSV (Long strain), or RSV (A/2001/2-20 strain) with multiplicity of infection 0.01 to 1. Levels of virus along with IFN-β and IFN-λ and IFN-stimulated gene expression (tracked by 2'-5'-oligoadenylate synthetase 1 and myxovirus (influenza virus) resistance 1 mRNA) were determined up to 72 hours postinoculation. RESULTS After IAV infection, viral levels were increased 2-fold in hBECs from asthmatic subjects compared with nonasthmatic control subjects (P < .05) and this increase occurred in concert with increased IFN-λ1 levels and no significant difference in IFNB1, 2'-5'-oligoadenylate synthetase 1, or myxovirus (influenza virus) resistance 1mRNA levels. After RSV infections, viral levels were not significantly increased in hBECs from asthmatic versus nonasthmatic subjects and the only significant difference between groups was a decrease in IFN-λ levels (P < .05) that correlated with a decrease in viral titer. All these differences were found only at isolated time points and were not sustained throughout the 72-hour infection period. CONCLUSIONS The results indicate that IAV and RSV control and IFN response to these viruses in airway epithelial cells is remarkably similar between subjects with and without asthma.
Collapse
Affiliation(s)
- Dhara A. Patel
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, St. Louis, MO
| | - Yingjian You
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, St. Louis, MO
| | - Guangming Huang
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, St. Louis, MO
| | - Derek E. Byers
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, St. Louis, MO
| | - Hyun Jik Kim
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, St. Louis, MO
| | - Eugene Agapov
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, St. Louis, MO
| | - Martin L. Moore
- Emory University Department of Pediatrics and Children's Healthcare of Atlanta, Atlanta, GA
| | - R. Stokes Peebles
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt School of Medicine, Nashville, TN
| | - Mario Castro
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, St. Louis, MO
| | - Kaharu Sumino
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, St. Louis, MO
| | - Adrian Shifren
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, St. Louis, MO
| | - Steven L. Brody
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, St. Louis, MO
| | - Michael J. Holtzman
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, St. Louis, MO
| |
Collapse
|
178
|
Manthei DM, Schwantes EA, Mathur SK, Guadarrama AG, Kelly EA, Gern JE, Jarjour NN, Denlinger LC. Nasal lavage VEGF and TNF-α levels during a natural cold predict asthma exacerbations. Clin Exp Allergy 2014; 44:1484-93. [PMID: 25109477 PMCID: PMC4247169 DOI: 10.1111/cea.12387] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 06/09/2014] [Accepted: 06/20/2014] [Indexed: 01/09/2023]
Abstract
BACKGROUND Asthma exacerbations contribute to significant morbidity, mortality and healthcare utilization. Furthermore, viral infections are associated with asthma exacerbations by mechanisms that are not fully understood. OBJECTIVE The aim of this analysis was to determine whether cytokine patterns in patients with colds could identify risks for subsequent asthma exacerbations. METHODS We analysed cytokine levels in nasal lavage fluid (NLF) in 59 subjects (46 with asthma) with acute upper respiratory symptoms and after symptomatic resolution. Analyte choice was based on potential relevance to asthma exacerbations: antiviral (IFN-α, IFN-β, IFN-γ, IFN-λ1, IP-10, TRAIL), cell recruiting (G-CSF, IL-1β, IL-8, MCP-1, MCP-3, TNF-α), polarizing (CXCL13, IL-10, IL-13, IL-17, TSLP), and injury remodelling (fibronectin, IL-33, MMP-9, VEGF). RESULTS The overall cytokine response induced during viral infections was not different between asthmatic and non-asthmatic individuals for a wide array of cytokines. However, mean levels of VEGF, TNF-α and IL-1β were 1.7-, 5.1- and 4.7-fold higher in samples from asthma subjects who exacerbated in the first 3 weeks of the cold compared with those who did not exacerbate (P = 0.006, 0.01, 0.048, respectively). Using receiver operating characteristic curve-defined thresholds, high VEGF and TNF-α levels predicted a shorter time-to-exacerbation after NLF sampling (25% exacerbation rate: 3 vs. 45 days, and 3 vs. 26 days; P = 0.03, 0.04, respectively). CONCLUSION AND CLINICAL RELEVANCE Although they produce similar cytokine responses to viral infection as non-asthmatics, asthmatics with higher levels of VEGF and TNF-α in NLF obtained during acute cold phases predicted subsequent asthma exacerbations in this cohort of patients with mild-to-moderate disease. In the future, stratifying the risk of an asthma exacerbation by cytokine profile may aid the targeting of personalized treatment and intervention strategies.
Collapse
Affiliation(s)
- D M Manthei
- University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | | | | | | | | | | | | |
Collapse
|
179
|
Yadava K, Massacand J, Mosconi I, Nicod LP, Harris NL, Marsland BJ. Thymic stromal lymphopoietin plays divergent roles in murine models of atopic and nonatopic airway inflammation. Allergy 2014; 69:1333-42. [PMID: 24961817 DOI: 10.1111/all.12469] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND Thymic stromal lymphopoietin (TSLP) is a cytokine primarily produced by epithelial cells, which has been shown to be a potent inducer of T-helper 2 (Th2)-type responses. However, TSLP has pleiotropic effects upon immune cells, and although extensively studied in the context of atopic asthma, its relevance as a therapeutic target and its role in the pathogenesis of nonatopic asthma remains unknown. We sought to investigate the role of TSLP in atopic, nonatopic and viral-induced exacerbations of pulmonary inflammation. METHODS Using stringently defined murine models of atopic, nonatopic and virally exacerbated forms of pulmonary inflammation, we compared inflammatory responses of C57BL/6 wild-type (WT) and TSLP receptor-deficient (TSLPR KO) mice. RESULTS Thymic stromal lymphopoietin receptor (TSLPR) signaling was crucial for the development of atopic asthma. Specifically, TSLPR signaling to lung recruited CD4+ T cells enhanced eosinophilia, goblet cell hyperplasia, and overall inflammation within the airways. In contrast, the absence of TSLPR signaling was associated with strikingly exaggerated pulmonary neutrophilic inflammation in a nonatopic model of airway inflammation. The inflammation was associated with excessive levels of interleukin (IL)-17A in the lungs, indicating that TSLP negatively regulates IL-17A. In addition, in a model of influenza-induced exacerbation of atopic airway inflammation, the absence of TSLPR signaling also led to exaggerated neutrophilic inflammation. CONCLUSION Thymic stromal lymphopoietin plays divergent roles in the pathogenesis of atopic and nonatopic asthma phenotypes by either enhancing Th2 responses or curtailing T-helper 17 responses. These findings raise important caveats for the design of therapeutic interventions targeting TSLP in asthma.
