1
|
Rupani H, Busse WW, Howarth PH, Bardin PG, Adcock IM, Konno S, Jackson DJ. Therapeutic relevance of eosinophilic inflammation and airway viral interactions in severe asthma. Allergy 2024; 79:2589-2604. [PMID: 39087443 DOI: 10.1111/all.16242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 06/21/2024] [Accepted: 07/08/2024] [Indexed: 08/02/2024]
Abstract
The role of eosinophils in airway inflammation and asthma pathogenesis is well established, with raised eosinophil counts in blood and sputum associated with increased disease severity and risk of asthma exacerbation. Conversely, there is also preliminary evidence suggesting antiviral properties of eosinophils in the airways. These dual roles for eosinophils are particularly pertinent as respiratory virus infections contribute to asthma exacerbations. Biologic therapies targeting key molecules implicated in eosinophil-associated pathologies have been approved in patients with severe asthma and, therefore, the effects of depleting eosinophils in a clinical setting are of considerable interest. This review discusses the pathological and antiviral roles of eosinophils in asthma and exacerbations. We also highlight the significant reduction in asthma exacerbations seen with biologic therapies, even at the height of the respiratory virus season. Furthermore, we discuss the implications of these findings in relation to the role of eosinophils in inflammation and antiviral responses to respiratory virus infection in asthma.
Collapse
Affiliation(s)
- Hitasha Rupani
- Department of Respiratory Medicine, University Hospital Southampton NHS Foundation Trust, Southampton, Hampshire, UK
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, Hampshire, UK
| | - William W Busse
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Peter H Howarth
- Global Medical, Global Specialty and Primary Care, GSK, Brentford, Middlesex, UK
| | - Philip G Bardin
- Monash Lung Sleep Allergy and Immunology, Monash University and Medical Centre and Hudson Institute, Melbourne, Victoria, Australia
| | - Ian M Adcock
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Satoshi Konno
- Department of Respiratory Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - David J Jackson
- Guy's Severe Asthma Centre, Guy's and St Thomas' Hospitals, London, UK
- School of Immunology and Microbial Sciences, King's College London, London, UK
| |
Collapse
|
2
|
Kicic-Starcevich E, Hancock DG, Iosifidis T, Agudelo-Romero P, Caparros-Martin JA, Karpievitch YV, Silva D, Turkovic L, Le Souef PN, Bosco A, Martino DJ, Kicic A, Prescott SL, Stick SM. Airway epithelium respiratory illnesses and allergy (AERIAL) birth cohort: study protocol. FRONTIERS IN ALLERGY 2024; 5:1349741. [PMID: 38666051 PMCID: PMC11043573 DOI: 10.3389/falgy.2024.1349741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
Introduction Recurrent wheezing disorders including asthma are complex and heterogeneous diseases that affect up to 30% of all children, contributing to a major burden on children, their families, and global healthcare systems. It is now recognized that a dysfunctional airway epithelium plays a central role in the pathogenesis of recurrent wheeze, although the underlying mechanisms are still not fully understood. This prospective birth cohort aims to bridge this knowledge gap by investigating the influence of intrinsic epithelial dysfunction on the risk for developing respiratory disorders and the modulation of this risk by maternal morbidities, in utero exposures, and respiratory exposures in the first year of life. Methods The Airway Epithelium Respiratory Illnesses and Allergy (AERIAL) study is nested within the ORIGINS Project and will monitor 400 infants from birth to 5 years. The primary outcome of the AERIAL study will be the identification of epithelial endotypes and exposure variables that influence the development of recurrent wheezing, asthma, and allergic sensitisation. Nasal respiratory epithelium at birth to 6 weeks, 1, 3, and 5 years will be analysed by bulk RNA-seq and DNA methylation sequencing. Maternal morbidities and in utero exposures will be identified on maternal history and their effects measured through transcriptomic and epigenetic analyses of the amnion and newborn epithelium. Exposures within the first year of life will be identified based on infant medical history as well as on background and symptomatic nasal sampling for viral PCR and microbiome analysis. Daily temperatures and symptoms recorded in a study-specific Smartphone App will be used to identify symptomatic respiratory illnesses. Discussion The AERIAL study will provide a comprehensive longitudinal assessment of factors influencing the association between epithelial dysfunction and respiratory morbidity in early life, and hopefully identify novel targets for diagnosis and early intervention.
