51
|
Xander N, Reddy Vari H, Eskandar R, Li W, Bolla S, Marchetti N, Sajjan US. Rhinovirus-Induced SIRT-1 via TLR2 Regulates Subsequent Type I and Type III IFN Responses in Airway Epithelial Cells. THE JOURNAL OF IMMUNOLOGY 2019; 203:2508-2519. [PMID: 31548332 DOI: 10.4049/jimmunol.1900165] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 08/21/2019] [Indexed: 01/10/2023]
Abstract
IFN responses to viral infection are necessary to establish intrinsic antiviral state, but if unchecked can lead to heightened inflammation. Recently, we showed that TLR2 activation contributes to limitation of rhinovirus (RV)-induced IFN response in the airway epithelial cells. We also demonstrated that compared with normal airway epithelial cells, those from patients with chronic obstructive pulmonary disease (COPD) show higher IFN responses to RV, but the underlying mechanisms are not known. Initially, RV-induced IFN responses depend on dsRNA receptor activation and then are amplified via IFN-stimulated activation of JAK/STAT signaling. In this study, we show that in normal cells, TLR2 limits RV-induced IFN responses by attenuating STAT1 and STAT2 phosphorylation and this was associated with TLR2-dependent SIRT-1 expression. Further, inhibition of SIRT-1 enhanced RV-induced IFN responses, and this was accompanied by increased STAT1/STAT2 phosphorylation, indicating that TLR2 may limit RV-induced IFN responses via SIRT-1. COPD airway epithelial cells showed attenuated IL-8 responses to TLR2 agonist despite expressing TLR2 similar to normal, indicating dysregulation in TLR2 signaling pathway. Unlike normal, COPD cells failed to show RV-induced TLR2-dependent SIRT-1 expression. Pretreatment with quercetin, which increases SIRT-1 expression, normalized RV-induced IFN levels in COPD airway epithelial cells. Inhibition of SIRT-1 in quercetin-pretreated COPD cells abolished the normalizing effects of quercetin on RV-induced IFN expression in these cells, confirming that quercetin exerts its effect via SIRT-1. In summary, we show that TLR2 is required for limiting RV-induced IFNs, and this pathway is dysregulated in COPD airway epithelial cells, leading to exaggerated IFN production.
Collapse
Affiliation(s)
- Nathaniel Xander
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA 19140; and
| | - Hymavathi Reddy Vari
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA 19140; and
| | - Rewees Eskandar
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA 19140; and
| | - Wuyan Li
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA 19140; and
| | - Sudhir Bolla
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA 19140; and
| | - Nathaniel Marchetti
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA 19140; and
| | - Umadevi S Sajjan
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA 19140; and .,Department of Physiology, Temple University, Philadelphia, PA 19140
| |
Collapse
|
52
|
van Eeden SF, Hogg JC. Immune-Modulation in Chronic Obstructive Pulmonary Disease: Current Concepts and Future Strategies. Respiration 2019; 99:550-565. [PMID: 31480060 DOI: 10.1159/000502261] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 07/08/2019] [Indexed: 11/19/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is caused by the chronic inhalation of toxic particles and gases that are primarily but not exclusively derived from cigarette smoke that may be either actively or passively inhaled, which initiates a persistent innate and adaptive immune response in the lung. This immune response is associated with an aberrant tissue repair and remodeling process that results in varying degrees of chronic inflammation with excess production of mucus in the central airways and permanent destruction of the smaller conducting airways and gas exchanging surface in the peripheral lung. Currently, the primary aims of treatment in COPD are bronchodilation (inhaled short- and long-acting β-agonist and antimuscarinic therapies), to control symptoms and nonspecific broad-acting anti-inflammatory agents (inhaled and oral corticosteroids, phosphor-di-esterase inhibitors, and macrolides). That provide symptomatic relief but have little or no impact on either disease progression or mortality. As our understanding of the immune pathogenesis of the COPD improves, available immune modulation therapies have the potential to alter or interfere with damaging immune pathways, thereby slowing relentless progression of lung tissue destruction. The purpose of this brief review is to discuss our current understanding of the immune pathogenesis of both the airways and parenchymal injury as well as the dysfunctional tissue repair process to propose immune modulating interventions in an attempt to stabilize or return these pathological changes to their normal state. The ultimate goal of the immune modulation therapy is to improve both morbidity and mortality associated with COPD.
Collapse
Affiliation(s)
- Stephan F van Eeden
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada, .,Pacific Lung Health Centre, St. Paul's Hospital, Vancouver, British Columbia, Canada,
| | - James C Hogg
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
53
|
García-Valero J, Olloquequi J, Montes JF, Rodríguez E, Martín-Satué M, Texidó L, Ferrer Sancho J. Deficient pulmonary IFN-β expression in COPD patients. PLoS One 2019; 14:e0217803. [PMID: 31170225 PMCID: PMC6553750 DOI: 10.1371/journal.pone.0217803] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/18/2019] [Indexed: 12/27/2022] Open
Abstract
COPD patients are prone to acute infectious exacerbations that impair their quality of life and hamper prognosis. The purpose of the present study was to investigate the in situ IFN-β response in the lungs of stable COPD and non-COPD patients. Lung samples from 70 subjects (9 control never smokers, 19 control smokers without COPD, 21 patients with moderate COPD and 21 patients with very severe COPD) were studied for the expression of IFN-β, its main transcription factor, IRF-7, and two products of its autocrine function, namely RIG-I and MDA-5, by immunohistochemical techniques and quantitative real-time PCR. IFN-β, IRF-7, RIG-I and MDA-5 were widely detected in most lung cell types. In epithelial tissues and alveolar macrophages, IFN-β and IRF-7 labeling scores were decreased up to 65% and 74%, respectively, for COPD patients, paralleling an analogous reduction (43% and 65%, respectively) in the amount of their lung mRNA. Moreover, this decreased production of IFN-β in COPD patients correlated with a similar decrease in the expression of RIG-I and MDA-5, two essential members of the innate immune system. Our study indicates that most lung cells from stable COPD patients show a constitutive decreased expression of IFN-β, IRF-7, RIG-I and MDA-5, suggesting that this deficiency is the main cause of their acute viral exacerbations.
Collapse
Affiliation(s)
- José García-Valero
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Jordi Olloquequi
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | - Juan F. Montes
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Esther Rodríguez
- Department of Pneumology, Vall d’Hebron University Hospital, Universitat Autònoma de Barcelona and CIBER de Enfermedades Respiratorias (CIBERES), Barcelona, Spain
| | - Mireia Martín-Satué
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine, University of Barcelona, Barcelona, Spain
- Institut d’Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Laura Texidó
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Jaume Ferrer Sancho
- Department of Pneumology, Vall d’Hebron University Hospital, Universitat Autònoma de Barcelona and CIBER de Enfermedades Respiratorias (CIBERES), Barcelona, Spain
| |
Collapse
|
54
|
Vielle NJ, García-Nicolás O, Oliveira Esteves BI, Brügger M, Summerfield A, Alves MP. The Human Upper Respiratory Tract Epithelium Is Susceptible to Flaviviruses. Front Microbiol 2019; 10:811. [PMID: 31057517 PMCID: PMC6477545 DOI: 10.3389/fmicb.2019.00811] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 03/29/2019] [Indexed: 01/24/2023] Open
Abstract
Flaviviruses replicate in a wide variety of species and have a broad cellular tropism. They are isolated from various body fluids, and Zika virus (ZIKV), Japanese encephalitis virus (JEV), and West Nile virus (WNV) RNAs have been detected in nasopharyngeal swabs. Consequently, we evaluated the cellular tropism and host responses upon ZIKV, JEV, WNV, and Usutu virus (USUV) infection using a relevant model of the human upper respiratory tract epithelium based on primary human nasal epithelial cells (NECs) cultured at the air-liquid interface. NECs were susceptible to all the viruses tested, and confocal analysis showed evidence of infection of ciliated and non-ciliated cells. Each flavivirus productively infected NECs, leading to apical and basolateral live virus shedding with particularly high basal release for JEV and WNV. As demonstrated by a paracellular permeability assay, the integrity of the epithelium was not affected by flavivirus infection, suggesting an active release of live virus through the basolateral surface. Also, we detected a significant secretion of interferon type III and the pro-inflammatory cytokine IP-10/CXCL10 upon infection with JEV. Taken together, our data suggest that the human upper respiratory tract epithelium is a target for flaviviruses and could potentially play a role in the spread of infection to other body compartments through basolateral virus release. Undoubtedly, further work is required to evaluate the risks and define the adapted measures to protect individuals exposed to flavivirus-contaminated body fluids.
Collapse
Affiliation(s)
- Nathalie J Vielle
- Institute of Virology and Immunology, Bern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.,Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Obdulio García-Nicolás
- Institute of Virology and Immunology, Bern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Blandina I Oliveira Esteves
- Institute of Virology and Immunology, Bern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Melanie Brügger
- Institute of Virology and Immunology, Bern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland.,Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Artur Summerfield
- Institute of Virology and Immunology, Bern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Marco P Alves
- Institute of Virology and Immunology, Bern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
55
|
Bentley JK, Han M, Jaipalli S, Hinde JL, Lei J, Ishikawa T, Goldsmith AM, Rajput C, Hershenson MB. Myristoylated rhinovirus VP4 protein activates TLR2-dependent proinflammatory gene expression. Am J Physiol Lung Cell Mol Physiol 2019; 317:L57-L70. [PMID: 30908938 DOI: 10.1152/ajplung.00365.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Asthma exacerbations are often caused by rhinovirus (RV). We and others have shown that Toll-like receptor 2 (TLR2), a membrane surface receptor that recognizes bacterial lipopeptides and lipoteichoic acid, is required and sufficient for RV-induced proinflammatory responses in vitro and in vivo. We hypothesized that viral protein-4 (VP4), an internal capsid protein that is myristoylated upon viral replication and externalized upon viral binding, is a ligand for TLR2. Recombinant VP4 and myristoylated VP4 (MyrVP4) were purified by Ni-affinity chromatography. MyrVP4 was also purified from RV-A1B-infected HeLa cells by urea solubilization and anti-VP4 affinity chromatography. Finally, synthetic MyrVP4 was produced by chemical peptide synthesis. MyrVP4-TLR2 interactions were assessed by confocal fluorescence microscopy, fluorescence resonance energy transfer (FRET), and monitoring VP4-induced cytokine mRNA expression in the presence of anti-TLR2 and anti-VP4. MyrVP4 and TLR2 colocalized in TLR2-expressing HEK-293 cells, mouse bone marrow-derived macrophages, human bronchoalveolar macrophages, and human airway epithelial cells. Colocalization was absent in TLR2-null HEK-293 cells and blocked by anti-TLR2 and anti-VP4. Cy3-labeled MyrVP4 and Cy5-labeled anti-TLR2 showed an average fractional FRET efficiency of 0.24 ± 0.05, and Cy5-labeled anti-TLR2 increased and unlabeled MyrVP4 decreased FRET efficiency. MyrVP4-induced chemokine mRNA expression was higher than that elicited by VP4 alone and was attenuated by anti-TLR2 and anti-VP4. Cytokine expression was similarly increased by MyrVP4 purified from RV-infected HeLa cells and synthetic MyrVP4. We conclude that, during RV infection, MyrVP4 and TLR2 interact to generate a proinflammatory response.
Collapse
Affiliation(s)
- J Kelley Bentley
- Departments of Pediatrics and Communicable Diseases, University of Michigan , Ann Arbor, Michigan
| | - Mingyuan Han
- Departments of Pediatrics and Communicable Diseases, University of Michigan , Ann Arbor, Michigan
| | - Suraj Jaipalli
- Departments of Pediatrics and Communicable Diseases, University of Michigan , Ann Arbor, Michigan
| | - Joanna L Hinde
- Departments of Pediatrics and Communicable Diseases, University of Michigan , Ann Arbor, Michigan
| | - Jing Lei
- Departments of Pediatrics and Communicable Diseases, University of Michigan , Ann Arbor, Michigan
| | - Tomoko Ishikawa
- Departments of Pediatrics and Communicable Diseases, University of Michigan , Ann Arbor, Michigan
| | - Adam M Goldsmith
- Departments of Pediatrics and Communicable Diseases, University of Michigan , Ann Arbor, Michigan
| | - Charu Rajput
- Departments of Pediatrics and Communicable Diseases, University of Michigan , Ann Arbor, Michigan
| | - Marc B Hershenson
- Departments of Pediatrics and Communicable Diseases, University of Michigan , Ann Arbor, Michigan.,Department of Molecular and Integrative Physiology, University of Michigan , Ann Arbor, Michigan
| |
Collapse
|
56
|
Linden D, Guo-Parke H, Coyle PV, Fairley D, McAuley DF, Taggart CC, Kidney J. Respiratory viral infection: a potential "missing link" in the pathogenesis of COPD. Eur Respir Rev 2019; 28:28/151/180063. [PMID: 30872396 DOI: 10.1183/16000617.0063-2018] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 11/19/2018] [Indexed: 02/07/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is currently the third most common cause of global mortality. Acute exacerbations of COPD frequently necessitate hospital admission to enable more intensive therapy, incurring significant healthcare costs. COPD exacerbations are also associated with accelerated lung function decline and increased risk of mortality. Until recently, bacterial pathogens were believed to be responsible for the majority of disease exacerbations. However, with the advent of culture-independent molecular diagnostic techniques it is now estimated that viruses are detected during half of all COPD exacerbations and are associated with poorer clinical outcomes. Human rhinovirus, respiratory syncytial virus and influenza are the most commonly detected viruses during exacerbation. The role of persistent viral infection (adenovirus) has also been postulated as a potential pathogenic mechanism in COPD. Viral pathogens may play an important role in driving COPD progression by acting as triggers for exacerbation and subsequent lung function decline whilst the role of chronic viral infection remains a plausible hypothesis that requires further evaluation. There are currently no effective antiviral strategies for patients with COPD. Herein, we focus on the current understanding of the cellular and molecular mechanisms of respiratory viral infection in COPD.