Collapse
Affiliation(s)
- K. Yadava
- Service de Pneumologie; Faculty of Biology and Medicine; University of Lausanne; CHUV; Lausanne Switzerland
| | - J. Massacand
- Global Health Institute; EPFL; Lausanne Switzerland
| | - I. Mosconi
- Global Health Institute; EPFL; Lausanne Switzerland
| | - L. P. Nicod
- Service de Pneumologie; Faculty of Biology and Medicine; University of Lausanne; CHUV; Lausanne Switzerland
| | - N. L. Harris
- Global Health Institute; EPFL; Lausanne Switzerland
| | - B. J. Marsland
- Service de Pneumologie; Faculty of Biology and Medicine; University of Lausanne; CHUV; Lausanne Switzerland
| |
Collapse
|
180
|
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
|
181
|
Herbert C, Zeng QX, Shanmugasundaram R, Garthwaite L, Oliver BG, Kumar RK. Response of airway epithelial cells to double-stranded RNA in an allergic environment. TRANSLATIONAL RESPIRATORY MEDICINE 2014; 2:11. [PMID: 25264520 PMCID: PMC4173067 DOI: 10.1186/s40247-014-0011-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 08/21/2014] [Indexed: 01/10/2023]
Abstract
Background Respiratory viral infections are the most common trigger of acute exacerbations in patients with allergic asthma. The anti-viral response of airway epithelial cells (AEC) may be impaired in asthmatics, while cytokines produced by AEC may drive the inflammatory response. We investigated whether AEC cultured in the presence of Th2 cytokines associated with an allergic environment exhibited altered responses to double-stranded RNA, a virus-like stimulus. Methods We undertook preliminary studies using the MLE-12 cell line derived from mouse distal respiratory epithelial cells, then confirmed and extended our findings using low-passage human AEC. Cells were cultured in the absence or presence of the Th2 cytokines IL-4 and IL-13 for 48 hours, then stimulated with poly I:C for 4 hours. Expression of relevant anti-viral response and cytokine genes was assessed by quantitative real-time PCR. Secretion of cytokine proteins was assessed by immunoassay. Results Following stimulation with poly I:C, MLE-12 cells pre-treated with Th2 cytokines exhibited significantly higher levels of expression of mRNA for the cytokine genes Cxcl10 and Cxcl11, as well as a trend towards increased expression of Cxcl9 and Il6. Expression of anti-viral response genes was mostly unchanged, although Stat1, Ifit1 and Ifitm3 were significantly increased in Th2 cytokine pre-treated cells. Human AEC pre-treated with IL-4 and IL-13, then stimulated with poly I:C, similarly exhibited significantly higher expression of IL8, CXCL9, CXCL10, CXCL11 and CCL5 genes. In parallel, there was significantly increased secretion of CXCL8 and CCL5, as well as a trend towards increased secretion of CXCL10 and IL-6. Again, expression of anti-viral response genes was not decreased. Rather, there was significantly enhanced expression of mRNA for type III interferons, RNA helicases and other interferon-stimulated genes. Conclusion The Th2 cytokine environment appears to promote increased production of pro-inflammatory chemokines by AEC in response to double-stranded RNA, which could help explain the exaggerated inflammatory response to respiratory viral infection in allergic asthmatics. However, any impairment of anti-viral host defences in asthmatics appears unlikely to be a consequence of Th2 cytokine-induced downregulation of the expression of viral response genes by AEC. Electronic supplementary material The online version of this article (doi:10.1186/s40247-014-0011-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Cristan Herbert
- Department of Pathology, School of Medical Sciences, UNSW Australia, Sydney, 2052 Australia
| | - Qing-Xiang Zeng
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, University of Sydney, Sydney, 2037 Australia ; Otorhinolaryngology Hospital, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080 China
| | | | - Linda Garthwaite
- Department of Pathology, School of Medical Sciences, UNSW Australia, Sydney, 2052 Australia
| | - Brian G Oliver
- Respiratory Cellular and Molecular Biology, Woolcock Institute of Medical Research, University of Sydney, Sydney, 2037 Australia ; School of Medical & Molecular Biosciences, University of Technology Sydney, Sydney, 2007 Australia
| | - Rakesh K Kumar
- Department of Pathology, School of Medical Sciences, UNSW Australia, Sydney, 2052 Australia
| |
Collapse
|
182
|
Djukanović R, Harrison T, Johnston SL, Gabbay F, Wark P, Thomson NC, Niven R, Singh D, Reddel HK, Davies DE, Marsden R, Boxall C, Dudley S, Plagnol V, Holgate ST, Monk P. The effect of inhaled IFN-β on worsening of asthma symptoms caused by viral infections. A randomized trial. Am J Respir Crit Care Med 2014; 190:145-54. [PMID: 24937476 DOI: 10.1164/rccm.201312-2235oc] [Citation(s) in RCA: 205] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
RATIONALE Ex vivo, bronchial epithelial cells from people with asthma are more susceptible to rhinovirus infection caused by deficient induction of the antiviral protein, IFN-β. Exogenous IFN-β restores antiviral activity. OBJECTIVES To compare the efficacy and safety of inhaled IFN-β with placebo administered to people with asthma after onset of cold symptoms to prevent or attenuate asthma symptoms caused by respiratory viruses. METHODS A total of 147 people with asthma on inhaled corticosteroids (British Thoracic Society Steps 2-5), with a history of virus-associated exacerbations, were randomized to 14-day treatment with inhaled IFN-β (n = 72) or placebo (n = 75) within 24 hours of developing cold symptoms and were assessed clinically, with relevant samples collected to assess virus infection and antiviral responses. MEASUREMENTS AND MAIN RESULTS A total of 91% of randomized patients developed a defined cold. In this modified intention-to-treat population, asthma symptoms did not get clinically significantly worse (mean change in six-item Asthma Control Questionnaire <0.5) and IFN-β treatment had no significant effect on this primary endpoint, although it enhanced morning peak expiratory flow recovery (P = 0.033), reduced the need for additional treatment, and boosted innate immunity as assessed by blood and sputum biomarkers. In an exploratory analysis of the subset of more difficult-to-treat, Step 4-5 people with asthma (n = 27 IFN-β; n = 31 placebo), Asthma Control Questionnaire-6 increased significantly on placebo; this was prevented by IFN-β (P = 0.004). CONCLUSIONS Although the trial did not meet its primary endpoint, it suggests that inhaled IFN-β is a potential treatment for virus-induced deteriorations of asthma in difficult-to-treat people with asthma and supports the need for further, adequately powered, trials in this population. Clinical trial registered with www.clinicaltrials.gov (NCT 01126177).