Collapse
Affiliation(s)
| | - David G. Hancock
- Wal-yan RespiratoryResearch Centre, Telethon Kids Institute, Perth, WA, Australia
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Nedlands, WA, Australia
- School of Medicine, The University of Western Australia, Nedlands, WA, Australia
| | - Thomas Iosifidis
- Wal-yan RespiratoryResearch Centre, Telethon Kids Institute, Perth, WA, Australia
- School of Population Health, Curtin University, Bentley, WA, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine, The University of Western Australia, Nedlands, WA, Australia
| | - Patricia Agudelo-Romero
- Wal-yan RespiratoryResearch Centre, Telethon Kids Institute, Perth, WA, Australia
- European Virus Bioinformatics Centre, Jena, Germany
| | | | | | - Desiree Silva
- School of Medicine, The University of Western Australia, Nedlands, WA, Australia
- Telethon Kids Institute, Perth, WA, Australia
- Department of Paediatrics and Neonatology, Joondalup Health Campus, Joondalup, WA, Australia
- School of Medicine and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | | | - Peter N. Le Souef
- Wal-yan RespiratoryResearch Centre, Telethon Kids Institute, Perth, WA, Australia
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Nedlands, WA, Australia
| | - Anthony Bosco
- School of Population Health, Curtin University, Bentley, WA, Australia
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, United States
- Department of Immunobiology, The University of Arizona College of Medicine, Tucson, AZ, United States
| | - David J. Martino
- Wal-yan RespiratoryResearch Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Anthony Kicic
- Wal-yan RespiratoryResearch Centre, Telethon Kids Institute, Perth, WA, Australia
- School of Population Health, Curtin University, Bentley, WA, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine, The University of Western Australia, Nedlands, WA, Australia
| | - Susan L. Prescott
- School of Medicine, The University of Western Australia, Nedlands, WA, Australia
- European Virus Bioinformatics Centre, Jena, Germany
| | - Stephen M. Stick
- Wal-yan RespiratoryResearch Centre, Telethon Kids Institute, Perth, WA, Australia
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Nedlands, WA, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
3
|
Russell RJ, Boulet LP, Brightling CE, Pavord ID, Porsbjerg C, Dorscheid D, Sverrild A. The airway epithelium: an orchestrator of inflammation, a key structural barrier and a therapeutic target in severe asthma. Eur Respir J 2024; 63:2301397. [PMID: 38453256 PMCID: PMC10991852 DOI: 10.1183/13993003.01397-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 02/15/2024] [Indexed: 03/09/2024]
Abstract
Asthma is a disease of heterogeneous pathology, typically characterised by excessive inflammatory and bronchoconstrictor responses to the environment. The clinical expression of the disease is a consequence of the interaction between environmental factors and host factors over time, including genetic susceptibility, immune dysregulation and airway remodelling. As a critical interface between the host and the environment, the airway epithelium plays an important role in maintaining homeostasis in the face of environmental challenges. Disruption of epithelial integrity is a key factor contributing to multiple processes underlying asthma pathology. In this review, we first discuss the unmet need in asthma management and provide an overview of the structure and function of the airway epithelium. We then focus on key pathophysiological changes that occur in the airway epithelium, including epithelial barrier disruption, immune hyperreactivity, remodelling, mucus hypersecretion and mucus plugging, highlighting how these processes manifest clinically and how they might be targeted by current and novel therapeutics.
Collapse
Affiliation(s)
- Richard J Russell
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | | | - Christopher E Brightling
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Ian D Pavord
- Respiratory Medicine, NIHR Oxford Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Celeste Porsbjerg
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg Hospital, Copenhagen University, Copenhagen, Denmark
| | - Del Dorscheid
- Centre for Heart Lung Innovation, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Asger Sverrild
- Department of Respiratory Medicine and Infectious Diseases, Bispebjerg Hospital, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
4
|
Yarlagadda T, Zhu Y, Snape N, Carey A, Bryan E, Maresco-Pennisi D, Coleman A, Cervin A, Spann K. Lactobacillus rhamnosus dampens cytokine and chemokine secretion from primary human nasal epithelial cells infected with rhinovirus. J Appl Microbiol 2024; 135:lxae018. [PMID: 38268489 DOI: 10.1093/jambio/lxae018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/15/2024] [Accepted: 01/22/2024] [Indexed: 01/26/2024]
Abstract
AIMS To investigate the effect of Lactobacillus rhamnosus on viral replication and cellular response to human rhinovirus (HRV) infection, including the secretion of antiviral and inflammatory mediators from well-differentiated nasal epithelial cells (WD-NECs). METHODS AND RESULTS The WD-NECs from healthy adult donors (N = 6) were cultured in vitro, exposed to different strains of L. rhamnosus (D3189, D3160, or LB21), and infected with HRV (RV-A16) after 24 h. Survival and adherence capacity of L. rhamnosus in a NEC environment were confirmed using CFSE-labelled isolates, immunofluorescent staining, and confocal microscopy. Shed virus and viral replication were quantified using TCID50 assays and RT-qPCR, respectively. Cytotoxicity was measured by lactate dehydrogenase (LDH) activity. Pro-inflammatory mediators were measured by multiplex immunoassay, and interferon (IFN)-λ1/3 was measured using a standard ELISA kit. Lactobacillus rhamnosus was able to adhere to and colonize WD-NECs prior to the RV-A16 infection. Lactobacillus rhamnosus did not affect shed RV-A16, viral replication, RV-A16-induced IFN-λ1/3 production, or LDH release. Pre-exposure to L. rhamnosus, particularly D3189, reduced the secretion of RV-A16-induced pro-inflammatory mediators by WD-NECs. CONCLUSIONS These findings demonstrate that L. rhamnosus differentially modulates RV-A16-induced innate inflammatory immune responses in primary NECs from healthy adults.