Collapse
Affiliation(s)
- Dermot Linden
- Airway Innate Immunity Research Group (AiiR), Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast, UK
| | - Hong Guo-Parke
- Airway Innate Immunity Research Group (AiiR), Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast, UK
| | - Peter V Coyle
- The Regional Virus Laboratory, Belfast Trust, Belfast, UK
| | - Derek Fairley
- The Regional Virus Laboratory, Belfast Trust, Belfast, UK
| | - Danny F McAuley
- Airway Innate Immunity Research Group (AiiR), Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast, UK
| | - Clifford C Taggart
- Airway Innate Immunity Research Group (AiiR), Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Belfast, UK
| | - Joe Kidney
- Dept of Respiratory Medicine, Mater Hospital Belfast, Belfast, UK
| |
Collapse
|
57
|
Pro-inflammatory effects of extracellular Hsp70 and cigarette smoke in primary airway epithelial cells from COPD patients. Biochimie 2019; 156:47-58. [DOI: 10.1016/j.biochi.2018.09.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/25/2018] [Indexed: 11/18/2022]
|
58
|
Abstract
Respiratory viral infections including human rhinovirus (RV) infection have been identified as the most important environmental trigger of exacerbations of chronic lung diseases. While well established as the most common viral infections associated with exacerbations of asthma and chronic obstructive pulmonary disease, RVs and other respiratory viruses are also now thought to be important in triggering exacerbations of cystic fibrosis and the interstitial lung diseases. Here, we summarize the epidemiological evidence the supports respiratory viruses including RV as triggers of exacerbations of chronic lung diseases. We propose that certain characteristics of RVs may explain why they are the most common trigger of exacerbations of chronic lung diseases. We further highlight the latest mechanistic evidence supporting how and why common respiratory viral infections may enhance and promote disease triggering exacerbation events, through their interactions with the host immune system, and may be affected by ongoing treatments. We also provide a commentary on how new treatments may better manage the disease burden associated with respiratory viral infections and the exacerbation events that they trigger.
Collapse
|
59
|
Jubrail J, Africano-Gomez K, Herit F, Baturcam E, Mayer G, Cunoosamy DM, Kurian N, Niedergang F. HRV16 Impairs Macrophages Cytokine Response to a Secondary Bacterial Trigger. Front Immunol 2018; 9:2908. [PMID: 30619272 PMCID: PMC6305396 DOI: 10.3389/fimmu.2018.02908] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 11/27/2018] [Indexed: 11/17/2022] Open
Abstract
Human rhinovirus is frequently seen as an upper respiratory tract infection but growing evidence proves the virus can cause lower respiratory tract infections in patients with chronic inflammatory lung diseases including chronic obstructive pulmonary disease (COPD). In addition to airway epithelial cells, macrophages are crucial for regulating inflammatory responses to viral infections. However, the response of macrophages to HRV has not been analyzed in detail. We used in vitro monocyte-derived human macrophages to study the cytokine secretion of macrophages in response to the virus. Our results showed that macrophages were competent at responding to HRV, as a robust cytokine response was detected. However, after subsequent exposure to non-typeable Haemophilus influenzae (NTHi) or to LPS, HRV-treated macrophages secreted reduced levels of pro-inflammatory or regulatory cytokines. This “paralyzed” phenotype was not mimicked if the macrophages were pre-treated with LPS or CpG instead of the virus. These results begin to deepen our understanding into why patients with COPD show HRV-induced exacerbations and why they mount a defective response toward NTHi.
Collapse
Affiliation(s)
- Jamil Jubrail
- Institut Cochin, Inserm U1016, Paris, France.,CNRS, UMR 8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Kshanti Africano-Gomez
- Institut Cochin, Inserm U1016, Paris, France.,CNRS, UMR 8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Floriane Herit
- Institut Cochin, Inserm U1016, Paris, France.,CNRS, UMR 8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Engin Baturcam
- IMED Biotech Unit, Target and Translational Science, Respiratory, Inflammation & Autoimmunity, AstraZeneca, Gothenburg, Sweden
| | - Gaell Mayer
- Clinical Development, Respiratory Inhalation & Oral Development, GMD, AstraZeneca, Gothenburg, Sweden
| | - Danen Mootoosamy Cunoosamy
- IMED Biotech Unit, Target and Translational Science, Respiratory, Inflammation & Autoimmunity, AstraZeneca, Gothenburg, Sweden
| | - Nisha Kurian
- Precision Medicine & Genomics, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Florence Niedergang
- Institut Cochin, Inserm U1016, Paris, France.,CNRS, UMR 8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
60
|
Bortz E, Wu TT, Patel P, Whitelegge JP, Sun R. Proteomics of Bronchoalveolar Lavage Fluid Reveals a Lung Oxidative Stress Response in Murine Herpesvirus-68 Infection. Viruses 2018; 10:v10120670. [PMID: 30486363 PMCID: PMC6316452 DOI: 10.3390/v10120670] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 11/15/2018] [Accepted: 11/20/2018] [Indexed: 12/25/2022] Open
Abstract
Murine herpesvirus-68 (MHV-68) productively infects mouse lungs, exhibiting a complex pathology characteristic of both acute viral infections and chronic respiratory diseases. We sought to discover proteins differentially expressed in bronchoalveolar lavage (BAL) from mice infected with MHV-68. Mice were infected intranasally with MHV-68. After nine days, as the lytic phase of infection resolved, differential BAL proteins were identified by two-dimensional (2D) electrophoresis and mass spectrometry. Of 23 unique proteins, acute phase proteins, vitamin A transport, and oxidative stress response factors Pdx6 and EC-SOD (Sod3) were enriched. Correspondingly, iNOS2 was induced in lung tissue by seven days post-infection. Oxidative stress was partly a direct result of MHV-68 infection, as reactive oxygen species (ROS) were induced in cultured murine NIH3T3 fibroblasts and human lung A549 cells infected with MHV-68. Finally, mice infected with a recombinant MHV-68 co-expressing inflammatory cytokine murine interleukin 6 (IL6) showed exacerbated oxidative stress and soluble type I collagen characteristic of tissue recovery. Thus, oxidative stress appears to be a salient feature of MHV-68 pathogenesis, in part caused by lytic replication of the virus and IL6. Proteins and small molecules in lung oxidative stress networks therefore may provide new therapeutic targets to ameliorate respiratory virus infections.
Collapse
Affiliation(s)
- Eric Bortz
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, AK 99508, USA.
| | - Ting-Ting Wu
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| | - Parthive Patel
- Center for Molecular Biology and German Cancer Research Center (DKFZ), University of Heidelberg (ZMBH), 69120 Heidelberg, Germany.
| | - Julian P Whitelegge
- The Pasarow Mass Spectrometry Laboratory & the Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| | - Ren Sun
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
61
|
Targeting Cytokines as Evolving Treatment Strategies in Chronic Inflammatory Airway Diseases. Int J Mol Sci 2018; 19:ijms19113402. [PMID: 30380761 PMCID: PMC6275012 DOI: 10.3390/ijms19113402] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/25/2018] [Accepted: 10/27/2018] [Indexed: 12/13/2022] Open
Abstract
Cytokines are key players in the initiation and propagation of inflammation in chronic inflammatory airway diseases such as chronic obstructive pulmonary disease (COPD), bronchiectasis and allergic asthma. This makes them attractive targets for specific novel anti-inflammatory treatment strategies. Recently, both interleukin-1 (IL-1) and IL-6 have been associated with negative health outcomes, mortality and a pro-inflammatory phenotype in COPD. IL-6 in COPD was shown to correlate negatively with lung function, and IL-1beta was induced by cigarette smoke in the bronchial epithelium, causing airway inflammation. Furthermore, IL-8 has been shown to be a pro-inflammatory marker in bronchiectasis, COPD and allergic asthma. Clinical trials using specific cytokine blockade therapies are currently emerging and have contributed to reduce exacerbations and steroid use in COPD. Here, we present a review of the current understanding of the roles of cytokines in the pathophysiology of chronic inflammatory airway diseases. Furthermore, outcomes of clinical trials in cytokine blockade as novel treatment strategies for selected patient populations with those diseases will be discussed.
Collapse
|
62
|
Lo D, Kennedy JL, Kurten RC, Panettieri RA, Koziol-White CJ. Modulation of airway hyperresponsiveness by rhinovirus exposure. Respir Res 2018; 19:208. [PMID: 30373568 PMCID: PMC6206673 DOI: 10.1186/s12931-018-0914-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 10/17/2018] [Indexed: 01/12/2023] Open
Abstract
Rhinovirus (RV) exposure has been implicated in childhood development of wheeze evoking asthma and exacerbations of underlying airways disease. Studies such as the Copenhagen Prospective Studies on Asthma in Childhood (COPSAC) and Childhood Origins of ASThma (COAST) have identified RV as a pathogen inducing severe respiratory disease. RVs also modulate airway hyperresponsiveness (AHR), a key characteristic of such diseases. Although potential factors underlying mechanisms by which RV induces AHR have been postulated, the precise mechanisms of AHR following RV exposure remain elusive. A challenge to RV-related research stems from inadequate models for study. While human models raise ethical concerns and are relatively difficult in terms of subject recruitment, murine models are limited by susceptibility of infection to the relatively uncommon minor group (RV-B) serotypes, strains that are generally associated with infrequent clinical respiratory virus infections. Although a transgenic mouse strain that has been developed has enhanced susceptibility for infection with the common major group (RV-A) serotypes, few studies have focused on RV in the context of allergic airways disease rather than understanding RV-induced AHR. Recently, the receptor for the virulent RV-C CDHR3, was identified, but a dearth of studies have examined RV-C-induced effects in humans. Currently, the mechanisms by which RV infections modulate airway smooth muscle (ASM) shortening or excitation-contraction coupling remain elusive. Further, only one study has investigated the effects of RV on bronchodilatory mechanisms, with only speculation as to mechanisms underlying RV-mediated modulation of bronchoconstriction.
Collapse
Affiliation(s)
- Dennis Lo
- Department of Medicine, Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, NJ, USA
| | - Joshua L Kennedy
- Department of Pediatrics, Division of Allergy and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Richard C Kurten
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Reynold A Panettieri
- Department of Medicine, Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, NJ, USA
| | - Cynthia J Koziol-White
- Department of Medicine, Rutgers Institute for Translational Medicine and Science, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
63
|
|
64
|
Jing H, Liu L, Zhou J, Yao H. Inhibition of C-X-C Motif Chemokine 10 (CXCL10) Protects Mice from Cigarette Smoke-Induced Chronic Obstructive Pulmonary Disease. Med Sci Monit 2018; 24:5748-5753. [PMID: 30118441 PMCID: PMC6109363 DOI: 10.12659/msm.909864] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a type of obstructive lung disease characterized by long-term breathing problems and poor airflow. COPD can progress to persistent decline of pulmonary function. This study explored the effect of CXCL10 on COPD induced by cigarette smoke (CS) and its underlying mechanism. Material/Methods Wild-type (WT) mice were randomly assigned into 3 groups: the control group, the CS group, and the intervention group. Mice in the CS group were exposed to CS and mice in the CXCL10 group were exposed to CS and CXCL10 neutralizing antibody. At 24 h after the last CS exposure, body weight and lung functions of each mouse were recorded. Mice were then anesthetized for collecting bronchoalveolar lavage fluid (BALF) and lung tissues. Levels of interleukin-6 (IL-6), keratinocyte chemotactic factor (KC), and monocyte chemoattractant protein-1 (MCP-1) in supernatant and lung homogenate were detected by ELISA and real-time PCR (RT-PCR), respectively. For in vitro experiments, human bronchial epithelial cells 16HBE were stimulated with different concentrations of cigarette smoke extract (CSE) and CXCL10. Cell viability and levels of inflammatory cytokines in the cell supernatant were detected by Cell Counting Kit-8 (CCK-8) and ELISA assay, respectively. Results Our data showed significant weight loss and reduction of lung functions in mice in the CS group compared with those in the control group and intervention group. Increased levels of IL-6, KC, and MCP-1 in BALF and lung homogenate were observed in mice in the model group compared to those in the control group and intervention group. In vitro experiments also confirmed that CXCL10-neutralizing antibody can inhibit CSE-induced cell necrosis and activation of inflammatory cytokines. Conclusions Inhibited CXCL10 protects against COPD progression by decreasing secretion of inflammatory factors, which provides a new direction for the clinical prevention and treatment of COPD.