Collapse
Affiliation(s)
- Ratko Djukanović
- 1 NIHR Southampton Respiratory Biomedical Research Unit, Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
183
|
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
|
184
|
Panjwani A, Strauss M, Gold S, Wenham H, Jackson T, Chou JJ, Rowlands DJ, Stonehouse NJ, Hogle JM, Tuthill TJ. Capsid protein VP4 of human rhinovirus induces membrane permeability by the formation of a size-selective multimeric pore. PLoS Pathog 2014; 10:e1004294. [PMID: 25102288 PMCID: PMC4125281 DOI: 10.1371/journal.ppat.1004294] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Accepted: 06/24/2014] [Indexed: 01/18/2023] Open
Abstract
Non-enveloped viruses must deliver their viral genome across a cell membrane without the advantage of membrane fusion. The mechanisms used to achieve this remain poorly understood. Human rhinovirus, a frequent cause of the common cold, is a non-enveloped virus of the picornavirus family, which includes other significant pathogens such as poliovirus and foot-and-mouth disease virus. During picornavirus cell entry, the small myristoylated capsid protein VP4 is released from the virus, interacts with the cell membrane and is implicated in the delivery of the viral RNA genome into the cytoplasm to initiate replication. In this study, we have produced recombinant C-terminal histidine-tagged human rhinovirus VP4 and shown it can induce membrane permeability in liposome model membranes. Dextran size-exclusion studies, chemical crosslinking and electron microscopy demonstrated that VP4 forms a multimeric membrane pore, with a channel size consistent with transfer of the single-stranded RNA genome. The membrane permeability induced by recombinant VP4 was influenced by pH and was comparable to permeability induced by infectious virions. These findings present a molecular mechanism for the involvement of VP4 in cell entry and provide a model system which will facilitate exploration of VP4 as a novel antiviral target for the picornavirus family. Human rhinovirus (HRV) is a non-enveloped virus of the picornavirus family and is responsible for respiratory infections (common colds) costing billions of dollars ($) annually. There remains no vaccine or licensed drug to prevent or reduce infection. Related members of the picornavirus family include significant pathogens such as poliovirus, enterovirus 71 and foot-and-mouth disease virus, for which improved control measures are also required. A fundamental step in virus infection is the delivery of the viral genetic material through the barrier of the cellular membrane. Viruses such as HIV and influenza are enveloped in an outer membrane which can fuse with the host cell membrane to allow the viral genome to penetrate into the cytoplasm. However, non-enveloped viruses such as picornaviruses lack a membrane and the mechanisms for penetration of the membrane by these viruses remain poorly understood. The capsid protein VP4 has previously been implicated in the delivery of the picornavirus genome. In this study we demonstrate that HRV VP4 interacts with membranes to make them permeable by the formation of multimeric, size-selective membrane pores with properties consistent with the transport of viral genome through the membrane. This function of VP4 provides a novel antiviral target for this family of viruses.
Collapse
Affiliation(s)
- Anusha Panjwani
- The Pirbright Institute, Pirbright, Surrey, United Kingdom
- School of Molecular and Cellular Biology & Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, West Yorkshire, United Kingdom
| | - Mike Strauss
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Sarah Gold
- The Pirbright Institute, Pirbright, Surrey, United Kingdom
| | - Hannah Wenham
- The Pirbright Institute, Pirbright, Surrey, United Kingdom
| | - Terry Jackson
- The Pirbright Institute, Pirbright, Surrey, United Kingdom
| | - James J. Chou
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - David J. Rowlands
- School of Molecular and Cellular Biology & Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, West Yorkshire, United Kingdom
| | - Nicola J. Stonehouse
- School of Molecular and Cellular Biology & Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, West Yorkshire, United Kingdom
| | - James M. Hogle
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, United States of America
| | | |
Collapse
|
185
|
Human rhinovirus 16 causes Golgi apparatus fragmentation without blocking protein secretion. J Virol 2014; 88:11671-85. [PMID: 25100828 PMCID: PMC4178721 DOI: 10.1128/jvi.01170-14] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The replication of picornaviruses has been described to cause fragmentation of the Golgi apparatus that blocks the secretory pathway. The inhibition of major histocompatibility complex class I upregulation and cytokine, chemokine and interferon secretion may have important implications for host defense. Previous studies have shown that disruption of the secretory pathway can be replicated by expression of individual nonstructural proteins; however the situation with different serotypes of human rhinovirus (HRV) is unclear. The expression of 3A protein from HRV14 or HRV2 did not cause Golgi apparatus disruption or a block in secretion, whereas other studies showed that infection of cells with HRV1A did cause Golgi apparatus disruption which was replicated by the expression of 3A. HRV16 is the serotype most widely used in clinical HRV challenge studies; consequently, to address the issue of Golgi apparatus disruption for HRV16, we have systematically and quantitatively examined the effect of HRV16 on both Golgi apparatus fragmentation and protein secretion in HeLa cells. First, we expressed each individual nonstructural protein and examined their cellular localization and their disruption of endoplasmic reticulum and Golgi apparatus architecture. We quantified their effects on the secretory pathway by measuring secretion of the reporter protein Gaussia luciferase. Finally, we examined the same outcomes following infection of cells with live virus. We demonstrate that expression of HRV16 3A and 3AB and, to a lesser extent, 2B caused dispersal of the Golgi structure, and these three nonstructural proteins also inhibited protein secretion. The infection of cells with HRV16 also caused significant Golgi apparatus dispersal; however, this did not result in the inhibition of protein secretion. IMPORTANCE The ability of replicating picornaviruses to influence the function of the secretory pathway has important implications for host defense. However, there appear to be differences between different members of the family and inconsistent results when comparing infection with live virus to expression of individual nonstructural proteins. We demonstrate that individual nonstructural HRV16 proteins, when expressed in HeLa cells, can both fragment the Golgi apparatus and block secretion, whereas viral infection fragments the Golgi apparatus without blocking secretion. This has major implications for how we interpret mechanistic evidence derived from the expression of single viral proteins.
Collapse
|
186
|
Zomer-Kooijker K, Uiterwaal CSPM, Verschueren KJC, Maitland-vd Zee AH, Balemans WAF, van Ewijk BE, van Velzen MF, van der Ent CK. Respiratory tract infections and asthma control in children. Respir Med 2014; 108:1446-52. [PMID: 25087902 DOI: 10.1016/j.rmed.2014.07.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 06/22/2014] [Accepted: 07/07/2014] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Asthma control is considered the major goal of asthma management, while many determinants of control are difficult to modify. We studied the association between respiratory infection episodes (RTIs) of various types and asthma control. METHODS Cross-sectional data were used from children aged 4-18 years with physician-diagnosed asthma who participated in a web-based electronic portal for children with asthma, allergies or infections. Asthma control was measured using the Childhood Asthma Control Test (C-ACT) or the Asthma Control Test (ACT). Linear regression was used to analyse the association between categories of numbers of various types of RTIs sustained in the preceding 12 months (categorized) and asthma control, adjusted for potential confounders. RESULTS Asthma control was assessed in 654 children, and 68.5% were clinically well controlled (ACT ≥ 20). Higher total numbers of RTIs in the last 12 months were strongly associated with a lower level of asthma control (p(trend) < 0.001). Similarly strong statistically significant associations were found for subtypes of RTI: ≥4 vs. 0 otitis episodes: coefficient -1.7 (95% CI -3.3 to -0.2); ≥5 vs.0 colds: coefficient -2.3 (95% CI -3.0 to -1.6); ≥3 vs. 0 bronchitis episodes: coefficient -3.1 (95% CI -4.0 to -2.3), each with p(trend) < 0.05. CONCLUSION Higher numbers of reported respiratory tract infections are associated with lower level of asthma control. The different type of respiratory tract infections contribute equally to less controlled asthma.