Collapse
Affiliation(s)
- Tejasri Yarlagadda
- Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane 4000, Australia
| | - Yanshan Zhu
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia 4072, Australia
| | - Natale Snape
- University of Queensland Frazer Institute, Woolloongabba 4102, Australia
| | - Alison Carey
- Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane 4000, Australia
| | - Emily Bryan
- Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane 4000, Australia
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Herston 4006, Australia
| | - Diane Maresco-Pennisi
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Herston 4006, Australia
| | - Andrea Coleman
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Herston 4006, Australia
| | - Anders Cervin
- Faculty of Medicine, University of Queensland Centre for Clinical Research, Herston 4006, Australia
| | - Kirsten Spann
- Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane 4000, Australia
| |
Collapse
|
5
|
Mitländer H, Yang Z, Krammer S, Grund JC, Zirlik S, Finotto S. Poly I:C Pre-Treatment Induced the Anti-Viral Interferon Response in Airway Epithelial Cells. Viruses 2023; 15:2328. [PMID: 38140569 PMCID: PMC10747011 DOI: 10.3390/v15122328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Type I and III interferons are among the most important antiviral mediators. Increased susceptibility to infections has been described as being associated with impaired interferon response in asthmatic patients. In this work, we focused on the modulation of interferon dysfunction after the rhinovirus infection of airway epithelial cells. Therefore, we tested polyinosinic:polycytidylic acid (poly I:C), a TLR3 agonist, as a possible preventive pre-treatment to improve this anti-viral response. In our human study on asthma, we found a deficiency in interferon levels in the nasal epithelial cells (NEC) from asthmatics at homeostatic level and after RV infection, which might contribute to frequent airway infection seen in asthmatic patients compared to healthy controls. Finally, pre-treatment with the immunomodulatory substance poly I:C before RV infection restored IFN responses in airway epithelial cells. Altogether, we consider poly I:C pre-treatment as a promising strategy for the induction of interferon response prior to viral infections. These results might help to improve current therapeutic strategies for allergic asthma exacerbations.
Collapse
Affiliation(s)
- Hannah Mitländer
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (H.M.); (Z.Y.); (S.K.); (J.C.G.)
| | - Zuqin Yang
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (H.M.); (Z.Y.); (S.K.); (J.C.G.)
| | - Susanne Krammer
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (H.M.); (Z.Y.); (S.K.); (J.C.G.)
| | - Janina C. Grund
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (H.M.); (Z.Y.); (S.K.); (J.C.G.)
| | - Sabine Zirlik
- Department of Medicine 1, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany;
| | - Susetta Finotto
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, 91054 Erlangen, Germany; (H.M.); (Z.Y.); (S.K.); (J.C.G.)
| |
Collapse
|
6
|
Fang L, Zhou L, Kulić Ž, Lehner MD, Tamm M, Roth M. EPs ® 7630 Stimulates Tissue Repair Mechanisms and Modifies Tight Junction Protein Expression in Human Airway Epithelial Cells. Int J Mol Sci 2023; 24:11230. [PMID: 37446408 DOI: 10.3390/ijms241311230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/29/2023] [Accepted: 07/01/2023] [Indexed: 07/15/2023] Open
Abstract
Airway epithelium repair after infection consists of wound repair, re-synthesis of the extracellular matrix (ECM), and tight junction proteins. In humans, EPs® 7630 obtained from Pelargonium sidoides roots reduces the severity and duration of acute respiratory tract infections. The effect of EPs® 7630 on tissue repair of rhinovirus-16 (RV-16) infected and control human airway epithelial cells was assessed for: (i) epithelial cell proliferation by manual cell counts, (ii) epithelial wound repair by "scratch assay", (iii) ECM composition by Western-blotting and cell-based ELISA, and (iv) epithelial tight junction proteins by Western-blotting. EPs® 7630 stimulated cell proliferation through cAMP, CREB, and p38 MAPK. EPs® 7630 significantly improved wound repair. Pro-inflammatory collagen type-I expression was reduced by EPs® 7630, while fibronectin was increased. Virus-binding tight junction proteins desmoglein2, desmocollin2, ZO-1, claudin1, and claudin4 were downregulated by EPs® 7630. The RV16-induced shift of the ECM towards the pro-inflammatory type was prevented by EPs® 7630. Most of the effects of EPs® 7630 on tissue repair and regeneration were sensitive to inhibition of cAMP-induced signaling. The data suggest that EPs® 7630-dependent modification of epithelial cell metabolism and function might underlie the faster recovery time from viral infections, as reported by others in clinical studies.