Collapse
Affiliation(s)
- Hongyu Jing
- Department of Respiratory Medicine, First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Lingyun Liu
- Department of Andrology, First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Junfeng Zhou
- Department of Dermatology and Venereology, First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Hanxin Yao
- Department of Clinical Laboratory, First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| |
Collapse
|
65
|
Khomich OA, Kochetkov SN, Bartosch B, Ivanov AV. Redox Biology of Respiratory Viral Infections. Viruses 2018; 10:392. [PMID: 30049972 PMCID: PMC6115776 DOI: 10.3390/v10080392] [Citation(s) in RCA: 257] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/17/2018] [Accepted: 07/24/2018] [Indexed: 12/16/2022] Open
Abstract
Respiratory viruses cause infections of the upper or lower respiratory tract and they are responsible for the common cold-the most prevalent disease in the world. In many cases the common cold results in severe illness due to complications, such as fever or pneumonia. Children, old people, and immunosuppressed patients are at the highest risk and require fast diagnosis and therapeutic intervention. However, the availability and efficiencies of existing therapeutic approaches vary depending on the virus. Investigation of the pathologies that are associated with infection by respiratory viruses will be paramount for diagnosis, treatment modalities, and the development of new therapies. Changes in redox homeostasis in infected cells are one of the key events that is linked to infection with respiratory viruses and linked to inflammation and subsequent tissue damage. Our review summarizes current knowledge on changes to redox homeostasis, as induced by the different respiratory viruses.
Collapse
Affiliation(s)
- Olga A Khomich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov str, 32, 119991 Moscow, Russia.
- Inserm U1052, Cancer Research Center Lyon, University of Lyon, 69000 Lyon, France.
| | - Sergey N Kochetkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov str, 32, 119991 Moscow, Russia.
| | - Birke Bartosch
- Inserm U1052, Cancer Research Center Lyon, University of Lyon, 69000 Lyon, France.
- DevWeCan Laboratories of Excellence Network (Labex), 69003 Lyon, France.
| | - Alexander V Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov str, 32, 119991 Moscow, Russia.
| |
Collapse
|
66
|
Jing Y, Gimenes JA, Mishra R, Pham D, Comstock AT, Yu D, Sajjan U. NOTCH3 contributes to rhinovirus-induced goblet cell hyperplasia in COPD airway epithelial cells. Thorax 2018; 74:18-32. [PMID: 29991510 DOI: 10.1136/thoraxjnl-2017-210593] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 05/15/2018] [Accepted: 06/11/2018] [Indexed: 11/04/2022]
Abstract
RATIONALE Goblet cell hyperplasia (GCH) is one of the cardinal features of chronic obstructive pulmonary disease (COPD) and contributes to airways obstruction. Rhinovirus (RV), which causes acute exacerbations in patients with COPD, also causes prolonged airways obstruction. Previously, we showed that RV enhances mucin gene expression and increases goblet cell number in a COPD mouse model. This study examines whether RV causes sustained GCH in relevant models of COPD. METHODS Mucociliary-differentiated COPD and normal airway epithelial cell cultures and mice with normal or COPD phenotype were infected with RV or sham and examined for GCH by immunofluorescence and/or mucin gene expression. In some experiments, RV-infected COPD cells and mice with COPD phenotype were treated with γ-secretase inhibitor or interleukin-13 neutralising antibody and assessed for GCH. To determine the contribution of NOTCH1/3 in RV-induced GCH, COPD cells transduced with NOTCH1/3 shRNA were used. RESULTS RV-infected COPD, but not normal cell cultures, showed sustained GCH and increased mucin genes expression. Microarray analysis indicated increased expression of NOTCH1, NOTCH3 and HEY1 only in RV-infected COPD cells. Blocking NOTCH3, but not NOTCH1, attenuated RV-induced GCH in vitro. Inhibition of NOTCH signalling by γ-secretase inhibitor, but not neutralising antibody to IL-13, abrogated RV-induced GCH and mucin gene expression. CONCLUSIONS RV induces sustained GCH via NOTCH3 particularly in COPD cells or mice with COPD phenotype. This may be one of the mechanisms that may contribute to RV-induced prolonged airways obstruction in COPD.
Collapse
Affiliation(s)
- Yaxun Jing
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Joao Antonio Gimenes
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Rahul Mishra
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Duc Pham
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Adam T Comstock
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Daohai Yu
- Department of Clinical Sciences, Temple University, Philadelphia, Pennsylvania, USA
| | - Umadevi Sajjan
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, USA.,Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, Pennsylvania, USA.,Department of Physiology, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
67
|
Farazuddin M, Mishra R, Jing Y, Srivastava V, Comstock AT, Sajjan US. Quercetin prevents rhinovirus-induced progression of lung disease in mice with COPD phenotype. PLoS One 2018; 13:e0199612. [PMID: 29975735 PMCID: PMC6033397 DOI: 10.1371/journal.pone.0199612] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/27/2018] [Indexed: 12/14/2022] Open
Abstract
Acute exacerbations are the major cause of morbidity and mortality in patients with chronic obstructive pulmonary disease (COPD). Rhinovirus, which causes acute exacerbations may also accelerate progression of lung disease in these patients. Current therapies reduces the respiratory symptoms and does not treat the root cause of exacerbations effectively. We hypothesized that quercetin, a potent antioxidant and anti-inflammatory agent with antiviral properties may be useful in treating rhinovirus-induced changes in COPD. Mice with COPD phenotype maintained on control or quercetin diet and normal mice were infected with sham or rhinovirus, and after 14 days mice were examined for changes in lung mechanics and lung inflammation. Rhinovirus-infected normal mice showed no changes in lung mechanics or histology. In contrast, rhinovirus-infected mice with COPD phenotype showed reduction in elastic recoiling and increase in lung inflammation, goblet cell metaplasia, and airways cholinergic responsiveness compared to sham-infected mice. Interestingly, rhinovirus-infected mice with COPD phenotype also showed accumulation of neutrophils, CD11b+/CD11c+ macrophages and CD8+ T cells in the lungs. Quercetin supplementation attenuated rhinovirus-induced all the pathologic changes in mice with COPD phenotype. Together these results indicate that quercetin effectively mitigates rhinovirus-induced progression of lung disease in a mouse model of COPD. Therefore, quercetin may be beneficial in the treatment of rhinovirus-associated exacerbations and preventing progression of lung disease in COPD.
Collapse
Affiliation(s)
- Mohammad Farazuddin
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Rahul Mishra
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Yaxun Jing
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Vikram Srivastava
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Adam T. Comstock
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Umadevi S. Sajjan
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, Pennsylvania, United States of America
- Department of Physiology, Temple University, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
68
|
Jiang D, Schaefer N, Chu HW. Air-Liquid Interface Culture of Human and Mouse Airway Epithelial Cells. Methods Mol Biol 2018; 1809:91-109. [PMID: 29987785 DOI: 10.1007/978-1-4939-8570-8_8] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Air-liquid interface culture enables airway epithelial cells to differentiate into a pseudostratified cell layer, consisting of ciliated cells, goblet/secretory cells, and basal cells (Ghio et al., Part Fibre Toxicol 10:25, 2013). This technique is critically important for in vitro studies of lung diseases such as asthma, chronic obstructive pulmonary disease, and cystic fibrosis, since differentiated airway epithelial cells are more representative of the in vivo lung environment than non-differentiated cells (Derichs et al., FASEB J 25:2325-2332, 2011; Hackett et al., Am J Respir Cell Mol Biol 45:1090-1100, 2011;Schneider et al., Am J Respir Crit Care Med 182: 332-340, 2010). Here we describe the process of isolating and expanding human and mouse airway epithelial cells, as well as differentiation of airway epithelial cells by air-liquid interface culture.
Collapse
Affiliation(s)
- Di Jiang
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | | | - Hong Wei Chu
- Department of Medicine, National Jewish Health, Denver, CO, USA.
| |
Collapse
|
69
|
The Role of Nicotinamide Adenine Dinucleotide Phosphate Oxidases in Lung Architecture Remodeling. Antioxidants (Basel) 2017; 6:antiox6040104. [PMID: 29257052 PMCID: PMC5745514 DOI: 10.3390/antiox6040104] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 12/08/2017] [Accepted: 12/14/2017] [Indexed: 02/07/2023] Open
Abstract
Chronic lung disorders, such as pulmonary artery hypertension (PAH), chronic obstructive pulmonary disease (COPD), asthma and neonatal bronchopulmonary dysplasia (BPD), are characterized by airway and/or vascular remodeling. Despite differences in the pathology, reactive oxygen species (ROS) have been highlighted as a critical contributor to the initiation and development of airway and vascular remodeling. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (Nox) appear to play a pivotal role in lung signaling, leading to marked changes in pulmonary airway and vascular cell phenotypes, including proliferation, hypertrophy and apoptosis. In this review, we summarized the current literature regarding the role of Nox in the airway and vascular remodeling.
Collapse
|
70
|
Wu Q, Jiang D, Schaefer NR, Harmacek L, O'Connor BP, Eling TE, Eickelberg O, Chu HW. Overproduction of growth differentiation factor 15 promotes human rhinovirus infection and virus-induced inflammation in the lung. Am J Physiol Lung Cell Mol Physiol 2017; 314:L514-L527. [PMID: 29192094 DOI: 10.1152/ajplung.00324.2017] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Human rhinovirus (HRV) is the most common virus contributing to acute exacerbations of chronic obstructive pulmonary disease (COPD) nearly year round, but the mechanisms have not been well elucidated. Recent clinical studies suggest that high levels of growth differentiation factor 15 (GDF15) protein in the blood are associated with an increased yearly rate of all-cause COPD exacerbations. Therefore, in the current study, we investigated whether GDF15 promotes HRV infection and virus-induced lung inflammation. We first examined the role of GDF15 in regulating host defense and HRV-induced inflammation using human GDF15 transgenic mice and cultured human GDF15 transgenic mouse tracheal epithelial cells. Next, we determined the effect of GDF15 on viral replication, antiviral responses, and inflammation in human airway epithelial cells with GDF15 knockdown and HRV infection. Finally, we explored the signaling pathways involved in airway epithelial responses to HRV infection in the context of GDF15. Human GDF15 protein overexpression in mice led to exaggerated inflammatory responses to HRV, increased infectious particle release, and decreased IFN-λ2/3 (IL-28A/B) mRNA expression in the lung. Moreover, GDF15 facilitated HRV replication and inflammation via inhibiting IFN-λ1/IL-29 protein production in human airway epithelial cells. Lastly, Smad1 cooperated with interferon regulatory factor 7 (IRF7) to regulate airway epithelial responses to HRV infection partly via GDF15 signaling. Our results reveal a novel function of GDF15 in promoting lung HRV infection and virus-induced inflammation, which may be a new mechanism for the increased susceptibility and severity of respiratory viral (i.e., HRV) infection in cigarette smoke-exposed airways with GDF15 overproduction.
Collapse
Affiliation(s)
- Qun Wu
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus , Aurora, Colorado.,Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health , Denver, Colorado
| | - Di Jiang
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health , Denver, Colorado
| | - Niccolette R Schaefer
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health , Denver, Colorado
| | - Laura Harmacek
- Center for Genes, Environment, and Health, National Jewish Health , Denver, Colorado
| | - Brian P O'Connor
- Center for Genes, Environment, and Health, National Jewish Health , Denver, Colorado
| | - Thomas E Eling
- The Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health , Research Triangle Park, North Carolina
| | - Oliver Eickelberg
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus , Aurora, Colorado
| | - Hong Wei Chu
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, National Jewish Health , Denver, Colorado
| |
Collapse
|
71
|
Jiang Q, Wang F, Shi L, Zhao X, Gong M, Liu W, Song C, Li Q, Chen Y, Wu H, Han D. C-X-C motif chemokine ligand 10 produced by mouse Sertoli cells in response to mumps virus infection induces male germ cell apoptosis. Cell Death Dis 2017; 8:e3146. [PMID: 29072682 PMCID: PMC5680925 DOI: 10.1038/cddis.2017.560] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 09/12/2017] [Accepted: 09/20/2017] [Indexed: 12/17/2022]
Abstract
Mumps virus (MuV) infection usually results in germ cell degeneration in the testis, which is an etiological factor for male infertility. However, the mechanisms by which MuV infection damages male germ cells remain unclear. The present study showed that C-X-C motif chemokine ligand 10 (CXCL10) is produced by mouse Sertoli cells in response to MuV infection, which induces germ cell apoptosis through the activation of caspase-3. CXC chemokine receptor 3 (CXCR3), a functional receptor of CXCL10, is constitutively expressed in male germ cells. Neutralizing antibodies against CXCR3 and an inhibitor of caspase-3 activation significantly inhibited CXCL10-induced male germ cell apoptosis. Furthermore, the tumor necrosis factor-α (TNF-α) upregulated CXCL10 production in Sertoli cells after MuV infection. The knockout of either CXCL10 or TNF-α reduced germ cell apoptosis in the co-cultures of germ cells and Sertoli cells in response to MuV infection. Local injection of MuV into the testes of mice confirmed the involvement of CXCL10 in germ cell apoptosis in vivo. These results provide novel insights into MuV-induced germ cell apoptosis in the testis.