Collapse
Affiliation(s)
- Kim Zomer-Kooijker
- Department of Paediatric Pulmonology and Allergology, University Medical Centre Utrecht, Lundlaan 6, 3508 AB Utrecht, The Netherlands.
| | - Cuno S P M Uiterwaal
- Julius Centre for Health Sciences and Primary Care, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Kim J C Verschueren
- Department of Paediatric Pulmonology and Allergology, University Medical Centre Utrecht, Lundlaan 6, 3508 AB Utrecht, The Netherlands
| | - Anke-Hilse Maitland-vd Zee
- Utrecht Institute for Pharmaceutical Sciences, Division of Pharmacoepidemiology & Clinical Pharmacology, David de Wied Building, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Walter A F Balemans
- St. Antonius Hospital, Department of Paediatrics, Postbus 2500, 3430 EM Nieuwegein, The Netherlands
| | - Bart E van Ewijk
- Tergooi Hospital, Department of Paediatrics, Rijksstraatweg 1, 1261 AN Blaricum, The Netherlands
| | - Maartje F van Velzen
- Meander Medisch Centrum, Department of Paediatrics, Postbus 1502, 3800 BM Amersfoort, The Netherlands
| | - Cornelis K van der Ent
- Department of Paediatric Pulmonology and Allergology, University Medical Centre Utrecht, Lundlaan 6, 3508 AB Utrecht, The Netherlands
| |
Collapse
|
187
|
Liang Z, Zhang Q, Thomas CM, Chana KK, Gibeon D, Barnes PJ, Chung KF, Bhavsar PK, Donnelly LE. Impaired macrophage phagocytosis of bacteria in severe asthma. Respir Res 2014; 15:72. [PMID: 24972601 PMCID: PMC4086996 DOI: 10.1186/1465-9921-15-72] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 06/20/2014] [Indexed: 01/18/2023] Open
Abstract
Background Bacteria are frequently cultured from sputum samples of severe asthma patients suggesting a defect in bacterial clearance from the airway. We measured the capacity of macrophages from patients with asthma to phagocytose bacteria. Methods Phagocytosis of fluorescently-labelled polystyrene beads, Haemophilus influenzae or Staphylococcus aureus by broncholaveolar lavage alveolar macrophages (AM) and by monocyte-derived macrophages (MDM) from non-asthmatics, mild-moderate and severe asthmatic patients was assessed using fluorimetry. Results There were no differences in phagocytosis of polystyrene beads by AMs or MDMs from any of the subject groups. There was reduced phagocytosis of Haemophilus influenzae and Staphylococcus aureus in MDMs from patients with severe asthma compared to non-severe asthma (p < 0.05 and p < 0.01, respectively) and healthy subjects (p < 0.01and p < 0.001, respectively). Phagocytosis of Haemophilus influenzae and Staphylococcus aureus by AM was also reduced in severe asthma compared to normal subjects (p < 0.05). Dexamethasone and formoterol did not suppress phagocytosis of bacteria by MDMs from any of the groups. Conclusions Persistence of bacteria in the lower airways may result partly from a reduced phagocytic capacity of macrophages for bacteria. This may contribute to increased exacerbations, airway colonization and persistence of inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Pankaj K Bhavsar
- Airway Disease, National Heart and Lung Institute, Imperial College London, & Biomedical Research Unit, Royal Brompton & Harefield NHS Trust, London SW3, UK.
| | | |
Collapse
|
188
|
Kumar RK, Foster PS, Rosenberg HF. Respiratory viral infection, epithelial cytokines, and innate lymphoid cells in asthma exacerbations. J Leukoc Biol 2014; 96:391-6. [PMID: 24904000 DOI: 10.1189/jlb.3ri0314-129r] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Exacerbations of asthma are most commonly triggered by viral infections, which amplify allergic inflammation. Cytokines released by virus-infected AECs may be important in driving this response. This review focuses on accumulating evidence in support of a role for epithelial cytokines, including IL-33, IL-25, and TSLP, as well as their targets, type 2 innate lymphoid cells (ILC2s), in the pathogenesis of virus-induced asthma exacerbations. Production and release of these cytokines lead to recruitment and activation of ILC2s, which secrete mediators, including IL-5 and IL-13, which augment allergic inflammation. However, little information is currently available about the induction of these responses by the respiratory viruses that are strongly associated with exacerbations of asthma, such as rhinoviruses. Further human studies, as well as improved animal experimental models, are needed to investigate appropriately the pathogenetic mechanisms in virus-induced exacerbations of asthma, including the role of ILCs.
Collapse
Affiliation(s)
- Rakesh K Kumar
- Department of Pathology, University of New South Wales, Sydney, Australia;
| | - Paul S Foster
- Centre for Asthma and Respiratory Disease, University of Newcastle and Hunter Medical Research Institute, Callaghan, Australia; and
| | - Helene F Rosenberg
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
189
|
Zhu L, Lee B, Zhao F, Zhou X, Chin V, Ling SC, Chen Y. Modulation of airway epithelial antiviral immunity by fungal exposure. Am J Respir Cell Mol Biol 2014; 50:1136-43. [PMID: 24428709 PMCID: PMC4068913 DOI: 10.1165/rcmb.2013-0357oc] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 12/26/2013] [Indexed: 01/18/2023] Open
Abstract
Multiple pathogens, such as bacteria, fungi, and viruses, have been frequently found in asthmatic airways and are associated with the pathogenesis and exacerbation of asthma. Among these pathogens, Alternaria alternata (Alt), a universally present fungus, and human rhinovirus have been extensively studied. However, their interactions have not been investigated. In the present study, we tested the effect of Alt exposure on virus-induced airway epithelial immunity using live virus and a synthetic viral mimicker, double-stranded RNA (dsRNA). Alt treatment was found to significantly enhance the production of proinflammatory cytokines (e.g., IL-6 and IL-8) induced by virus infection or dsRNA treatment. In contrast to this synergistic effect, Alt significantly repressed type I and type III IFN production, and this impairment led to elevated viral replication. Mechanistic studies suggested the positive role of NF-κB and mitogen-activated protein kinase pathways in the synergism and the attenuation of the TBK1-IRF3 pathway in the inhibition of IFN production. These opposite effects are caused by separate fungal components. Protease-dependent and -independent mechanisms appear to be involved. Thus, Alt exposure alters the airway epithelial immunity to viral infection by shifting toward more inflammatory but less antiviral responses.
Collapse
Affiliation(s)
- Lingxiang Zhu
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, Arizona; and
| | - Boram Lee
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, Arizona; and
| | - Fangkun Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, Arizona; and
- Department of Ophthalmology, School of Medicine, China Medical University, Shenyang, China
| | - Xu Zhou
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, Arizona; and
| | - Vanessa Chin
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, Arizona; and
| | - Serena C. Ling
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, Arizona; and
| | - Yin Chen
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, Arizona; and
| |
Collapse
|
190
|
Saraya T, Kurai D, Ishii H, Ito A, Sasaki Y, Niwa S, Kiyota N, Tsukagoshi H, Kozawa K, Goto H, Takizawa H. Epidemiology of virus-induced asthma exacerbations: with special reference to the role of human rhinovirus. Front Microbiol 2014; 5:226. [PMID: 24904541 PMCID: PMC4033317 DOI: 10.3389/fmicb.2014.00226] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Accepted: 04/28/2014] [Indexed: 11/13/2022] Open
Abstract
Viral respiratory infections may be associated with the virus-induced asthma in adults as well as children. Particularly, human rhinovirus is strongly suggested a major candidate for the associations of the virus-induced asthma. Thus, in this review, we reviewed and focused on the epidemiology, pathophysiology, and treatment of virus-induced asthma with special reference on human rhinovirus. Furthermore, we added our preliminary data regarding the clinical and virological findings in the present review.