Collapse
Affiliation(s)
- Lei Fang
- Pulmonary Cell Research, Department of Biomedicine & Clinic of Pneumology, University and University Hospital Basel, CH-4031 Basel, Switzerland
| | - Liang Zhou
- Pulmonary Cell Research, Department of Biomedicine & Clinic of Pneumology, University and University Hospital Basel, CH-4031 Basel, Switzerland
| | - Žarko Kulić
- Preclinical Research and Development, Dr. Willmar Schwabe GmbH & Co. KG, D-76227 Karlsruhe, Germany
| | - Martin D Lehner
- Preclinical Research and Development, Dr. Willmar Schwabe GmbH & Co. KG, D-76227 Karlsruhe, Germany
| | - Michael Tamm
- Pulmonary Cell Research, Department of Biomedicine & Clinic of Pneumology, University and University Hospital Basel, CH-4031 Basel, Switzerland
| | - Michael Roth
- Pulmonary Cell Research, Department of Biomedicine & Clinic of Pneumology, University and University Hospital Basel, CH-4031 Basel, Switzerland
| |
Collapse
|
7
|
Kicic-Starcevich E, Hancock DG, Iosifidis T, Agudelo-Romero P, Caparros-Martin JA, Silva D, Turkovic L, Le Souef PN, Bosco A, Martino DJ, Kicic A, Prescott SL, Stick SM. Airway Epithelium Respiratory Illnesses and Allergy (AERIAL) birth cohort: study protocol. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.29.23289314. [PMID: 37205501 PMCID: PMC10187351 DOI: 10.1101/2023.04.29.23289314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Introduction Recurrent wheezing disorders including asthma are complex and heterogeneous diseases that affect up to 30% of all children, contributing to a major burden on children, their families, and global healthcare systems. It is now recognized that a dysfunctional airway epithelium plays a central role in the pathogenesis of recurrent wheeze, although the underlying mechanisms are still not fully understood. This prospective birth cohort aims to bridge this knowledge gap by investigating the influence of intrinsic epithelial dysfunction on the risk for developing respiratory disorders and the modulation of this risk by maternal morbidities, in utero exposures, and respiratory exposures in the first year of life. Methods and Analysis The Airway Epithelium Respiratory Illnesses and Allergy (AERIAL) study is nested within the ORIGINS Project and will monitor 400 infants from birth to five years. The primary outcome of the AERIAL study will be the identification of epithelial endotypes and exposure variables that influence the development of recurrent wheezing, asthma, and allergic sensitisation. Nasal respiratory epithelium at birth to six weeks, one, three, and five years will be analysed by bulk RNA-seq and DNA methylation sequencing. Maternal morbidities and in utero exposures will be identified on maternal history and their effects measured through transcriptomic and epigenetic analyses of the amnion and newborn epithelium. Exposures within the first year of life will be identified based on infant medical history as well as on background and symptomatic nasal sampling for viral PCR and microbiome analysis. Daily temperatures and symptoms recorded in a study-specific Smartphone App will be used to identify symptomatic respiratory illnesses. Ethics and Dissemination Ethical approval has been obtained from Ramsey Health Care HREC WA-SA (#1908). Results will be disseminated through open-access peer-reviewed manuscripts, conference presentations, and through different media channels to consumers, ORIGINS families, and the wider community.
Collapse
|
8
|
Landwehr KR, Nabi MN, Rasul MG, Kicic A, Mullins BJ. Biodiesel Exhaust Toxicity with and without Diethylene Glycol Dimethyl Ether Fuel Additive in Primary Airway Epithelial Cells Grown at the Air-Liquid Interface. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:14640-14648. [PMID: 36177943 DOI: 10.1021/acs.est.2c03806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Biodiesel usage is increasing steadily worldwide as the push for renewable fuel sources increases. The increased oxygen content in biodiesel fuel is believed to cause decreased particulate matter (PM) and increased nitrous oxides within its exhaust. The addition of fuel additives to further increase the oxygen content may contribute to even further benefits in exhaust composition. The aim of this study was to assess the toxicity of 10% (v/v) diethylene glycol dimethyl ether (DGDME) added as a biodiesel fuel additive. Primary human airway epithelial cells were grown at the air-liquid interface and exposed to diluted exhaust from an engine running on either grapeseed, bran, or coconut biodiesel or the same three biodiesels with 10% (v/v) DGDME added to them; mineral diesel and air were used as controls. Exhaust properties, culture permeability, epithelial cell damage, and IL-6 and IL-8 release were measured postexposure. The fuel additive DGDME caused a decrease in PM and nitrous oxide concentrations. However, exhaust exposure with DGDME also caused decreased permeability, increased epithelial cell damage, and increased release of IL-6 and IL-8 (p < 0.05). Despite the fuel additive having beneficial effects on the exhaust properties of the biodiesel, it was found to be more toxic.