Collapse
Affiliation(s)
- Qian Jiang
- Department of Cell Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Fei Wang
- Department of Cell Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Lili Shi
- Department of Cell Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Xiang Zhao
- Department of Cell Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Maolei Gong
- Department of Cell Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Weihua Liu
- Department of Cell Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Chengyi Song
- Joint International Research Laboratory of Agriculture and Agri-product Safety, Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Qihan Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences, Kunming, China
| | - Yongmei Chen
- Department of Cell Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Han Wu
- Department of Cell Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China.,Joint International Research Laboratory of Agriculture and Agri-product Safety, Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Daishu Han
- Department of Cell Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| |
Collapse
|
72
|
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is characterized by irreversible airflow limitation. It is a global disease and expected to be the third leading cause of death. Respiratory exacerbations are associated with increased mortality and morbidity in this patient population. Respiratory viruses were isolated from at least 30 to 50% of the infectious respiratory COPD exacerbations with rhinovirus being most commonly isolated pathogen. Although rhinovirus does not cause airway epithelial damage like influenza and other respiratory viruses, it may further impair innate immunity of airway epithelium, which is the first line of defense in the lungs. This may increase susceptibility to secondary bacterial infections leading to progression of lung disease. Currently, there arc no therapies available to treat rhinovirus infection in COPD and therefore understanding the mechanisms underlying RV pathogenesis in COPD is essential to identify molecular target to develop new therapeutic strategies. Quercetin, a plant polyphenol, which modulates innate immunity and effectively blocks viral replication may be useful in treating rhinovirus associated COPD exacerbations.
Collapse
Affiliation(s)
- Nicole Owuor
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA, USA
| | - Nisha Nalamala
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA, USA
| | - Joao Antonio Gimenes
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA, USA
| | - Uma S Sajjan
- Department of Thoracic Surgery and Medicine, Temple University, Philadelphia, PA, USA.,Department of Physiology, Temple University, Philadelphia, PA, USA
| |
Collapse
|
73
|
Mertens TCJ, Karmouty-Quintana H, Taube C, Hiemstra PS. Use of airway epithelial cell culture to unravel the pathogenesis and study treatment in obstructive airway diseases. Pulm Pharmacol Ther 2017; 45:101-113. [PMID: 28502841 DOI: 10.1016/j.pupt.2017.05.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 04/19/2017] [Accepted: 05/10/2017] [Indexed: 12/12/2022]
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) are considered as two distinct obstructive diseases. Both chronic diseases share a component of airway epithelial dysfunction. The airway epithelium is localized to deal with inhaled substances, and functions as a barrier preventing penetration of such substances into the body. In addition, the epithelium is involved in the regulation of both innate and adaptive immune responses following inhalation of particles, allergens and pathogens. Through triggering and inducing immune responses, airway epithelial cells contribute to the pathogenesis of both asthma and COPD. Various in vitro research models have been described to study airway epithelial cell dysfunction in asthma and COPD. However, various considerations and cautions have to be taken into account when designing such in vitro experiments. Epithelial features of asthma and COPD can be modelled by using a variety of disease-related invoking substances either alone or in combination, and by the use of primary cells isolated from patients. Differentiation is a hallmark of airway epithelial cells, and therefore models should include the ability of cells to differentiate, as can be achieved in air-liquid interface models. More recently developed in vitro models, including precision cut lung slices, lung-on-a-chip, organoids and human induced pluripotent stem cells derived cultures, provide novel state-of-the-art alternatives to the conventional in vitro models. Furthermore, advanced models in which cells are exposed to respiratory pathogens, aerosolized medications and inhaled toxic substances such as cigarette smoke and air pollution are increasingly used to model e.g. acute exacerbations. These exposure models are relevant to study how epithelial features of asthma and COPD are affected and provide a useful tool to study the effect of drugs used in treatment of asthma and COPD. These new developments are expected to contribute to a better understanding of the complex gene-environment interactions that contribute to development and progression of asthma and COPD.
Collapse
Affiliation(s)
- Tinne C J Mertens
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands; Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Christian Taube
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
74
|
Acute cigarette smoke exposure activates apoptotic and inflammatory programs but a second stimulus is required to induce epithelial to mesenchymal transition in COPD epithelium. Respir Res 2017; 18:82. [PMID: 28468623 PMCID: PMC5415733 DOI: 10.1186/s12931-017-0565-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 04/27/2017] [Indexed: 12/23/2022] Open
Abstract
Background Smoking and aberrant epithelial responses are risk factors for lung cancer as well as chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. In these conditions, disease progression is associated with epithelial damage and fragility, airway remodelling and sub-epithelial fibrosis. The aim of this study was to assess the acute effects of cigarette smoke on epithelial cell phenotype and pro-fibrotic responses in vitro and in vivo. Results Apoptosis was significantly greater in unstimulated cells from COPD patients compared to control, but proliferation and CXCL8 release were not different. Cigarette smoke dose-dependently induced apoptosis, proliferation and CXCL8 release with normal epithelial cells being more responsive than COPD patient derived cells. Cigarette smoke did not induce epithelial-mesenchymal transition. In vivo, cigarette smoke exposure promoted epithelial apoptosis and proliferation. Moreover, mimicking a virus-induced exacerbation by exposing to mice to poly I:C, exaggerated the inflammatory responses, whereas expression of remodelling genes was similar in both. Conclusions Collectively, these data indicate that cigarette smoke promotes epithelial cell activation and hyperplasia, but a secondary stimulus is required for the remodelling phenotype associated with COPD. Electronic supplementary material The online version of this article (doi:10.1186/s12931-017-0565-2) contains supplementary material, which is available to authorized users.
Collapse
|
75
|
Jiang Y, Wang X, Hu D. Mitochondrial alterations during oxidative stress in chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis 2017; 12:1153-1162. [PMID: 28458526 PMCID: PMC5402882 DOI: 10.2147/copd.s130168] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The high incidence of chronic obstructive pulmonary disease (COPD), one of the most prevalent diseases worldwide, has attracted growing attention. Cigarette smoking is considered a major contributory factor in the pathogenesis and progression of COPD due to the tremendous oxidative burden that it causes, which induces an oxidant/antioxidant imbalance. Excessive oxidation induced by the excessive generation of mitochondrial reactive oxygen species disturbs the antioxidant systems and plays an important role in triggering and promoting chronic inflammation of airways. Given that mitochondria is one of the main sites of reactive oxygen species production by the oxidative phosphorylation process, oxidative stress may affect mitochondrial function by changing its structure and morphology and by affecting a series of mitochondrial proteins. In particular, PTEN-induced putative kinase 1/Parkin and p62 play critical roles in mitophagy. During the process, the Akt ubiquitin E3 ligase is an important mediator associated with cigarette smoke exposure-induced pulmonary endothelial cell death and dysfunction. Thus, understanding the underlying mechanisms of the signaling pathway may provide important information regarding the therapeutic treatment of COPD by application of alternative PTEN-induced putative kinase 1 targets or ubiquitin E3 ligase.
Collapse
Affiliation(s)
- Ying Jiang
- Department of Clinical Pharmacology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xiaoqin Wang
- Department of Clinical Pharmacology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Daode Hu
- Department of Clinical Pharmacology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
76
|
Hung IFN, Zhang AJ, To KKW, Chan JFW, Zhu SHS, Zhang R, Chan TC, Chan KH, Yuen KY. Unexpectedly Higher Morbidity and Mortality of Hospitalized Elderly Patients Associated with Rhinovirus Compared with Influenza Virus Respiratory Tract Infection. Int J Mol Sci 2017; 18:ijms18020259. [PMID: 28134768 PMCID: PMC5343795 DOI: 10.3390/ijms18020259] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 01/19/2017] [Accepted: 01/23/2017] [Indexed: 11/17/2022] Open
Abstract
Rhinovirus is a common cause of upper and lower respiratory tract infections in adults, especially among the elderly and immunocompromised. Nevertheless, its clinical characteristics and mortality risks have not been well described. A retrospective analysis on a prospective cohort was conducted in a single teaching hospital center over a one-year period. We compared adult patients hospitalized for pneumonia caused by rhinovirus infection with those hospitalized for influenza infection during the same period. All recruited patients were followed up for at least 3 months up to 15 months. Independent risk factors associated with mortality for rhinovirus infection were identified. Between 1 March 2014 and 28 February 2015, a total of 1946 patients were consecutively included for analysis. Of these, 728 patients were hospitalized for rhinovirus infection and 1218 patients were hospitalized for influenza infection. Significantly more rhinovirus patients were elderly home residents and had chronic lung diseases (p < 0.001), whereas more influenza patients had previous stroke (p = 0.02); otherwise, there were no differences in the Charlson comorbidity indexes between the two groups. More patients in the rhinovirus group developed pneumonia complications (p = 0.03), required oxygen therapy, and had a longer hospitalization period (p < 0.001), whereas more patients in the influenza virus group presented with fever (p < 0.001) and upper respiratory tract symptoms of cough and sore throat (p < 0.001), and developed cardiovascular complications (p < 0.001). The 30-day (p < 0.05), 90-day (p < 0.01), and 1-year (p < 0.01) mortality rate was significantly higher in the rhinovirus group than the influenza virus group. Intensive care unit admission (odds ratio (OR): 9.56; 95% confidence interval (C.I.) 2.17–42.18), elderly home residents (OR: 2.60; 95% C.I. 1.56–4.33), requirement of oxygen therapy during hospitalization (OR: 2.62; 95% C.I. 1.62–4.24), and hemoglobin level <13.3 g/dL upon admission (OR: 2.43; 95% C.I. 1.16–5.12) were independent risk factors associated with 1-year mortality in patients hospitalized for rhinovirus infection. Rhinovirus infection in the adults was associated with significantly higher mortality and longer hospitalization when compared with influenza virus infection. Institutionalized older adults were particularly at risk. More stringent infection control among health care workers in elderly homes could lower the infection rate before an effective vaccine and antiviral become available.
Collapse
Affiliation(s)
- Ivan F N Hung
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China.
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Pokfulam, Hong Kong SAR, China.
| | - Anna Jinxia Zhang
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China.
| | - Kelvin K W To
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China.
| | - Jasper F W Chan
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China.
| | - Shawn H S Zhu
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China.
| | - Ricky Zhang
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China.
| | - Tuen-Ching Chan
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, 102 Pokfulam Road, Pokfulam, Hong Kong SAR, China.
| | - Kwok-Hung Chan
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China.
| | - Kwok-Yung Yuen
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu's Centre for Infection and Division of Infectious Diseases, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China.
| |
Collapse
|
77
|
Abstract
Chronic obstructive pulmonary disease (COPD) is a disease with high prevalence and substantial associated economical burden. A significant determinant of quality of life, long-term survival, and health care costs is an acute exacerbation of COPD. Acute exacerbations are provoked by respiratory viruses, altered airway microbiome, and environmental factors. The current treatment options are limited. In order to develop specific therapeutic measures, it is important to understand how acute exacerbations evolve. This review focuses on pathophysiology of stable and exacerbated COPD.
Collapse
Affiliation(s)
- Xianghui Zhou
- Department of Respiratory Medicine, The First Xuzhou People's Hospital, 19 Zhongshan North Road, Xuzhou, 221003, Jiangsu, China
| | - Qingling Li
- Department of Respiratory Medicine, The First Xuzhou People's Hospital, 19 Zhongshan North Road, Xuzhou, 221003, Jiangsu, China.
| | - Xincan Zhou
- Department of Respiratory Medicine, The First Xuzhou People's Hospital, 19 Zhongshan North Road, Xuzhou, 221003, Jiangsu, China
| |
Collapse
|
78
|
Zavala J, O'Brien B, Lichtveld K, Sexton KG, Rusyn I, Jaspers I, Vizuete W. Assessment of biological responses of EpiAirway 3-D cell constructs versus A549 cells for determining toxicity of ambient air pollution. Inhal Toxicol 2017; 28:251-9. [PMID: 27100558 DOI: 10.3109/08958378.2016.1157227] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONTEXT EpiAirway™ 3-D constructs are human-derived cell cultures of differentiated airway epithelial cells that may represent a more biologically relevant model of the human lung. However, limited information is available on their utility for exposures to air pollutants at the air-liquid interface (ALI). OBJECTIVE To assess the biological responses of EpiAirway™ cells in comparison to the responses of A549 human alveolar epithelial cells after exposure to air pollutants at ALI. METHODS Cells were exposed to filtered air, 400 ppb of ozone (O3) or a photochemically aged Synthetic Urban Mixture (SynUrb54) consisting of hydrocarbons, nitrogen oxides, O3 and other secondary oxidation products for 4 h. Basolateral supernatants and apical washes were collected at 9 and 24 h post-exposure. We assessed cytotoxicity by measuring lactate dehydrogenase (LDH) release into the culture medium and apical surface. Interleukin 6 (IL-6) and interleukin 8 (IL-8) proteins were measured in the culture medium and in the apical washes to determine the inflammatory response after exposure. RESULTS Both O3 and SynUrb54 significantly increased basolateral levels of LDH and IL-8 in A549 cells. No significant changes in LDH and IL-8 levels were observed in the EpiAirway™ cells, however, IL-6 in the apical surface was significantly elevated at 24 h after O3 exposure. CONCLUSION LDH and IL-8 are robust endpoints for assessing toxicity in A549 cells. The EpiAirway™ cells show minimal adverse effects after exposure suggesting that they are more toxicologically resistant compared to A549 cells. Higher concentrations or longer exposure times are needed to induce effects on EpiAirway™ cells.