Collapse
Affiliation(s)
- Takeshi Saraya
- Department of Respiratory Medicine, School of Medicine, Kyorin University Mitaka, Tokyo, Japan
| | - Daisuke Kurai
- Department of Respiratory Medicine, School of Medicine, Kyorin University Mitaka, Tokyo, Japan
| | - Haruyuki Ishii
- Department of Respiratory Medicine, School of Medicine, Kyorin University Mitaka, Tokyo, Japan
| | - Anri Ito
- Department of Respiratory Medicine, School of Medicine, Kyorin University Mitaka, Tokyo, Japan
| | - Yoshiko Sasaki
- Gunma Prefectural Institute of Public Health and Environmental Sciences Gunma, Japan
| | - Shoichi Niwa
- Gunma Prefectural Institute of Public Health and Environmental Sciences Gunma, Japan
| | - Naoko Kiyota
- Kumamoto Prefectural Institute of Public Health and Environmental Sciences Kumamoto, Japan
| | - Hiroyuki Tsukagoshi
- Gunma Prefectural Institute of Public Health and Environmental Sciences Gunma, Japan
| | - Kunihisa Kozawa
- Gunma Prefectural Institute of Public Health and Environmental Sciences Gunma, Japan
| | - Hajime Goto
- Department of Respiratory Medicine, School of Medicine, Kyorin University Mitaka, Tokyo, Japan
| | - Hajime Takizawa
- Department of Respiratory Medicine, School of Medicine, Kyorin University Mitaka, Tokyo, Japan
| |
Collapse
|
191
|
Jartti T, Palomares O, Waris M, Tastan O, Nieminen R, Puhakka T, Rückert B, Aab A, Vuorinen T, Allander T, Vahlberg T, Ruuskanen O, Akdis M, Akdis CA. Distinct regulation of tonsillar immune response in virus infection. Allergy 2014; 69:658-67. [PMID: 24684577 PMCID: PMC7159333 DOI: 10.1111/all.12396] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2014] [Indexed: 01/05/2023]
Abstract
Background The relationships between tonsillar immune responses, and viral infection and allergy are incompletely known. Objective To study intratonsillar/nasopharyngeal virus detections and in vivo expressions of T‐cell‐ and innate immune response‐specific cytokines, transcription factors, and type I/II/III interferons in human tonsils. Methods Palatine tonsil samples were obtained from 143 elective tonsillectomy patients. Adenovirus, bocavirus‐1, coronavirus, enteroviruses, influenza virus, metapneumovirus, parainfluenza virus, rhinovirus, and respiratory syncytial virus were detected using PCR. The mRNA expression levels of IFN‐α, IFN‐β, IFN‐γ, IL‐10, IL‐13, IL‐17, IL‐28, IL‐29, IL‐37, TGF‐β, FOXP3, GATA3, RORC2, and Tbet were directly analyzed by quantitative RT‐PCR. Results Fifty percentage of subjects reported allergy, 59% had ≥1 nasopharyngeal viruses, and 24% had ≥1 intratonsillar viruses. Tonsillar virus detection showed a strong negative association with age; especially rhinovirus or parainfluenza virus detection showed positive association with IFN‐γ and Tbet expressions. IL‐37 expression was positively associated with atopic dermatitis, whereas IFN‐α, IL‐13, IL‐28, and Tbet expressions were negatively associated with allergic diseases. Network analyses demonstrated strongly polarized clusters of immune regulatory (IL‐10, IL‐17, TGF‐β, FOXP3, GATA3, RORC2, Tbet) and antiviral (IFN‐α, IFN‐β, IL‐28, IL‐29) genes. These two clusters became more distinctive in the presence of viral infection or allergy. A negative correlation between antiviral cytokines and IL‐10, IL‐17, IL‐37, FOXP3, and RORC2 was observed only in the presence of viruses, and interestingly, IL‐13 strongly correlated with antiviral cytokines. Conclusions Tonsillar cytokine expression is closely related to existing viral infections, age, and allergic illnesses and shows distinct clusters between antiviral and immune regulatory genes.
Collapse
Affiliation(s)
- T. Jartti
- Department of Pediatrics; Turku University Hospital; Turku Finland
| | - O. Palomares
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zürich; Christine Kühne-Center for Allergy Research and Education (CK-CARE); Davos Switzerland
- Department of Biochemistry and Molecular Biology; School of Chemistry; Complutense University of Madrid (UCM); Madrid Spain
| | - M. Waris
- Department of Virology; University of Turku; Turku Finland
| | - O. Tastan
- Department of Computer Engineering; Bilkent University; Ankara Turkey
| | - R. Nieminen
- Department of Pediatrics; Turku University Hospital; Turku Finland
| | - T. Puhakka
- Department of Otorhinolaryngology; Turku University Hospital; Turku Finland
- Department of Otorhinolaryngology; Satakunta Central Hospital; Pori Finland
| | - B. Rückert
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zürich; Christine Kühne-Center for Allergy Research and Education (CK-CARE); Davos Switzerland
| | - A. Aab
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zürich; Christine Kühne-Center for Allergy Research and Education (CK-CARE); Davos Switzerland
| | - T. Vuorinen
- Department of Virology; University of Turku; Turku Finland
| | - T. Allander
- Department of Clinical Microbiology; Karolinska University Hospital; Stockholm Sweden
| | - T. Vahlberg
- Department of Biostatistics; University of Turku; Turku Finland
| | - O. Ruuskanen
- Department of Pediatrics; Turku University Hospital; Turku Finland
| | - M. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zürich; Christine Kühne-Center for Allergy Research and Education (CK-CARE); Davos Switzerland
| | - C. A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF); University of Zürich; Christine Kühne-Center for Allergy Research and Education (CK-CARE); Davos Switzerland
| |
Collapse
|
192
|
Ichikawa T, Sugiura H, Koarai A, Minakata Y, Kikuchi T, Morishita Y, Oka A, Kanai K, Kawabata H, Hiramatsu M, Akamatsu K, Hirano T, Nakanishi M, Matsunaga K, Yamamoto N, Ichinose M. TLR3 activation augments matrix metalloproteinase production through reactive nitrogen species generation in human lung fibroblasts. THE JOURNAL OF IMMUNOLOGY 2014; 192:4977-88. [PMID: 24760149 DOI: 10.4049/jimmunol.1302919] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Viral infection often triggers asthma exacerbation and contributes to airway remodeling. Cell signaling in viral infection is mainly mediated through TLR3. Many mediators are involved in airway remodeling, but matrix metalloproteinases (MMPs) are key players in this process in asthma. However, the role of TLR3 activation in production of MMPs is unknown. In this study, we examined the effects of polyinosinic-polycytidylic acid [poly(I:C)], a ligand for TLR3, on production of MMPs in human lung fibroblasts, with a focus on nitrosative stress in TLR3 modulation of MMP production. After lung fibroblasts were treated with poly(I:C), production of MMP-1, -2, and -9 and inducible NO synthase (iNOS) was assessed. The roles of NF-κB and IFN regulatory factor-3 (IRF-3) in the poly(I:C)-mediated production of MMPs and the responsiveness to poly(I:C) of normal lung fibroblasts and asthmatic lung fibroblasts were also investigated. Poly(I:C) augmented production of MMPs and iNOS in fibroblasts, and an iNOS inhibitor diminished this production of MMPs. Poly(I:C) stimulated translocation of NF-κB and IRF-3 into the nucleus in fibroblasts and inhibition of NF-κB or IRF-3 abrogated the poly(I:C)-induced increase in both iNOS expression and release of MMPs. Poly(I:C)-induced production of iNOS and MMPs was greater in asthmatic fibroblasts than in normal fibroblasts. We conclude that viral infection may induce nitrosative stress and subsequent MMP production via NF-κB- and IRF-3-dependent pathways, thus potentiating viral-induced airway remodeling in asthmatic airways.