Collapse
Affiliation(s)
- Katherine R Landwehr
- Occupation, Environment & Safety, School of Population Health, Curtin University, Perth, Western Australia 6102, Australia
- Respiratory Environmental Health, Telethon Kids Institute, Perth, Western Australia 6009, Australia
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, Western Australia 6009, Australia
| | - Md Nurun Nabi
- School of Engineering and Technology, Fuel and Energy Research Group, Central Queensland University, Perth, Western Australia 6000, Australia
| | - Mohammad G Rasul
- School of Engineering and Technology, Fuel and Energy Research Group, Central Queensland University, Rockhampton, Queensland 4701, Australia
| | - Anthony Kicic
- Occupation, Environment & Safety, School of Population Health, Curtin University, Perth, Western Australia 6102, Australia
- Respiratory Environmental Health, Telethon Kids Institute, Perth, Western Australia 6009, Australia
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, Western Australia 6009, Australia
- Department of of Respiratory and Sleep Medicine, Perth Children's Hospital, Perth, Western Australia 6009, Australia
- Centre for Cell Therapy and Regenerative Medicine, The University of Western Australia, Perth, Western Australia 6009, Australia
| | - Benjamin J Mullins
- Occupation, Environment & Safety, School of Population Health, Curtin University, Perth, Western Australia 6102, Australia
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Perth, Western Australia 6009, Australia
- St. John of God Hospital, Subiaco, Western Australia 6008, Australia
| |
Collapse
|
9
|
Kim SR. Viral Infection and Airway Epithelial Immunity in Asthma. Int J Mol Sci 2022; 23:9914. [PMID: 36077310 PMCID: PMC9456547 DOI: 10.3390/ijms23179914] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 12/19/2022] Open
Abstract
Viral respiratory tract infections are associated with asthma development and exacerbation in children and adults. In the course of immune responses to viruses, airway epithelial cells are the initial platform of innate immunity against viral invasion. Patients with severe asthma are more vulnerable than those with mild to moderate asthma to viral infections. Furthermore, in most cases, asthmatic patients tend to produce lower levels of antiviral cytokines than healthy subjects, such as interferons produced from immune effector cells and airway epithelial cells. The epithelial inflammasome appears to contribute to asthma exacerbation through overactivation, leading to self-damage, despite its naturally protective role against infectious pathogens. Given the mixed and complex immune responses in viral-infection-induced asthma exacerbation, this review examines the diverse roles of airway epithelial immunity and related potential therapeutic targets and discusses the mechanisms underlying the heterogeneous manifestations of asthma exacerbations.
Collapse
Affiliation(s)
- So Ri Kim
- Division of Respiratory Medicine and Allergy, Department of Internal Medicine, Medical School of Jeonbuk National University, 20 Geonji-ro, Deokjin-gu, Jeonju 54907, Korea
| |
Collapse
|
10
|
Likońska A, Gawrysiak M, Gajewski A, Klimczak K, Michlewska S, Szewczyk R, Gulbas I, Chałubiński M. Human lung vascular endothelium may limit viral replication and recover in time upon the infection with rhinovirus HRV16. APMIS 2022; 130:678-685. [PMID: 35959516 DOI: 10.1111/apm.13269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022]
Abstract
Vascular endothelium is a semi-permeable barrier that regulates the flow of nutrients, ions, cytokines, and immune cells between blood and tissues. Barrier properties of endothelium, its ability to regenerate, and the potential for secretion of inflammatory mediators play a crucial role in maintaining local tissue homeostasis. The lung vascular endothelial cells was shown to be infected by human rhinovirus and generate antiviral, inflammatory and cytopathic responses. The current study reveals that in the long-time manner the lung vascular endothelium may efficiently limit the HRV replication via the IFN-dependent 2'-5'-oligoadenylate synthetase 1 (OAS1) activation. This leads to the restoration of integrity accompanied by the up-regulation of adherens and tight junctions, increase of metabolic activity, and proliferation rate. Secondly, HRV16-infected cells show delayed and transient up-regulation of the expression of vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), angiopoietin 1 and 2, and neurophilin-1 (NRP-1), as well as VEGF receptors. The lung vascular endothelium infected with HRV may limit the infection, recover in time, and regain barrier properties and metabolic functions, thus leading to the restoration of integrated barrier tissue.