Collapse
Affiliation(s)
- Jose Zavala
- a Department of Environmental Sciences & Engineering , University of North Carolina at Chapel Hill , Chapel Hill , NC, USA
| | - Bridget O'Brien
- a Department of Environmental Sciences & Engineering , University of North Carolina at Chapel Hill , Chapel Hill , NC, USA
| | - Kim Lichtveld
- a Department of Environmental Sciences & Engineering , University of North Carolina at Chapel Hill , Chapel Hill , NC, USA
| | - Kenneth G Sexton
- a Department of Environmental Sciences & Engineering , University of North Carolina at Chapel Hill , Chapel Hill , NC, USA
| | - Ivan Rusyn
- a Department of Environmental Sciences & Engineering , University of North Carolina at Chapel Hill , Chapel Hill , NC, USA
| | - Ilona Jaspers
- a Department of Environmental Sciences & Engineering , University of North Carolina at Chapel Hill , Chapel Hill , NC, USA .,b Department of Pediatrics , University of North Carolina at Chapel Hill , Chapel Hill , NC, USA , and.,c Center for Environmental Medicine and Lung Biology, University of North Carolina at Chapel Hill , Chapel Hill , NC, USA
| | - William Vizuete
- a Department of Environmental Sciences & Engineering , University of North Carolina at Chapel Hill , Chapel Hill , NC, USA
| |
Collapse
|
79
|
Shukla SD, Mahmood MQ, Weston S, Latham R, Muller HK, Sohal SS, Walters EH. The main rhinovirus respiratory tract adhesion site (ICAM-1) is upregulated in smokers and patients with chronic airflow limitation (CAL). Respir Res 2017; 18:6. [PMID: 28056984 PMCID: PMC5217320 DOI: 10.1186/s12931-016-0483-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 12/06/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND ICAM-1 is a major receptor for ~60% of human rhinoviruses, and non-typeable Haemophilus influenzae, two major pathogens in COPD. Increased cell-surface expression of ICAM-1 in response to tobacco smoke exposure has been suggested. We have investigated epithelial ICAM-1 expression in both the large and small airways, and lung parenchyma in smoking-related chronic airflow limitation (CAL) patients. METHODS We evaluated epithelial ICAM-1 expression in resected lung tissue: 8 smokers with normal spirometry (NLFS); 29 CAL patients (10 small-airway disease; 9 COPD-smokers; 10 COPD ex-smokers); Controls (NC): 15 normal airway/lung tissues. Immunostaining with anti-ICAM-1 monoclonal antibody was quantified with computerized image analysis. The percent and type of cells expressing ICAM-1 in large and small airway epithelium and parenchyma were enumerated, plus percentage of epithelial goblet and submucosal glands positive for ICAM- 1. RESULTS A major increase in ICAM-1 expression in epithelial cells was found in both large (p < 0.006) and small airways (p < 0.004) of CAL subjects compared to NC, with NLFS being intermediate. In the CAL group, both basal and luminal areas stained heavily for ICAM-1, so did goblet cells and sub-mucosal glands, however in either NC or NLFS subjects, only epithelial cell luminal surfaces stained. ICAM-1 expression on alveolar pneumocytes (mainly type II) was slightly increased in CAL and NLFS (p < 0.01). Pack-years of smoking correlated with ICAM-1 expression (r = 0.49; p < 0.03). CONCLUSION Airway ICAM-1 expression is markedly upregulated in CAL group, which could be crucial in rhinoviral and NTHi infections. The parenchymal ICAM-1 is affected by smoking, with no further enhancement in CAL subjects.
Collapse
Affiliation(s)
- Shakti Dhar Shukla
- NHMRC Centre of Research Excellence for Chronic Respiratory Disease, School of Medicine, University of Tasmania, MS1, 17 Liverpool Street, Private Bag 23, Hobart, Tasmania, 7000, Australia
| | - Malik Quasir Mahmood
- NHMRC Centre of Research Excellence for Chronic Respiratory Disease, School of Medicine, University of Tasmania, MS1, 17 Liverpool Street, Private Bag 23, Hobart, Tasmania, 7000, Australia
| | - Steven Weston
- NHMRC Centre of Research Excellence for Chronic Respiratory Disease, School of Medicine, University of Tasmania, MS1, 17 Liverpool Street, Private Bag 23, Hobart, Tasmania, 7000, Australia
| | - Roger Latham
- NHMRC Centre of Research Excellence for Chronic Respiratory Disease, School of Medicine, University of Tasmania, MS1, 17 Liverpool Street, Private Bag 23, Hobart, Tasmania, 7000, Australia
| | - Hans Konrad Muller
- NHMRC Centre of Research Excellence for Chronic Respiratory Disease, School of Medicine, University of Tasmania, MS1, 17 Liverpool Street, Private Bag 23, Hobart, Tasmania, 7000, Australia
| | - Sukhwinder Singh Sohal
- NHMRC Centre of Research Excellence for Chronic Respiratory Disease, School of Medicine, University of Tasmania, MS1, 17 Liverpool Street, Private Bag 23, Hobart, Tasmania, 7000, Australia.,School of Health Sciences, University of Tasmania, Launceston, Tasmania, 7248, Australia
| | - Eugene Haydn Walters
- NHMRC Centre of Research Excellence for Chronic Respiratory Disease, School of Medicine, University of Tasmania, MS1, 17 Liverpool Street, Private Bag 23, Hobart, Tasmania, 7000, Australia.
| |
Collapse
|
80
|
Faris AN, Ganesan S, Chattoraj A, Chattoraj SS, Comstock AT, Unger BL, Hershenson MB, Sajjan US. Rhinovirus Delays Cell Repolarization in a Model of Injured/Regenerating Human Airway Epithelium. Am J Respir Cell Mol Biol 2016; 55:487-499. [PMID: 27119973 DOI: 10.1165/rcmb.2015-0243oc] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Rhinovirus (RV), which causes exacerbation in patients with chronic airway diseases, readily infects injured airway epithelium and has been reported to delay wound closure. In this study, we examined the effects of RV on cell repolarization and differentiation in a model of injured/regenerating airway epithelium (polarized, undifferentiated cells). RV causes only a transient barrier disruption in a model of normal (mucociliary-differentiated) airway epithelium. However, in the injury/regeneration model, RV prolongs barrier dysfunction and alters the differentiation of cells. The prolonged barrier dysfunction caused by RV was not a result of excessive cell death but was instead associated with epithelial-to-mesenchymal transition (EMT)-like features, such as reduced expression of the apicolateral junction and polarity complex proteins, E-cadherin, occludin, ZO-1, claudins 1 and 4, and Crumbs3 and increased expression of vimentin, a mesenchymal cell marker. The expression of Snail, a transcriptional repressor of tight and adherence junctions, was also up-regulated in RV-infected injured/regenerating airway epithelium, and inhibition of Snail reversed RV-induced EMT-like features. In addition, compared with sham-infected cells, the RV-infected injured/regenerating airway epithelium showed more goblet cells and fewer ciliated cells. Inhibition of epithelial growth factor receptor promoted repolarization of cells by inhibiting Snail and enhancing expression of E-cadherin, occludin, and Crumbs3 proteins, reduced the number of goblet cells, and increased the number of ciliated cells. Together, these results suggest that RV not only disrupts barrier function, but also interferes with normal renewal of injured/regenerating airway epithelium by inducing EMT-like features and subsequent goblet cell hyperplasia.
Collapse
Affiliation(s)
- Andrea N Faris
- 1 Departments of Pediatrics and Communicable Diseases and
| | | | | | | | | | | | - Marc B Hershenson
- 1 Departments of Pediatrics and Communicable Diseases and.,2 Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | | |
Collapse
|
81
|
Abstract
Chronic airway diseases are a significant cause of morbidity and mortality worldwide, and their prevalence is predicted to increase in the future. Respiratory viruses are the most common cause of acute pulmonary infection, and there is clear evidence of their role in acute exacerbations of inflammatory airway diseases such as asthma and chronic obstructive pulmonary disease. Studies have reported impaired host responses to virus infection in these diseases, and a better understanding of the mechanisms of these abnormal immune responses has the potential to lead to the development of novel therapeutic targets for virus-induced exacerbations. The aim of this article is to review the current knowledge regarding the role of viruses and immune modulation in acute exacerbations of chronic pulmonary diseases and to discuss exciting areas for future research and novel treatments.
Collapse
|
82
|
Ganesan S, Pham D, Jing Y, Farazuddin M, Hudy MH, Unger B, Comstock AT, Proud D, Lauring AS, Sajjan US. TLR2 Activation Limits Rhinovirus-Stimulated CXCL-10 by Attenuating IRAK-1-Dependent IL-33 Receptor Signaling in Human Bronchial Epithelial Cells. THE JOURNAL OF IMMUNOLOGY 2016; 197:2409-20. [PMID: 27503209 DOI: 10.4049/jimmunol.1502702] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 07/07/2016] [Indexed: 12/15/2022]
Abstract
Airway epithelial cells are the major target for rhinovirus (RV) infection and express proinflammatory chemokines and antiviral cytokines that play a role in innate immunity. Previously, we demonstrated that RV interaction with TLR2 causes ILR-associated kinase-1 (IRAK-1) depletion in both airway epithelial cells and macrophages. Further, IRAK-1 degradation caused by TLR2 activation was shown to inhibit ssRNA-induced IFN expression in dendritic cells. Therefore, in this study, we examined the role of TLR2 and IRAK-1 in RV-induced IFN-β, IFN-λ1, and CXCL-10, which require signaling by viral RNA. In airway epithelial cells, blocking TLR2 enhanced RV-induced expression of IFNs and CXCL-10. By contrast, IRAK-1 inhibition abrogated RV-induced expression of CXCL-10, but not IFNs in these cells. Neutralization of IL-33 or its receptor, ST2, which requires IRAK-1 for signaling, inhibited RV-stimulated CXCL-10 expression. In addition, RV induced expression of both ST2 and IL-33 in airway epithelial cells. In macrophages, however, RV-stimulated CXCL-10 expression was primarily dependent on TLR2/IL-1R. Interestingly, in a mouse model of RV infection, blocking ST2 not only attenuated RV-induced CXCL-10, but also lung inflammation. Finally, influenza- and respiratory syncytial virus-induced CXCL-10 was also found to be partially dependent on IL-33/ST2/IRAK-1 signaling in airway epithelial cells. Together, our results indicate that RV stimulates CXCL-10 expression via the IL-33/ST2 signaling axis, and that TLR2 signaling limits RV-induced CXCL-10 via IRAK-1 depletion at least in airway epithelial cells. To our knowledge, this is the first report to demonstrate the role of respiratory virus-induced IL-33 in the induction of CXCL-10 in airway epithelial cells.
Collapse
Affiliation(s)
- Shyamala Ganesan
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109
| | - Duc Pham
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109
| | - Yaxun Jing
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109
| | - Mohammad Farazuddin
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109
| | - Magdalena H Hudy
- Department of Physiology & Pharmacology, University of Calgary Faculty of Medicine, Calgary, Alberta T2N 4N1, Canada
| | - Benjamin Unger
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109
| | - Adam T Comstock
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109
| | - David Proud
- Department of Physiology & Pharmacology, University of Calgary Faculty of Medicine, Calgary, Alberta T2N 4N1, Canada
| | - Adam S Lauring
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109; and Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109
| | - Uma S Sajjan
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109;
| |
Collapse
|
83
|
Jamieson KC, Warner SM, Leigh R, Proud D. Rhinovirus in the Pathogenesis and Clinical Course of Asthma. Chest 2016; 148:1508-1516. [PMID: 26270739 DOI: 10.1378/chest.15-1335] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
In healthy individuals, human rhinovirus (HRV) infections are the major cause of the common cold. These are generally uncomplicated infections except for occasional cases of otitis media or sinusitis. In individuals with asthma, however, HRV infections can have a major impact on disease development and progression. HRV-induced wheezing illnesses in early life are a significant risk factor for subsequent development of asthma, and growing evidence supports a role of recurrent HRV infections in the development and progression of several aspects of airway remodeling in asthma. In addition, HRV infections are one of the most common triggers for acute exacerbations of asthma, which represent a major burden to health-care systems around the world. None of the currently prescribed medications for asthma are effective in preventing or reversing asthma development and airway remodeling or are ideal for treating HRV-induced exacerbations of asthma. Thus, a better understanding of the role of HRV in asthma is important if we are to develop more effective therapies. In the past decade, we have gained new insights into the role of HRV infections in the development and progression of airway remodeling as well as a new appreciation for the proinflammatory and host defense responses to HRV infections that may help to regulate susceptibility to asthma exacerbations. This article reviews the current understanding of the role HRV infections play in the pathogenesis of asthma and identifies possible avenues to new therapeutic strategies for limiting the effects of HRV infections in asthma.