Collapse
Affiliation(s)
- Tomohiro Ichikawa
- Third Department of Internal Medicine, Wakayama Medical University, School of Medicine, Wakayama 641-8509, Japan; and
| | - Hisatoshi Sugiura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Akira Koarai
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Yoshiaki Minakata
- Third Department of Internal Medicine, Wakayama Medical University, School of Medicine, Wakayama 641-8509, Japan; and
| | - Takashi Kikuchi
- Third Department of Internal Medicine, Wakayama Medical University, School of Medicine, Wakayama 641-8509, Japan; and
| | - Yukiko Morishita
- Third Department of Internal Medicine, Wakayama Medical University, School of Medicine, Wakayama 641-8509, Japan; and
| | - Asako Oka
- Third Department of Internal Medicine, Wakayama Medical University, School of Medicine, Wakayama 641-8509, Japan; and
| | - Kuninobu Kanai
- Third Department of Internal Medicine, Wakayama Medical University, School of Medicine, Wakayama 641-8509, Japan; and
| | - Hiroki Kawabata
- Third Department of Internal Medicine, Wakayama Medical University, School of Medicine, Wakayama 641-8509, Japan; and
| | - Masataka Hiramatsu
- Third Department of Internal Medicine, Wakayama Medical University, School of Medicine, Wakayama 641-8509, Japan; and
| | - Keiichiro Akamatsu
- Third Department of Internal Medicine, Wakayama Medical University, School of Medicine, Wakayama 641-8509, Japan; and
| | - Tsunahiko Hirano
- Third Department of Internal Medicine, Wakayama Medical University, School of Medicine, Wakayama 641-8509, Japan; and
| | - Masanori Nakanishi
- Third Department of Internal Medicine, Wakayama Medical University, School of Medicine, Wakayama 641-8509, Japan; and
| | - Kazuto Matsunaga
- Third Department of Internal Medicine, Wakayama Medical University, School of Medicine, Wakayama 641-8509, Japan; and
| | - Nobuyuki Yamamoto
- Third Department of Internal Medicine, Wakayama Medical University, School of Medicine, Wakayama 641-8509, Japan; and
| | - Masakazu Ichinose
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| |
Collapse
|
193
|
Gulraiz F, Bellinghausen C, Dentener MA, Reynaert NL, Gaajetaan GR, Beuken EV, Rohde GG, Bruggeman CA, Stassen FR. Efficacy of IFN-λ1 to protect human airway epithelial cells against human rhinovirus 1B infection. PLoS One 2014; 9:e95134. [PMID: 24751942 PMCID: PMC3994020 DOI: 10.1371/journal.pone.0095134] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 03/24/2014] [Indexed: 12/24/2022] Open
Abstract
Impaired interferon (IFN) production has been observed in various obstructive respiratory diseases. This contributes to enhanced sensitivity towards viral infections triggering acute exacerbations. To compensate for this impaired host IFN response, there is need to explore new therapeutic strategies, like exogenous administration of IFNs as prophylactic treatment. In the present study, we examined the protective potential of IFN-λ1 and compared it with the previously established protecting effect of IFN-β. A549 cells and human primary bronchial epithelial cells were first treated with either IFN-β (500 IU/ml) or IFN-λ1 (500 ng/ml) for 18 h. For infection, two approaches were adopted: i) Continuous scenario: after pre-treatment, cells were infected immediately for 24 h with human rhinovirus 1B (HRV1B) in IFN-containing medium, or were cultured for another 72 h in IFN-containing medium, and then infected for 24 h with HRV1B, ii) Pre-treatment scenario: IFN-containing medium was replaced after 18 h and cells were infected for 4 h either immediately after pre-treatment or after additional culturing for 72 h in IFN-free medium. The protective effect was evaluated in terms of reduction in the number of viral copies/infectious progeny, and enhanced expression of IFN-stimulated genes (ISGs). In both cell types and in both approaches, IFN-λ1 and IFN-β treatment resulted in pronounced and long-lasting antiviral effects exemplified by significantly reduced viral copy numbers and diminished infectious progeny. This was associated with strong up-regulation of multiple ISGs. However, in contrast to the IFN-β induced expression of ISGs, which decreased over time, expression of ISGs induced by IFN-λ1 was sustained or even increased over time. Here we demonstrate that the protective potential of IFN-λ1 is comparable to IFN-β. Yet, the long-lasting induction of ISGs by IFN-λ1 and most likely less incitement of side effects due to more localized expression of its receptors could make it an even more promising candidate for prophylactic treatment than IFN-β.
Collapse
Affiliation(s)
- Fahad Gulraiz
- Department of Medical Microbiology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Carla Bellinghausen
- Department of Medical Microbiology, Maastricht University Medical Centre, Maastricht, the Netherlands
- Department of Respiratory Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Mieke A. Dentener
- Department of Respiratory Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Niki L. Reynaert
- Department of Respiratory Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Giel R. Gaajetaan
- Department of Medical Microbiology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Erik V. Beuken
- Department of Medical Microbiology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Gernot G. Rohde
- Department of Respiratory Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Cathrien A. Bruggeman
- Department of Medical Microbiology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Frank R. Stassen
- Department of Medical Microbiology, Maastricht University Medical Centre, Maastricht, the Netherlands
| |
Collapse
|
194
|
Rhinovirus-16 induced release of IP-10 and IL-8 is augmented by Th2 cytokines in a pediatric bronchial epithelial cell model. PLoS One 2014; 9:e94010. [PMID: 24705919 PMCID: PMC3976391 DOI: 10.1371/journal.pone.0094010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 03/12/2014] [Indexed: 01/01/2023] Open
Abstract
Background In response to viral infection, bronchial epithelial cells increase inflammatory cytokine release to activate the immune response and curtail viral replication. In atopic asthma, enhanced expression of Th2 cytokines is observed and we postulated that Th2 cytokines may augment the effects of rhinovirus-induced inflammation. Methods Primary bronchial epithelial cell cultures from pediatric subjects were treated with Th2 cytokines for 24 h before infection with RV16. Release of IL-8, IP-10 and GM-CSF was measured by ELISA. Infection was quantified using RTqPCR and TCID50. Phosphatidyl inositol 3-kinase (PI3K) and P38 mitogen activated protein kinase (MAPK) inhibitors and dexamethasone were used to investigate differences in signaling pathways. Results The presence of Th2 cytokines did not affect RV replication or viral titre, yet there was a synergistic increase in IP-10 release from virally infected cells in the presence of Th2 cytokines. Release of IL-8 and GM-CSF was also augmented. IP-10 release was blocked by a PI3K inhibitor and IL-8 by dexamethasone. Conclusion Th2 cytokines increase release of inflammatory cytokines in the presence of rhinovirus infection. This increase is independent of effects of virus replication. Inhibition of the PI3K pathway inhibits IP-10 expression.