Collapse
Affiliation(s)
- Aleksandra Likońska
- Department of Immunology and Allergy, Chair of Pulmonology, Rheumatology and Clinical Immunology, Medical University of Lodz, Poland;Pomorska 251, 92-213 Lodz, Poland
| | - Mateusz Gawrysiak
- Department of Immunology and Allergy, Chair of Pulmonology, Rheumatology and Clinical Immunology, Medical University of Lodz, Poland;Pomorska 251, 92-213 Lodz, Poland
| | - Adrian Gajewski
- Department of Immunology and Allergy, Chair of Pulmonology, Rheumatology and Clinical Immunology, Medical University of Lodz, Poland;Pomorska 251, 92-213 Lodz, Poland
| | - Kinga Klimczak
- Department of Immunology and Allergy, Chair of Pulmonology, Rheumatology and Clinical Immunology, Medical University of Lodz, Poland;Pomorska 251, 92-213 Lodz, Poland
| | - Sylwia Michlewska
- Laboratory of Microscopic Imaging and Specialized Biological Techniques, Faculty of Biology and Environmental Protection, University of Lodz, Poland;Banacha12/16, 90-237 Lodz, Poland
| | - Robert Szewczyk
- Department of Immunology and Allergy, Chair of Pulmonology, Rheumatology and Clinical Immunology, Medical University of Lodz, Poland;Pomorska 251, 92-213 Lodz, Poland
| | - Izabela Gulbas
- Department of Immunology and Allergy, Chair of Pulmonology, Rheumatology and Clinical Immunology, Medical University of Lodz, Poland;Pomorska 251, 92-213 Lodz, Poland
| | - Maciej Chałubiński
- Department of Immunology and Allergy, Chair of Pulmonology, Rheumatology and Clinical Immunology, Medical University of Lodz, Poland;Pomorska 251, 92-213 Lodz, Poland
| |
Collapse
|
11
|
Zhu Y, Chew KY, Wu M, Karawita AC, McCallum G, Steele LE, Yamamoto A, Labzin LI, Yarlagadda T, Khromykh AA, Wang X, Sng JDJ, Stocks CJ, Xia Y, Kollmann TR, Martino D, Joensuu M, Meunier FA, Balistreri G, Bielefeldt-Ohmann H, Bowen AC, Kicic A, Sly PD, Spann KM, Short KR. Ancestral SARS-CoV-2, but not Omicron, replicates less efficiently in primary pediatric nasal epithelial cells. PLoS Biol 2022; 20:e3001728. [PMID: 35913989 PMCID: PMC9371332 DOI: 10.1371/journal.pbio.3001728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/11/2022] [Accepted: 06/24/2022] [Indexed: 01/02/2023] Open
Abstract
Children typically experience more mild symptoms of Coronavirus Disease 2019 (COVID-19) when compared to adults. There is a strong body of evidence that children are also less susceptible to Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection with the ancestral viral isolate. However, the emergence of SARS-CoV-2 variants of concern (VOCs) has been associated with an increased number of pediatric infections. Whether this is the result of widespread adult vaccination or fundamental changes in the biology of SARS-CoV-2 remain to be determined. Here, we use primary nasal epithelial cells (NECs) from children and adults, differentiated at an air-liquid interface to show that the ancestral SARS-CoV-2 replicates to significantly lower titers in the NECs of children compared to those of adults. This was associated with a heightened antiviral response to SARS-CoV-2 in the NECs of children. Importantly, the Delta variant also replicated to significantly lower titers in the NECs of children. This trend was markedly less pronounced in the case of Omicron. It is also striking to note that, at least in terms of viral RNA, Omicron replicated better in pediatric NECs compared to both Delta and the ancestral virus. Taken together, these data show that the nasal epithelium of children supports lower infection and replication of ancestral SARS-CoV-2, although this may be changing as the virus evolves.
Collapse
Affiliation(s)
- Yanshan Zhu
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Keng Yih Chew
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Melanie Wu
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Anjana C. Karawita
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Georgina McCallum
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Lauren E. Steele
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Ayaho Yamamoto
- Child Health Research Centre, The University of Queensland, South Brisbane, Queensland, Australia
| | - Larisa I. Labzin
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Tejasri Yarlagadda
- Centre for Immunology and Infection Control, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Alexander A. Khromykh
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
- Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Brisbane, Queensland, Australia
| | - Xiaohui Wang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Julian D. J. Sng
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Claudia J. Stocks
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Yao Xia
- School of Science, Edith Cowan University, Joondalup, Western Australia, Australia
- School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Tobias R. Kollmann
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - David Martino
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
| | - Merja Joensuu
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, Australia
| | - Frédéric A. Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Giuseppe Balistreri
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Helle Bielefeldt-Ohmann
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
- Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Brisbane, Queensland, Australia
| | - Asha C. Bowen
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
- Department of Infectious Diseases, Perth Children’s Hospital, Nedlands, Perth, Western Australia, Australia
- Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Anthony Kicic
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, The University of Western Australia, Perth, Western Australia, Australia
- Occupation and Environment, School of Public Health, Curtin University, Perth, Western Australia, Australia
- Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Perth, Western Australia, Australia
| | - Peter D. Sly
- Child Health Research Centre, The University of Queensland, South Brisbane, Queensland, Australia
- Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Brisbane, Queensland, Australia
| | - Kirsten M. Spann
- Centre for Immunology and Infection Control, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Kirsty R. Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