Collapse
Affiliation(s)
- Kyla C Jamieson
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Stephanie M Warner
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Richard Leigh
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - David Proud
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
84
|
Yamaya M, Nomura K, Arakawa K, Nishimura H, Lusamba Kalonji N, Kubo H, Nagatomi R, Kawase T. Increased rhinovirus replication in nasal mucosa cells in allergic subjects is associated with increased ICAM-1 levels and endosomal acidification and is inhibited by L-carbocisteine. IMMUNITY INFLAMMATION AND DISEASE 2016; 4:166-181. [PMID: 27957326 PMCID: PMC4879463 DOI: 10.1002/iid3.102] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 02/09/2016] [Accepted: 02/10/2016] [Indexed: 01/22/2023]
Abstract
Increased viral replication and cytokine production may be associated with the pathogenesis of asthma attacks in rhinovirus (RV) infections. However, the association between increased RV replication and enhanced expression of intercellular adhesion molecule‐1 (ICAM‐1), a receptor for a major RV group, in airway epithelial cells has remained unclear. Furthermore, the inhibitory effects of mucolytics, which have clinical benefits in asthmatic subjects, are uncertain. Human nasal epithelial (HNE) cells were infected with type 14 rhinovirus (RV14), a major RV group. RV14 titers and cytokine concentrations, including interleukin (IL)‐6 and IL‐8, in supernatants, RV14 RNA replication and susceptibility to RV14 infection were higher in HNE cells obtained from subjects in the allergic group (allergic subjects) than in those from subjects in the non‐allergic group (non‐allergic subjects). ICAM‐1 expression and the number and fluorescence intensity of acidic endosomes from which RV14 RNA enters the cytoplasm were higher in HNE cells from allergic subjects, though substantial amounts of interferon (IFN)‐γ and IFN‐λ were not detected in the supernatant. The abundance of p50 and p65 subunits of transcription factor nuclear factor kappa B (NF‐κB) in nuclear extracts of the cells from allergic subjects was higher compared to non‐allergic subjects, and an inhibitor of NF‐κB, caffeic acid phenethyl ester, reduced the fluorescence intensity of acidic endosomes as well as RV titers and RNA. Furthermore, a mucolytic agent, L‐carbocisteine, reduced RV14 titers and RNA levels, cytokine release, ICAM‐1 expression, the fluorescence intensity of acidic endosomes, and NF‐κB activation. The increased RV14 replication observed in HNE cells from allergic subjects might be partly associated with enhanced ICAM‐1 expression and decreased endosomal pH through NF‐κB activation. L‐Carbocisteine inhibits RV14 infection by reducing ICAM‐1 and acidic endosomes and may, therefore, modulate airway inflammation caused by RV infection in allergic subjects.
Collapse
Affiliation(s)
- Mutsuo Yamaya
- Department of Advanced Preventive Medicine for Infectious Disease Tohoku University Graduate School of Medicine Sendai 980-8575 Japan
| | - Kazuhiro Nomura
- Department of Otolaryngology-Head and Neck Surgery Tohoku University Graduate School of Medicine Sendai 980-8575 Japan
| | - Kazuya Arakawa
- Department of Otolaryngology-Head and Neck Surgery Tohoku University Graduate School of Medicine Sendai 980-8575 Japan
| | - Hidekazu Nishimura
- Virus Research Center, Clinical Research Division Sendai Medical Center Sendai 983-8520 Japan
| | - Nadine Lusamba Kalonji
- Department of Advanced Preventive Medicine for Infectious Disease Tohoku University Graduate School of Medicine Sendai 980-8575 Japan
| | - Hiroshi Kubo
- Department of Advanced Preventive Medicine for Infectious Disease Tohoku University Graduate School of Medicine Sendai 980-8575 Japan
| | - Ryoichi Nagatomi
- Medicine and Science in Sports and Exercise Tohoku University Graduate School of Medicine Sendai 980-8575 Japan
| | - Tetsuaki Kawase
- Laboratory of Rehabilitative Auditory Science Tohoku University Graduate School of Biomedical Engineering Sendai 980-8575 Japan
| |
Collapse
|
85
|
Hewitt R, Farne H, Ritchie A, Luke E, Johnston SL, Mallia P. The role of viral infections in exacerbations of chronic obstructive pulmonary disease and asthma. Ther Adv Respir Dis 2016; 10:158-74. [PMID: 26611907 PMCID: PMC5933560 DOI: 10.1177/1753465815618113] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) are major causes of global morbidity and mortality worldwide. The clinical course of both asthma and COPD are punctuated by the occurrence of exacerbations, acute events characterized by increased symptoms and airflow obstruction. Exacerbations contribute most of the morbidity, mortality and excess healthcare costs associated with both asthma and COPD. COPD and asthma exacerbations are frequently associated with respiratory virus infections and this has led to an intense research focus into the mechanisms of virus-induced exacerbations over the past decade. Current therapies are effective in reducing chronic symptoms but are less effective in preventing exacerbations, particularly in COPD. Understanding the mechanisms of virus-induced exacerbation will lead to the development of new targeted therapies that can reduce the burden of virus-induced exacerbations. In this review we discuss current knowledge of virus-induced exacerbations of asthma and COPD with a particular focus on mechanisms, human studies, virus-bacteria interactions and therapeutic advances.
Collapse
Affiliation(s)
- Richard Hewitt
- National Heart and Lung Institute, Imperial College London, UK
| | - Hugo Farne
- National Heart and Lung Institute, Imperial College London, UK
| | - Andrew Ritchie
- National Heart and Lung Institute, Imperial College London, UK
| | - Emma Luke
- Imperial Healthcare NHS Trust, London, UK
| | | | - Patrick Mallia
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
86
|
Wang Q, Chen D, Xie H, Lin X, Wang X, Yao Q, Zheng X, Xu C, Chen L, He S, Zhang H. Altered Expression of IFN-λ2 in Allergic Airway Disorders and Identification of Its Cell Origins. Mediators Inflamm 2016; 2016:5759496. [PMID: 27057098 PMCID: PMC4737047 DOI: 10.1155/2016/5759496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 11/06/2015] [Accepted: 11/22/2015] [Indexed: 02/05/2023] Open
Abstract
This study investigated the expression levels of interferon- (IFN-) λ2 in peripheral blood and tissues. The results showed that the levels of IFN-λ2 were elevated by 17.9% and 14.2% in the plasma of allergic rhinitis (AR) and combined rhinitis with asthma (AR + AS), which was positively correlated with the level of tryptase but negatively correlated with the level of IL-10. IFN-λ2 was predominately expressed in the CD16+ cells and CD14+ cells in healthy control subjects (HC) but upregulated only in CD8+ cells of AR and in eosinophils of asthma. It was observed that approximately 6.6% and 7.0% dispersed tonsil cells and 5.8% and 0.44% dispersed lung cells are IFN-λ2+ mast cells and macrophages. Moreover, tryptase and agonist peptides of PAR-2 induced enhanced IFN-λ2 mRNA expression in A549 cells. In conclusion, the elevated levels of IFN-λ2 in the plasma of AR and AR + AS indicate that IFN-λ2 is likely to contribute to the pathogenesis of allergic airway disorders. The potential origins of the elevated plasma IFN-λ2 include mast cells, macrophages, and epithelial cells in tissues, neutrophils, monocytes, CD8+ T cells, and eosinophils in peripheral blood. Development of IFN-λ2 related therapy may help to treat or prevent allergic airway disorders.
Collapse
Affiliation(s)
- Qiuli Wang
- Department of ENT, Allergy and Clinical Immunology Research Centre, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning 121001, China
| | - Dong Chen
- Department of ENT, Allergy and Clinical Immunology Research Centre, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning 121001, China
| | - Hua Xie
- Department of Respiratory Medicine, The General Hospital of Shenyang Military Region, Shenyang, Liaoning 110016, China
| | - Xiaoping Lin
- Department of Respiratory Medicine, The General Hospital of Shenyang Military Region, Shenyang, Liaoning 110016, China
| | - Xuefeng Wang
- Department of ENT, Allergy and Clinical Immunology Research Centre, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning 121001, China
| | - Qijian Yao
- Allergy and Inflammation Research Institute, Shantou University Medical College, Shantou 515031, China
| | - Xiaoxuan Zheng
- Allergy and Inflammation Research Institute, Shantou University Medical College, Shantou 515031, China
| | - Chiyan Xu
- Allergy and Inflammation Research Institute, Shantou University Medical College, Shantou 515031, China
| | - Lingfei Chen
- Allergy and Inflammation Research Institute, Shantou University Medical College, Shantou 515031, China
| | - Shaoheng He
- Department of ENT, Allergy and Clinical Immunology Research Centre, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning 121001, China
| | - Huiyun Zhang
- Department of ENT, Allergy and Clinical Immunology Research Centre, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning 121001, China
| |
Collapse
|
87
|
Zhang Q, Wan H, Huang S, Zhang Y, Wang Y, Guo X, He P, Zhou M. Critical role of RIG-I-like receptors in inflammation in chronic obstructive pulmonary disease. THE CLINICAL RESPIRATORY JOURNAL 2016; 10:22-31. [PMID: 24992168 PMCID: PMC7162323 DOI: 10.1111/crj.12177] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 06/10/2014] [Accepted: 06/22/2014] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Viral infection is a significant cause of chronic obstructive pulmonary disease (COPD) and acute exacerbation of COPD. Retinoic acid inducible gene I (RIG-I)-like receptors (RLRs), including RIG-I and melanoma differentiation associated gene 5 (MDA-5), are important pattern recognition receptors for viral elimination. OBJECTIVE The study aims to investigate the role of RIG-I and MDA-5 in COPD pathogenesis. METHODS We examined the expression of RIG-I and MDA-5 by immunohistochemistry, real-time PCR and Western blots in COPD patients and control subjects. RESULTS Our results showed that MDA-5 expression was upregulated in lung tissues and peripheral blood mononuclear cells of COPD patients and there was a negative correlation between MDA-5 mRNA levels and forced expiratory volume in 1 s %pred. COPD patients had higher interleukin (IL)-1 and IL-8 mRNA expression levels, and these inflammatory cytokines positively correlate with MDA-5 levels. However, there was no difference in the expression of RIG-I between COPD patients and control subjects. CONCLUSION Our results suggested that MDA-5, but not RIG-I, may play a critical role in airway inflammation in COPD.
Collapse
Affiliation(s)
- Qiurui Zhang
- Department of Respiratory MedicineRuijin HospitalShanghai Jiaotong University School of MedicineShanghaiChina
| | - Huanying Wan
- Department of Respiratory MedicineRuijin HospitalShanghai Jiaotong University School of MedicineShanghaiChina
| | - Shaoguang Huang
- Department of Respiratory MedicineRuijin HospitalShanghai Jiaotong University School of MedicineShanghaiChina
| | - Yan Zhang
- Department of Microbiology and ParasitologyInstitutes of Medical SciencesShanghai Jiaotong University School of MedicineShanghaiChina
| | - Yanchun Wang
- Department of Microbiology and ParasitologyInstitutes of Medical SciencesShanghai Jiaotong University School of MedicineShanghaiChina
| | - Xiaokui Guo
- Department of Microbiology and ParasitologyInstitutes of Medical SciencesShanghai Jiaotong University School of MedicineShanghaiChina
| | - Ping He
- Department of Microbiology and ParasitologyInstitutes of Medical SciencesShanghai Jiaotong University School of MedicineShanghaiChina
| | - Min Zhou
- Department of Respiratory MedicineRuijin HospitalShanghai Jiaotong University School of MedicineShanghaiChina
| |
Collapse
|
88
|
Rowell TR, Tarran R. Will chronic e-cigarette use cause lung disease? Am J Physiol Lung Cell Mol Physiol 2015; 309:L1398-409. [PMID: 26408554 PMCID: PMC4683316 DOI: 10.1152/ajplung.00272.2015] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 09/22/2015] [Indexed: 12/22/2022] Open
Abstract
Chronic tobacco smoking is a major cause of preventable morbidity and mortality worldwide. In the lung, tobacco smoking increases the risk of lung cancer, and also causes chronic obstructive pulmonary disease (COPD), which encompasses both emphysema and chronic bronchitis. E-cigarettes (E-Cigs), or electronic nicotine delivery systems, were developed over a decade ago and are designed to deliver nicotine without combusting tobacco. Although tobacco smoking has declined since the 1950s, E-Cig usage has increased, attracting both former tobacco smokers and never smokers. E-Cig liquids (e-liquids) contain nicotine in a glycerol/propylene glycol vehicle with flavorings, which are vaporized and inhaled. To date, neither E-Cig devices, nor e-liquids, are regulated by the Food and Drug Administration (FDA). The FDA has proposed a deeming rule, which aims to initiate legislation to regulate E-Cigs, but the timeline to take effect is uncertain. Proponents of E-Cigs say that they are safe and should not be regulated. Opposition is varied, with some opponents proposing that E-Cig usage will introduce a new generation to nicotine addiction, reversing the decline seen with tobacco smoking, or that E-Cigs generally may not be safe and will trigger diseases like tobacco. In this review, we shall discuss what is known about the effects of E-Cigs on the mammalian lung and isolated lung cells in vitro. We hope that collating this data will help illustrate gaps in the knowledge of this burgeoning field, directing researchers toward answering whether or not E-Cigs are capable of causing disease.