Collapse
|
195
|
Synthesis, biological activity and structure–activity relationship of 4,5-dimethoxybenzene derivatives inhibitor of rhinovirus 14 infection. Eur J Med Chem 2014; 76:445-59. [DOI: 10.1016/j.ejmech.2014.01.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 01/14/2014] [Accepted: 01/18/2014] [Indexed: 11/23/2022]
|
196
|
Rhinovirus exacerbates house-dust-mite induced lung disease in adult mice. PLoS One 2014; 9:e92163. [PMID: 24632596 PMCID: PMC3954893 DOI: 10.1371/journal.pone.0092163] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 02/19/2014] [Indexed: 01/01/2023] Open
Abstract
Human rhinovirus is a key viral trigger for asthma exacerbations. To date, murine studies investigating rhinovirus-induced exacerbation of allergic airways disease have employed systemic sensitisation/intranasal challenge with ovalbumin. In this study, we combined human-rhinovirus infection with a clinically relevant mouse model of aero-allergen exposure using house-dust-mite in an attempt to more accurately understand the links between human-rhinovirus infection and exacerbations of asthma. Adult BALB/c mice were intranasally exposed to low-dose house-dust-mite (or vehicle) daily for 10 days. On day 9, mice were inoculated with human-rhinovirus-1B (or UV-inactivated human-rhinovirus-1B). Forty-eight hours after inoculation, we assessed bronchoalveolar cellular inflammation, levels of relevant cytokines/serum antibodies, lung function and responsiveness/sensitivity to methacholine. House-dust-mite exposure did not result in a classical TH2-driven response, but was more representative of noneosinophilic asthma. However, there were significant effects of house-dust-mite exposure on most of the parameters measured including increased cellular inflammation (primarily macrophages and neutrophils), increased total IgE and house-dust-mite-specific IgG1 and increased responsiveness/sensitivity to methacholine. There were limited effects of human-rhinovirus-1B infection alone, and the combination of the two insults resulted in additive increases in neutrophil levels and lung parenchymal responses to methacholine (tissue elastance). We conclude that acute rhinovirus infection exacerbates house-dust-mite-induced lung disease in adult mice. The similarity of our results using the naturally occurring allergen house-dust-mite, to previous studies using ovalbumin, suggests that the exacerbation of allergic airways disease by rhinovirus infection could act via multiple or conserved mechanisms.
Collapse
|
197
|
Grainge CL, Davies DE. Epithelial injury and repair in airways diseases. Chest 2014; 144:1906-1912. [PMID: 24297122 DOI: 10.1378/chest.12-1944] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Asthma is a common chronic disease characterized by variable respiratory distress with underlying airway inflammation and airflow obstruction. The incidence of asthma has risen inexorably over the past 50 years, suggesting that environmental factors are important in its etiology. All inhaled environmental stimuli interact with the lung at the respiratory epithelium, and it is a testament to the effectiveness of the airway innate defenses that the majority of inhaled substances are cleared without the need to elicit an inflammatory response. However, once this barrier is breached, effective communication with immune and inflammatory cells is required to protect the internal milieu of the lung. In asthma, the respiratory epithelium is known to be structurally and functionally abnormal. Structurally, the epithelium shows evidence of damage and has more mucus-producing cells than normal airways. Functionally, the airway epithelial barrier can be more permeable and more sensitive to oxidants and show a deficient innate immune response to respiratory virus infection compared with that in normal individuals. The potential of a susceptible epithelium and the underlying mesenchyme to create a microenvironment that enables deviation of immune and inflammatory responses to external stimuli may be crucial in the development and progression of asthma. In this review, we consider three important groups of environmental stimuli on the epithelium in asthma: oxidants, such as environmental pollution and acetaminophen; viruses, including rhinovirus; and agents that cause barrier disruption, such as house dust mite allergens. The pathology associated with each stimulus is considered, and potential future treatments arising from research on their effects are presented.
Collapse
Affiliation(s)
- Christopher L Grainge
- Academic Unit of Clinical and Experimental Sciences, University Hospital Southampton, Southampton, England.
| | - Donna E Davies
- Academic Unit of Clinical and Experimental Sciences, University Hospital Southampton, Southampton, England; University of Southampton Faculty of Medicine, and NIHR Respiratory Biomedical Research Unit, University Hospital Southampton, Southampton, England
| |
Collapse
|
198
|
Wickert LE, Karta MR, Audhya A, Gern JE, Bertics PJ. Simvastatin attenuates rhinovirus-induced interferon and CXCL10 secretion from monocytic cells in vitro. J Leukoc Biol 2014; 95:951-9. [PMID: 24532643 DOI: 10.1189/jlb.0713413] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
RV infections frequently trigger exacerbations of respiratory diseases, such as asthma, yet treatment and intervention options remain limited. Statin drugs are the treatment of choice for dyslipidemia and can also modulate immune cell function. To determine whether statin drugs modify antiviral responses of human monocytic cells, we obtained blood monocytes from donors with allergies and/or asthma and treated the cells with sim prior to challenge with RV. RV-induced secretion of CXCL10 was attenuated significantly, irrespective of RV type (RV-16, -14, or -1A), which corresponded with decreases in IFN-α secretion and pSTAT1. Sim pretreatment also reduced RV-induced CXCL10 secretion from human alveolar macrophages. The addition of mev and GGPP-two intermediates of the cholesterol biosynthesis pathway-was able to rescue CXCL10 release fully, demonstrating that effects of sim were related to inhibition of cholesterol biosynthesis and not to an off-target effect. In addition, sim pretreatment attenuated IFN-α-induced pSTAT1 and CXCL10 secretion, providing evidence that sim additionally can affect type I IFNR signaling. SOCS1 and 3 mRNA are both induced with RV stimulation, but sim did not elevate SOCS1 or SOCS3 mRNA expression basally or in the presence of RV. Our findings suggest that sim inhibition of the cholesterol biosynthesis pathway leads to decreased RV-induced chemokine secretion in monocytes and macrophages. These findings suggest that statin drugs have the potential to curb the inflammatory response to RV infection.