- Australian Infectious Diseases Research Centre, Global Virus Network Centre of Excellence, Brisbane, Queensland, Australia
| |
Collapse
|
12
|
Liew KY, Koh SK, Hooi SL, Ng MKL, Chee HY, Harith HH, Israf DA, Tham CL. Rhinovirus-Induced Cytokine Alterations With Potential Implications in Asthma Exacerbations: A Systematic Review and Meta-Analysis. Front Immunol 2022; 13:782936. [PMID: 35242128 PMCID: PMC8886024 DOI: 10.3389/fimmu.2022.782936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 01/13/2022] [Indexed: 12/01/2022] Open
Abstract
Background Rhinovirus (RV) infections are a major cause of asthma exacerbations. Unlike other respiratory viruses, RV causes minimal cytotoxic effects on airway epithelial cells and cytokines play a critical role in its pathogenesis. However, previous findings on RV-induced cytokine responses were largely inconsistent. Thus, this study sought to identify the cytokine/chemokine profiles induced by RV infection and their correlations with airway inflammatory responses and/or respiratory symptoms using systematic review, and to determine whether a quantitative difference exists in cytokine levels between asthmatic and healthy individuals via meta-analysis. Methods Relevant articles were obtained from PubMed, Scopus, and ScienceDirect databases. Studies that compared RV-induced cytokine responses between asthmatic and healthy individuals were included in the systematic review, and their findings were categorized based on the study designs, which were ex vivo primary bronchial epithelial cells (PBECs), ex vivo peripheral blood mononuclear cells (PBMCs), and human experimental studies. Data on cytokine levels were also extracted and analyzed using Review Manager 5.4. Results Thirty-four articles were included in the systematic review, with 18 of these further subjected to meta-analysis. Several studies reported the correlations between the levels of cytokines, such as IL-8, IL-4, IL-5, and IL-13, and respiratory symptoms. Evidence suggests that IL-25 and IL-33 may be the cytokines that promote type 2 inflammation in asthmatics after RV infection. Besides that, a meta-analysis revealed that PBECs from children with atopic asthma produced significantly lower levels of IFN-β [Effect size (ES): -0.84, p = 0.030] and IFN-λ (ES: -1.00, p = 0.002), and PBECs from adult atopic asthmatics produced significantly lower levels of IFN-β (ES: -0.68, p = 0.009), compared to healthy subjects after RV infection. A trend towards a deficient production of IFN-γ (ES: -0.56, p = 0.060) in PBMCs from adult atopic asthmatics was observed. In lower airways, asthmatics also had significantly lower baseline IL-15 (ES: -0.69, p = 0.020) levels. Conclusion Overall, RV-induced asthma exacerbations are potentially caused by an imbalance between Th1 and Th2 cytokines, which may be contributed by defective innate immune responses at cellular levels. Exogenous IFNs delivery may be beneficial as a prophylactic approach for RV-induced asthma exacerbations. Systematic Review Registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=184119, identifier CRD42020184119.
Collapse
Affiliation(s)
- Kong Yen Liew
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Sue Kie Koh
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Suet Li Hooi
- School of Science, Monash University Malaysia, Subang Jaya, Malaysia
| | | | - Hui-Yee Chee
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Hanis Hazeera Harith
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Daud Ahmad Israf
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Chau Ling Tham
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
13
|
Celebi Sozener Z, Ozdel Ozturk B, Cerci P, Turk M, Gorgulu Akin B, Akdis M, Altiner S, Ozbey U, Ogulur I, Mitamura Y, Yilmaz I, Nadeau K, Ozdemir C, Mungan D, Akdis CA. Epithelial barrier hypothesis: Effect of the external exposome on the microbiome and epithelial barriers in allergic disease. Allergy 2022; 77:1418-1449. [PMID: 35108405 PMCID: PMC9306534 DOI: 10.1111/all.15240] [Citation(s) in RCA: 155] [Impact Index Per Article: 77.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/25/2022] [Accepted: 01/29/2022] [Indexed: 12/11/2022]
Abstract
Environmental exposure plays a major role in the development of allergic diseases. The exposome can be classified into internal (e.g., aging, hormones, and metabolic processes), specific external (e.g., chemical pollutants or lifestyle factors), and general external (e.g., broader socioeconomic and psychological contexts) domains, all of which are interrelated. All the factors we are exposed to, from the moment of conception to death, are part of the external exposome. Several hundreds of thousands of new chemicals have been introduced in modern life without our having a full understanding of their toxic health effects and ways to mitigate these effects. Climate change, air pollution, microplastics, tobacco smoke, changes and loss of biodiversity, alterations in dietary habits, and the microbiome due to modernization, urbanization, and globalization constitute our surrounding environment and external exposome. Some of these factors disrupt the epithelial barriers of the skin and mucosal surfaces, and these disruptions have been linked in the last few decades to the increasing prevalence and severity of allergic and inflammatory diseases such as atopic dermatitis, food allergy, allergic rhinitis, chronic rhinosinusitis, eosinophilic esophagitis, and asthma. The epithelial barrier hypothesis provides a mechanistic explanation of how these factors can explain the rapid increase in allergic and autoimmune diseases. In this review, we discuss factors affecting the planet's health in the context of the 'epithelial barrier hypothesis,' including climate change, pollution, changes and loss of biodiversity, and emphasize the changes in the external exposome in the last few decades and their effects on allergic diseases. In addition, the roles of increased dietary fatty acid consumption and environmental substances (detergents, airborne pollen, ozone, microplastics, nanoparticles, and tobacco) affecting epithelial barriers are discussed. Considering the emerging data from recent studies, we suggest stringent governmental regulations, global policy adjustments, patient education, and the establishment of individualized control measures to mitigate environmental threats and decrease allergic disease.