Collapse
Affiliation(s)
- Temperance R Rowell
- Marsico Lung Institute and Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Robert Tarran
- Marsico Lung Institute and Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
89
|
Alves MP, Schögler A, Ebener S, Vielle NJ, Casaulta C, Jung A, Moeller A, Geiser T, Regamey N. Comparison of innate immune responses towards rhinovirus infection of primary nasal and bronchial epithelial cells. Respirology 2015; 21:304-12. [DOI: 10.1111/resp.12692] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 07/14/2015] [Accepted: 08/26/2015] [Indexed: 12/27/2022]
Affiliation(s)
- Marco P. Alves
- Department of Clinical ResearchUniversity Hospital of Bern Bern Switzerland
- Division of Pediatric Respiratory MedicineUniversity Children's Hospital Bern Switzerland
| | - Aline Schögler
- Department of Clinical ResearchUniversity Hospital of Bern Bern Switzerland
- Graduate School for Cellular and Biomedical SciencesUniversity Hospital of Bern Bern Switzerland
- Division of Pediatric Respiratory MedicineUniversity Children's Hospital Bern Switzerland
| | - Simone Ebener
- Department of Clinical ResearchUniversity Hospital of Bern Bern Switzerland
- Graduate School for Cellular and Biomedical SciencesUniversity Hospital of Bern Bern Switzerland
- Division of Pediatric Respiratory MedicineUniversity Children's Hospital Bern Switzerland
| | - Nathalie J. Vielle
- Department of Clinical ResearchUniversity Hospital of Bern Bern Switzerland
- Division of Pediatric Respiratory MedicineUniversity Children's Hospital Bern Switzerland
| | - Carmen Casaulta
- Division of Pediatric Respiratory MedicineUniversity Children's Hospital Bern Switzerland
| | - Andreas Jung
- Division of Respiratory MedicineUniversity Children's Hospital Zürich Switzerland
| | - Alexander Moeller
- Division of Respiratory MedicineUniversity Children's Hospital Zürich Switzerland
| | - Thomas Geiser
- Department of Clinical ResearchUniversity Hospital of Bern Bern Switzerland
- Division of Respiratory MedicineUniversity Hospital of Bern Bern Switzerland
| | - Nicolas Regamey
- Department of Clinical ResearchUniversity Hospital of Bern Bern Switzerland
- Children's Hospital Lucerne Lucerne Switzerland
| |
Collapse
|
90
|
Interferon response of the cystic fibrosis bronchial epithelium to major and minor group rhinovirus infection. J Cyst Fibros 2015; 15:332-9. [PMID: 26613982 PMCID: PMC7185532 DOI: 10.1016/j.jcf.2015.10.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 10/19/2015] [Accepted: 10/29/2015] [Indexed: 12/01/2022]
Abstract
Rhinoviruses (RVs) are associated with exacerbations of cystic fibrosis (CF), asthma and COPD. There is growing evidence suggesting the involvement of the interferon (IFN) pathway in RV-associated morbidity in asthma and COPD. The mechanisms of RV-triggered exacerbations in CF are poorly understood. In a pilot study, we assessed the antiviral response of CF and healthy bronchial epithelial cells (BECs) to RV infection, we measured the levels of IFNs, pattern recognition receptors (PRRs) and IFN-stimulated genes (ISGs) upon infection with major and minor group RVs and poly(IC) stimulation. Major group RV infection of CF BECs resulted in a trend towards a diminished IFN response at the level of IFNs, PRRs and ISGs in comparison to healthy BECs. Contrary to major group RV, the IFN pathway induction upon minor group RV infection was significantly increased at the level of IFNs and PRRs in CF BECs compared to healthy BECs.
Collapse
|
91
|
A short-term mouse model that reproduces the immunopathological features of rhinovirus-induced exacerbation of COPD. Clin Sci (Lond) 2015; 129:245-58. [PMID: 25783022 PMCID: PMC4557402 DOI: 10.1042/cs20140654] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Viral exacerbations of chronic obstructive pulmonary disease (COPD), commonly caused by rhinovirus (RV) infections, are poorly controlled by current therapies. This is due to a lack of understanding of the underlying immunopathological mechanisms. Human studies have identified a number of key immune responses that are associated with RV-induced exacerbations including neutrophilic inflammation, expression of inflammatory cytokines and deficiencies in innate anti-viral interferon. Animal models of COPD exacerbation are required to determine the contribution of these responses to disease pathogenesis. We aimed to develop a short-term mouse model that reproduced the hallmark features of RV-induced exacerbation of COPD. Evaluation of complex protocols involving multiple dose elastase and lipopolysaccharide (LPS) administration combined with RV1B infection showed suppression rather than enhancement of inflammatory parameters compared with control mice infected with RV1B alone. Therefore, these approaches did not accurately model the enhanced inflammation associated with RV infection in patients with COPD compared with healthy subjects. In contrast, a single elastase treatment followed by RV infection led to heightened airway neutrophilic and lymphocytic inflammation, increased expression of tumour necrosis factor (TNF)-α, C-X-C motif chemokine 10 (CXCL10)/IP-10 (interferon γ-induced protein 10) and CCL5 [chemokine (C-C motif) ligand 5]/RANTES (regulated on activation, normal T-cell expressed and secreted), mucus hypersecretion and preliminary evidence for increased airway hyper-responsiveness compared with mice treated with elastase or RV infection alone. In summary, we have developed a new mouse model of RV-induced COPD exacerbation that mimics many of the inflammatory features of human disease. This model, in conjunction with human models of disease, will provide an essential tool for studying disease mechanisms and allow testing of novel therapies with potential to be translated into clinical practice.
Collapse
|
92
|
Solleti SK, Simon DM, Srisuma S, Arikan MC, Bhattacharya S, Rangasamy T, Bijli KM, Rahman A, Crossno JT, Shapiro SD, Mariani TJ. Airway epithelial cell PPARγ modulates cigarette smoke-induced chemokine expression and emphysema susceptibility in mice. Am J Physiol Lung Cell Mol Physiol 2015; 309:L293-304. [PMID: 26024894 DOI: 10.1152/ajplung.00287.2014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 05/26/2015] [Indexed: 11/22/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a highly prevalent, chronic inflammatory lung disease with limited existing therapeutic options. While modulation of peroxisome proliferator-activating receptor (PPAR)-γ activity can modify inflammatory responses in several models of lung injury, the relevance of the PPARG pathway in COPD pathogenesis has not been previously explored. Mice lacking Pparg specifically in airway epithelial cells displayed increased susceptibility to chronic cigarette smoke (CS)-induced emphysema, with excessive macrophage accumulation associated with increased expression of chemokines, Ccl5, Cxcl10, and Cxcl15. Conversely, treatment of mice with a pharmacological PPARγ activator attenuated Cxcl10 and Cxcl15 expression and macrophage accumulation in response to CS. In vitro, CS increased lung epithelial cell chemokine expression in a PPARγ activation-dependent fashion. The ability of PPARγ to regulate CS-induced chemokine expression in vitro was not specifically associated with peroxisome proliferator response element (PPRE)-mediated transactivation activity but was correlated with PPARγ-mediated transrepression of NF-κB activity. Pharmacological or genetic activation of PPARγ activity abrogated CS-dependent induction of NF-κB activity. Regulation of NF-κB activity involved direct PPARγ-NF-κB interaction and PPARγ-mediated effects on IKK activation, IκBα degradation, and nuclear translocation of p65. Our data indicate that PPARG represents a disease-relevant pathophysiological and pharmacological target in COPD. Its activation state likely contributes to NF-κB-dependent, CS-induced chemokine-mediated regulation of inflammatory cell accumulation.
Collapse
Affiliation(s)
- Siva Kumar Solleti
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York
| | - Dawn M Simon
- Emory-Children's Center Pulmonary, Apnea, Cystic Fibrosis and Sleep Clinic, Atlanta, Georgia
| | - Sorachai Srisuma
- Faculty of Medicine, Department of Physiology, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Meltem C Arikan
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Soumyaroop Bhattacharya
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York;
| | - Tirumalai Rangasamy
- Division of Pulmonary & Critical Care Medicine, University of Rochester Medical Center, Rochester, New York
| | - Kaiser M Bijli
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York; Atlanta VA and Division of Pulmonary, Allergy, and Critical Care Medicine, Emory University, Atlanta, Georgia
| | - Arshad Rahman
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York
| | - Joseph T Crossno
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Health Sciences Center, Denver, Colorado
| | - Steven D Shapiro
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Thomas J Mariani
- Division of Neonatology and Pediatric Molecular and Personalized Medicine Program, University of Rochester Medical Center, Rochester, New York;
| |
Collapse
|
93
|
Leigh R, Proud D. Virus-induced modulation of lower airway diseases: pathogenesis and pharmacologic approaches to treatment. Pharmacol Ther 2014; 148:185-98. [PMID: 25550230 PMCID: PMC7173263 DOI: 10.1016/j.pharmthera.2014.12.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 12/24/2014] [Indexed: 02/08/2023]
Abstract
Uncomplicated upper respiratory viral infections are the most common cause of days lost from work and school and exert a major economic burden. In susceptible individuals, however, common respiratory viruses, particularly human rhinoviruses, also can have a major impact on diseases that involve the lower airways, including asthma, chronic obstructive pulmonary diseases (COPD) and cystic fibrosis (CF). Respiratory virus-induced wheezing illnesses in early life are a significant risk factor for the subsequent development of asthma, and virus infections may also play a role in the development and progression of airway remodeling in asthma. It is clear that upper respiratory tract virus infections can spread to the lower airway and trigger acute attacks of asthma, COPD or CF. These exacerbations can be life-threatening, and exert an enormous burden on health care systems. In recent years we have gained new insights into the mechanisms by which respiratory viruses may induce acute exacerbations of lower airway diseases, as well as into host defense pathways that may regulate the outcomes to viral infections. In the current article we review the role of viruses in lower airway diseases, including our current understanding on pathways by which they may cause remodeling and trigger acute exacerbations. We also review the efficacy of current and emerging therapies used to treat these lower airway diseases on the outcomes due to viral infection, and discuss alternative therapeutic approaches for the management of virus-induced airway inflammation.
Collapse
Affiliation(s)
- Richard Leigh
- Airway Inflammation Research Group, Snyder Institute for Chronic Diseases and Department of Medicine, University of Calgary Faculty of Medicine, Calgary, Canada; Airway Inflammation Research Group, Snyder Institute for Chronic Diseases and Department of Physiology & Pharmacology, University of Calgary Faculty of Medicine, Calgary, Canada
| | - David Proud
- Airway Inflammation Research Group, Snyder Institute for Chronic Diseases and Department of Physiology & Pharmacology, University of Calgary Faculty of Medicine, Calgary, Canada.
| |
Collapse
|
94
|
Holtzman MJ, Byers DE, Alexander-Brett J, Wang X. The role of airway epithelial cells and innate immune cells in chronic respiratory disease. Nat Rev Immunol 2014; 14:686-98. [PMID: 25234144 PMCID: PMC4782595 DOI: 10.1038/nri3739] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
An abnormal immune response to environmental agents is generally thought to be responsible for causing chronic respiratory diseases, such as asthma and chronic obstructive pulmonary disease (COPD). Based on studies of experimental models and human subjects, there is increasing evidence that the response of the innate immune system is crucial for the development of this type of airway disease. Airway epithelial cells and innate immune cells represent key components of the pathogenesis of chronic airway disease and are emerging targets for new therapies. In this Review, we summarize the innate immune mechanisms by which airway epithelial cells and innate immune cells regulate the development of chronic respiratory diseases. We also explain how these pathways are being targeted in the clinic to treat patients with these diseases.
Collapse
Affiliation(s)
- Michael J Holtzman
- 1] Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63110, USA. [2] Department of Cell Biology, Washington University School of Medicine, Saint Louis, Missouri 63110, USA
| | - Derek E Byers
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63110, USA
| | - Jennifer Alexander-Brett
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63110, USA
| | - Xinyu Wang
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63110, USA
| |
Collapse
|
95
|
Kapur N, Mackay IM, Sloots TP, Masters IB, Chang AB. Respiratory viruses in exacerbations of non-cystic fibrosis bronchiectasis in children. Arch Dis Child 2014; 99:749-53. [PMID: 24819370 DOI: 10.1136/archdischild-2013-305147] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Respiratory viral infections precipitate exacerbations of chronic respiratory diseases such as asthma and chronic obstructive pulmonary disease though similar data in non-cystic fibrosis (CF) bronchiectasis are missing. Our study aimed to determine the point prevalence of viruses associated with exacerbations and evaluate clinical and investigational differences between virus-positive and -negative exacerbations in children with bronchiectasis. METHODS A cohort of 69 children (median age 7 years) with non-CF bronchiectasis was prospectively followed for 900 child-months. PCR for 16 respiratory viruses was performed on nasopharyngeal aspirates collected during 77 paediatric pulmonologist-defined exacerbations. Clinical data, systemic (C reactive protein (CRP), IL-6, procalcitonin, amyloid-A, fibrinogen) and lung function parameters were also collected. FINDINGS Respiratory viruses were detected during 37 (48%) exacerbations: human rhinovirus (HRV) in 20; an enterovirus or bocavirus in four each; adenoviruses, metapneumovirus, influenza A virus, respiratory syncytial virus, parainfluenza virus 3 or 4 in two each; coronavirus or parainfluenza virus 1 and 2 in one each. Viral codetections occurred in 6 (8%) exacerbations. HRV-As (n=9) were more likely to be present than HRV-Cs (n=2). Children with virus-positive exacerbations were more likely to require hospitalisation (59% vs 32.5% (p=0.02)) and have fever (OR 3.1, 95% CI 1.2 to 11.1), hypoxia (OR 25.5, 95% CI 2.0 to 322.6), chest signs (OR 3.3, 95% CI 1.1 to 10.2) and raised CRP (OR 4.7, 95% CI 1.7 to 13.1) when compared with virus-negative exacerbations. INTERPRETATION Respiratory viruses are commonly detected during pulmonary exacerbations of children with bronchiectasis. HRV-As were the most frequently detected viruses with viral codetection being rare. Time-sequenced cohort studies are needed to determine the role of viral-bacterial interactions in exacerbations of bronchiectasis.