Collapse
Affiliation(s)
- Lisa E Wickert
- Departments of Biomolecular Chemistry, Nutritional Sciences, and
| | | | | | - James E Gern
- Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | |
Collapse
|
199
|
Custovic A, Johnston SL, Pavord I, Gaga M, Fabbri L, Bel EH, Le Souëf P, Lötvall J, Demoly P, Akdis CA, Ryan D, Mäkelä MJ, Martinez F, Holloway JW, Saglani S, O'Byrne P, Papi A, Sergejeva S, Magnan A, Del Giacco S, Kalayci O, Hamelmann E, Papadopoulos NG. EAACI position statement on asthma exacerbations and severe asthma. Allergy 2013; 68:1520-31. [PMID: 24410781 PMCID: PMC7159478 DOI: 10.1111/all.12275] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2013] [Indexed: 02/02/2023]
Abstract
Asthma exacerbations and severe asthma are linked with high morbidity, significant mortality and high treatment costs. Recurrent asthma exacerbations cause a decline in lung function and, in childhood, are linked to development of persistent asthma. This position paper, from the European Academy of Allergy and Clinical Immunology, highlights the shortcomings of current treatment guidelines for patients suffering from frequent asthma exacerbations and those with difficult‐to‐treat asthma and severe treatment‐resistant asthma. It reviews current evidence that supports a call for increased awareness of (i) the seriousness of asthma exacerbations and (ii) the need for novel treatment strategies in specific forms of severe treatment‐resistant asthma. There is strong evidence linking asthma exacerbations with viral airway infection and underlying deficiencies in innate immunity and evidence of a synergism between viral infection and allergic mechanisms in increasing risk of exacerbations. Nonadherence to prescribed medication has been identified as a common clinical problem amongst adults and children with difficult‐to‐control asthma. Appropriate diagnosis, assessment of adherence and other potentially modifiable factors (such as passive or active smoking, ongoing allergen exposure, psychosocial factors) have to be a priority in clinical assessment of all patients with difficult‐to‐control asthma. Further studies with improved designs and new diagnostic tools are needed to properly characterize (i) the pathophysiology and risk of asthma exacerbations, and (ii) the clinical and pathophysiological heterogeneity of severe asthma.
Collapse
Affiliation(s)
- A. Custovic
- Centre Lead for Respiratory Medicine; Institute of Inflammation & Repair; University of Manchester; University Hospital of South Manchester; Manchester UK
| | - S. L. Johnston
- Airway Disease Infection Section; National Heart & Lung Institute; Imperial College London; Norfolk Place London UK
| | - I. Pavord
- Department of Respiratory Medicine; Thoracic Surgery and Allergy University Hospitals of Leicester NHS Trust; Glenfield Hospital; Leicester UK
| | - M. Gaga
- 7th Respiratory Medicine Department and Asthma Centre; Athens Chest Hospital; Athens Greece
| | - L. Fabbri
- Department of Oncology Haematology and Respiratory Diseases; University of Modena & Reggio Emilia; Modena Italy
| | - E. H. Bel
- Department of Respiratory Medicine; Academic Medical Centre; University of Amsterdam; Amsterdam the Netherlands
| | - P. Le Souëf
- Department of Respiratory Medicine; University of Western Australia; Princess Margaret Hospital for Children; Perth WA Australia
| | - J. Lötvall
- Krefting Research Centre Sahlgrenska Academy; University of Gothenburg; Göteborg Sweden
| | - P. Demoly
- Allergology Unit; Département de Pneumologie-Addictologie; Hôpital de Villeneuve - Inserm U657; Montpellier France
| | - C. A. Akdis
- Swiss Institute of Allergy & Asthma Research (SIAF); University of Zurich; Davos
- Christine-Kühne Center for Allergy Research and Education (CK-CARE); Davos Switzerland
| | - D. Ryan
- Woodbrook Medical Centre; Loughborough UK
| | - M. J. Mäkelä
- Skin and Allergy Hospital; Helsinki University Central Hospital; HUS Finland
| | - F. Martinez
- University of Arizona; Arizona Health Sciences Center; Pediatric Pulmonary Center; Tucson AZ USA
| | - J. W. Holloway
- University of Southampton; Faculty of Medicine Southampton General Hospital; Southampton UK
| | - S. Saglani
- National Heart & Lung Institute; Imperial College London; London UK
| | - P. O'Byrne
- Department of Medicine; Michael G DeGroote School of Medicine; McMaster University Faculty of Health Sciences; Hamilton ON Canada
| | - A. Papi
- Research Center on Asthma and COPD; University of Ferrara; Ferrara Italy
| | - S. Sergejeva
- Institute of Technology; University of Tartu; Tartu Estonia
| | - A. Magnan
- INSERM UMR915; l'institut du thorax; Faculté de Médecine; & Université de Nantes; Nantes France
| | - S. Del Giacco
- Department of Medical Science; University of Cagliari; Cagliari Italy
| | - O. Kalayci
- Pediatric Allergy and Asthma Unit; Ihsan Dogramaci Children's Hospital; Hacettepe University School of Medicine; Ankara Turkey
| | - E. Hamelmann
- Department of Pediatric Pneumology & Immunology; Charité Universitäts Berlin; Berlin Germany
- Department of Pediatrics; Ruhr-University Bochum; Bochum Germany
| | | |
Collapse
|
200
|
Katoh S, Shimizu H, Obase Y, Oomizu S, Niki T, Ikeda M, Mouri K, Kobashi Y, Hirashima M, Oka M. Preventive effect of galectin-9 on double-stranded RNA-induced airway hyperresponsiveness in an exacerbation model of mite antigen-induced asthma in mice. Exp Lung Res 2013; 39:453-62. [PMID: 24246013 DOI: 10.3109/01902148.2013.850558] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Viral respiratory infection is the most common cause of acute asthma exacerbation in patients with stable asthma. The replication of most respiratory viruses requires the generation of double-stranded RNA (dsRNA), resulting in the activation of host immune responses. Synthetic dsRNA, polyinosinic-polycytidylic acid (PolyIC), mimics the effects of viruses in various cell types. To evaluate new therapies for mite antigen-induced chronic asthma, we developed an acute exacerbation model of mouse chronic asthma using mite antigen and PolyIC. We also examined the preventive effects of recombinant galectin-9 (Gal-9) on acute asthma exacerbation in this model. METHODS Airway hyperresponsiveness (AHR) was examined to evaluate the exacerbation of chronic asthma. To analyze airway inflammation, the numbers of inflammatory cells and concentrations of cytokines in the bronchoalveolar lavage fluid (BALF) were estimated by flow cytometry and enzyme-linked immunosorbent assay, respectively. RESULTS AHR was accelerated by intranasal administration of PolyIC in addition to mite antigen. Levels of cytokines that contribute to AHR, including interferon-γ, tumor necrosis factor-α, and RANTES (CCR5), and of Gal-9 in the BALF were elevated in this acute asthma exacerbation mouse model. Intranasal administration of recombinant Gal-9 reduced the PolyIC-induced AHR and levels of these cytokines in the BALF. Further, Gal-9 suppressed the production of cytokines induced by PolyIC in the alveolar macrophages. CONCLUSIONS. Our findings demonstrated that exogenous Gal-9 suppressed dsRNA-induced AHR in an acute exacerbation model of chronic asthma in mice, and suggest that recombinant Gal-9 could be therapeutically effective for preventing acute asthma exacerbation.
Collapse
Affiliation(s)
- Shigeki Katoh
- 1Department of Respiratory Medicine, Kawasaki Medical School, Okayama, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|