Collapse
Affiliation(s)
- Nitin Kapur
- Department of Respiratory Medicine, Royal Children's Hospital, Queensland, Australia
| | - Ian M Mackay
- Queensland Paediatric Infectious Diseases Laboratory, SASVRC, QCMRI, Herston, Queensland, Australia
| | - Theo P Sloots
- Queensland Paediatric Infectious Diseases Laboratory, SASVRC, QCMRI, Herston, Queensland, Australia
| | - Ian B Masters
- Department of Respiratory Medicine, Royal Children's Hospital, Queensland, Australia
| | - Anne B Chang
- Department of Respiratory Medicine, Royal Children's Hospital, Queensland, Australia Child Health Division, Menzies School of Health Research, Darwin, Australia
| |
Collapse
|
96
|
Pu X, Wang L, Chang JY, Hildebrandt MAT, Ye Y, Lu C, Skinner HD, Niu N, Jenkins GD, Komaki R, Minna JD, Roth JA, Weinshilboum RM, Wu X. Inflammation-related genetic variants predict toxicity following definitive radiotherapy for lung cancer. Clin Pharmacol Ther 2014; 96:609-15. [PMID: 25054431 PMCID: PMC4206576 DOI: 10.1038/clpt.2014.154] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 07/16/2014] [Indexed: 12/25/2022]
Abstract
Definitive radiotherapy improves locoregional control and survival in inoperable non-small cell lung cancer (NSCLC) patients. However, radiation-induced toxicities (pneumonitis/esophagitis) are common dose-limiting inflammatory conditions. We therefore conducted a pathway-based analysis to identify inflammation-related SNPs associated with radiation-induced pneumonitis or esophagitis. 11,930 SNPs were genotyped in 201 stage I-III NSCLC patients treated with definitive radiotherapy. Validation was performed in an additional 220 NSCLC cases. After validation, 19 SNPs remained significant. A polygenic risk score (PRS) was generated to summarize the effect from validated SNPs. Significant improvements in discriminative ability were observed by adding the PRS into the clinical/epidemiological variable-based model. We then used 277 lymphoblastoid cell-lines to assess radiation sensitivity and eQTL relationships of the identified SNPs. Three genes (PRKCE,DDX58 and TNFSF7) were associated with radiation sensitivity. We concluded that inflammation-related genetic variants could contribute to the development of radiation-induced toxicities. These loci could assist in predicting those unfavorable events.
Collapse
Affiliation(s)
- X Pu
- Department of Epidemiology, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - L Wang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - J Y Chang
- Department of Radiation Oncology, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - M A T Hildebrandt
- Department of Epidemiology, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Y Ye
- Department of Epidemiology, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - C Lu
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - H D Skinner
- Department of Radiation Oncology, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - N Niu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - G D Jenkins
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - R Komaki
- Department of Radiation Oncology, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - J D Minna
- Hamon Center for Therapeutic Oncology Research, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - J A Roth
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - R M Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - X Wu
- Department of Epidemiology, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
97
|
Yamaya M, Nishimura H, Nadine L, Kubo H, Nagatomi R. Formoterol and budesonide inhibit rhinovirus infection and cytokine production in primary cultures of human tracheal epithelial cells. Respir Investig 2014; 52:251-60. [PMID: 24998372 DOI: 10.1016/j.resinv.2014.03.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 02/17/2014] [Accepted: 03/10/2014] [Indexed: 11/25/2022]
Abstract
BACKGROUND Long-acting β(2) agonists (LABAs) and inhaled corticosteroids (ICSs) reduce the frequency of exacerbations of chronic obstructive pulmonary disease and bronchial asthma. However, inhibitory effects of LABAs and ICSs on the replication of rhinovirus (RV), the major cause of exacerbations, have not been demonstrated. METHODS Primary cultures of human tracheal epithelial cells were infected with a major group RV, type 14 rhinovirus (RV14), to examine the effects of formoterol and budesonide on RV infection and infection-induced airway inflammation. RESULTS Treatment with formoterol and budesonide 72 h before and after RV14 infection reduced RV14 titers and cytokine concentrations, including interleukin (IL)-1β, IL-6 and IL-8, in supernatants and viral RNA within cells. Formoterol and budesonide reduced mRNA expression and protein concentration of intercellular adhesion molecule-1 (ICAM-1), the receptor for RV14. Formoterol reduced the number and fluorescence intensity of acidic endosomes through which RV RNA enters the cytoplasm. Formoterol and budesonide reduced the activation of the nuclear factor kappa-B protein p65 in nuclear extracts. The effects of formoterol plus budesonide were additive with respect to RV14 replication, cytokine production, ICAM-1 expression, acidic endosome fluorescence intensity, and p65 activation. The selective β(2)-adrenergic receptor antagonist, ICI 118551 [erythro-dl-1-(7-methylindan-4-yloxy)-3-isopropylaminobutan-2-ol], reversed the inhibitory effects of formoterol on RV14 titers and RNA levels, the susceptibility of cells to RV14 infection, cytokine production, acidic endosomes, ICAM-1 expression, and p65 activation. CONCLUSIONS Formoterol and budesonide may inhibit RV infection by reducing the ICAM-1 levels and/or acidic endosomes and modulate airway inflammation associated with RV infections.
Collapse
Affiliation(s)
- Mutsuo Yamaya
- Department of Advanced Preventive Medicine for Infectious Disease, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| | - Hidekazu Nishimura
- Virus Research Center, Clinical Research Division, Sendai National Hospital, Sendai, Japan.
| | - Lusamba Nadine
- Department of Advanced Preventive Medicine for Infectious Disease, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| | - Hiroshi Kubo
- Department of Advanced Preventive Medicine for Infectious Disease, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
| | - Ryoichi Nagatomi
- Medicine and Science in Sports and Exercise, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
98
|
Ramirez IA, Caverly LL, Kalikin LM, Goldsmith AM, Lewis TC, Burke DT, LiPuma JJ, Sajjan US, Hershenson MB. Differential responses to rhinovirus- and influenza-associated pulmonary exacerbations in patients with cystic fibrosis. Ann Am Thorac Soc 2014; 11:554-61. [PMID: 24641803 PMCID: PMC4225796 DOI: 10.1513/annalsats.201310-346oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 01/26/2014] [Indexed: 12/25/2022] Open
Abstract
RATIONALE The mechanism by which viruses cause exacerbations of chronic airway disease and the capacity of patients with cystic fibrosis (CF) to respond to viral infection are not precisely known. OBJECTIVES To determine the antiviral response to infection in patients with CF. METHODS Sputum was collected from patients with CF with respiratory exacerbation. Viruses were detected in multiplex polymerase chain reaction (PCR)-based assays. Gene expression of 84 antiviral response genes was measured, using a focused quantitative PCR gene array. MEASUREMENTS AND MAIN RESULTS We examined 36 samples from 23 patients with respiratory exacerbation. Fourteen samples tested virus-positive and 22 virus-negative. When we compared exacerbations associated with rhinovirus (RV, n = 9) and influenza (n = 5) with virus-negative specimens, we found distinct patterns of antiviral gene expression. RV was associated with greater than twofold induction of five genes, including those encoding the monocyte-attracting chemokines CXCL10, CXCL11, and CXCL9. Influenza was associated with overexpression of 20 genes, including those encoding the cytokines tumor necrosis factor and IL-12; the kinases MEK, TBK-1, and STAT-1; the apoptosis proteins caspase-8 and caspase-10; the influenza double-stranded RNA receptor RIG-I and its downstream effector MAVS; and pyrin, an IFN-stimulated protein involved in influenza resistance. CONCLUSIONS We conclude that virus-induced exacerbations of CF are associated with immune responses tailored to specific infections. Influenza induced a more potent response consisting of inflammation, whereas RV infection had a pronounced effect on chemokine expression. As far as we are aware, this study is the first to compare specific responses to different viruses in live patients with chronic airway disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Marc B. Hershenson
- Department of Pediatrics and Communicable Diseases
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
99
|
Foronjy RF, Dabo AJ, Taggart CC, Weldon S, Geraghty P. Respiratory syncytial virus infections enhance cigarette smoke induced COPD in mice. PLoS One 2014; 9:e90567. [PMID: 24587397 PMCID: PMC3938768 DOI: 10.1371/journal.pone.0090567] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 02/03/2014] [Indexed: 12/02/2022] Open
Abstract
Respiratory syncytial viral (RSV) infections are a frequent cause of chronic obstructive pulmonary disease (COPD) exacerbations, which are a major factor in disease progression and mortality. RSV is able to evade antiviral defenses to persist in the lungs of COPD patients. Though RSV infection has been identified in COPD, its contribution to cigarette smoke-induced airway inflammation and lung tissue destruction has not been established. Here we examine the long-term effects of cigarette smoke exposure, in combination with monthly RSV infections, on pulmonary inflammation, protease production and remodeling in mice. RSV exposures enhanced the influx of macrophages, neutrophils and lymphocytes to the airways of cigarette smoke exposed C57BL/6J mice. This infiltration of cells was most pronounced around the vasculature and bronchial airways. By itself, RSV caused significant airspace enlargement and fibrosis in mice and these effects were accentuated with concomitant smoke exposure. Combined stimulation with both smoke and RSV synergistically induced cytokine (IL-1α, IL-17, IFN-γ, KC, IL-13, CXCL9, RANTES, MIF and GM-CSF) and protease (MMP-2, -8, -12, -13, -16 and cathepsins E, S, W and Z) expression. In addition, RSV exposure caused marked apoptosis within the airways of infected mice, which was augmented by cigarette smoke exposure. RSV and smoke exposure also reduced protein phosphatase 2A (PP2A) and protein tyrosine phosphates (PTP1B) expression and activity. This is significant as these phosphatases counter smoke-induced inflammation and protease expression. Together, these findings show for the first time that recurrent RSV infection markedly enhances inflammation, apoptosis and tissue destruction in smoke-exposed mice. Indeed, these results indicate that preventing RSV transmission and infection has the potential to significantly impact on COPD severity and progression.
Collapse
Affiliation(s)
- Robert F. Foronjy
- St. Luke’s Roosevelt Hospital, Mount Sinai Health System, Division of Pulmonary and Critical Care Medicine, New York, New York, United States of America
| | - Abdoulaye J. Dabo
- St. Luke’s Roosevelt Hospital, Mount Sinai Health System, Division of Pulmonary and Critical Care Medicine, New York, New York, United States of America
| | - Clifford C. Taggart
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Sinead Weldon
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Patrick Geraghty
- St. Luke’s Roosevelt Hospital, Mount Sinai Health System, Division of Pulmonary and Critical Care Medicine, New York, New York, United States of America
| |
Collapse
|
100
|
Combined exposure to cigarette smoke and nontypeable Haemophilus influenzae drives development of a COPD phenotype in mice. Respir Res 2014; 15:11. [PMID: 24495712 PMCID: PMC3926338 DOI: 10.1186/1465-9921-15-11] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 02/03/2014] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Cigarette smoke (CS) is the major etiologic factor of chronic obstructive pulmonary disease (COPD). CS-exposed mice develop emphysema and mild pulmonary inflammation but no airway obstruction, which is also a prominent feature of COPD. Therefore, CS may interact with other factors, particularly respiratory infections, in the pathogenesis of airway remodeling in COPD. METHODS C57BL/6 mice were exposed to CS for 2 h a day, 5 days a week for 8 weeks. Mice were also exposed to heat-killed non-typeable H. influenzae (HK-NTHi) on days 7 and 21. One day after the last exposure to CS, mice were sacrificed and lung inflammation and mechanics, emphysematous changes, and goblet cell metaplasia were assessed. Mice exposed to CS or HK-NTHi alone or room air served as controls. To determine the susceptibility to viral infections, we also challenged these mice with rhinovirus (RV). RESULTS Unlike mice exposed to CS or HK-NTHi alone, animals exposed to CS/HK-NTHi developed emphysema, lung inflammation and goblet cell metaplasia in both large and small airways. CS/HK-NTHi-exposed mice also expressed increased levels of mucin genes and cytokines compared to mice in other groups. CS/HK-NTHi-exposed mice infected with RV demonstrated increased viral persistence, sustained neutrophilia, and further increments in mucin gene and chemokine expression compared to other groups. CONCLUSIONS These findings indicate that in addition to CS, bacteria may also contribute to development of COPD, particularly changes in airways. Mice exposed to CS/HK-NTHi are also more susceptible to subsequent viral infection than mice exposed to either CS or HK-NTHi alone.
Collapse
|