Collapse
Affiliation(s)
| | - Betul Ozdel Ozturk
- School of MedicineDepartment of Chest DiseasesDivision of Immunology and Allergic DiseasesAnkara UniversityAnkaraTurkey
| | - Pamir Cerci
- Clinic of Immunology and Allergic DiseasesEskisehir City HospitalEskisehirTurkey
| | - Murat Turk
- Clinic of Immunology and Allergic DiseasesKayseri City HospitalKayseriTurkey
| | - Begum Gorgulu Akin
- Clinic of Immunology and Allergic DiseasesAnkara City HospitalAnkaraTurkey
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
| | - Seda Altiner
- Clinic of Internal Medicine Division of Immunology and Allergic DiseasesKahramanmaras Necip Fazil City HospitalKahramanmarasTurkey
| | - Umus Ozbey
- Department of Nutrition and DietAnkara UniversityAnkaraTurkey
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
| | - Insu Yilmaz
- Department of Chest DiseasesDivision of Immunology and Allergic DiseasesErciyes UniversityKayseriTurkey
| | - Kari Nadeau
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University School of MedicineDivision of Pulmonary and Critical Care MedicineDepartment of MedicineStanford UniversityStanfordCaliforniaUSA
| | - Cevdet Ozdemir
- Institute of Child HealthDepartment of Pediatric Basic SciencesIstanbul UniversityIstanbulTurkey
- Istanbul Faculty of MedicineDepartment of PediatricsDivision of Pediatric Allergy and ImmunologyIstanbul UniversityIstanbulTurkey
| | - Dilsad Mungan
- School of MedicineDepartment of Chest DiseasesDivision of Immunology and Allergic DiseasesAnkara UniversityAnkaraTurkey
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
- Christine Kühne‐Center for Allergy Research and Education (CK‐CARE)DavosSwitzerland
| |
Collapse
|
14
|
Hernandez-Pacheco N, Kere M, Melén E. Gene-environment interactions in childhood asthma revisited; expanding the interaction concept. Pediatr Allergy Immunol 2022; 33:e13780. [PMID: 35616899 PMCID: PMC9325482 DOI: 10.1111/pai.13780] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 04/13/2022] [Indexed: 01/04/2023]
Abstract
Investigation of gene-environment interactions (GxE) may provide important insights into the gene regulatory framework in response to environmental factors of relevance for childhood asthma. Over the years, different methodological strategies have been applied, more recently using genome-wide approaches. The best example to date is the major asthma locus on the 17q12-21 chromosome region, viral infections, and airway epithelium processes where recent studies have shed much light on mechanisms in childhood asthma. However, there are challenges with the traditional single variant-single exposure interaction models, as they do not encompass the complexity and cumulative effects of multiple exposures or multiple genetic variants. As such, we need to redefine our traditional GxE thinking, and we propose in this review to expand the GxE concept by also evaluating other omics layers, such as epigenetics, transcriptomics, metabolomics, and proteomics. In addition, host factors such as age, gender, and other exposures are very likely to influence GxE effects and need firmly to be considered in future studies.
Collapse
Affiliation(s)
- Natalia Hernandez-Pacheco
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden.,CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Maura Kere
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Erik Melén
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden.,Sachs' Children's Hospital, South General Hospital, Stockholm, Sweden
| |
Collapse
|
15
|
Feng X, Lawrence MG, Payne SC, Mattos J, Etter E, Negri JA, Murphy D, Kennedy JL, Steinke JW, Borish L. Lower viral loads in subjects with rhinovirus-challenged allergy despite reduced innate immunity. Ann Allergy Asthma Immunol 2022; 128:414-422.e2. [PMID: 35031416 PMCID: PMC10666001 DOI: 10.1016/j.anai.2022.01.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/04/2021] [Accepted: 01/05/2022] [Indexed: 02/02/2023]
Abstract
BACKGROUND Viral infections, especially those caused by rhinovirus, are the most common cause of asthma exacerbations. Previous studies have argued that impaired innate antiviral immunity and, as a consequence, more severe infections contribute to these exacerbations. OBJECTIVE These studies explored the innate immune response in the upper airway of volunteers with allergic rhinitis and asthma in comparison to healthy controls and interrogated how these differences corresponded to severity of infection. METHODS Volunteers with allergic rhinitis, those with asthma, and those who are healthy were inoculated with rhinovirus A16 and monitored for clinical symptoms. Tissue and nasal wash samples were evaluated for antiviral signature and viral load. RESULTS Both subjects with allergic rhinitis and asthma were found to have more severe cold symptoms. Subjects with asthma had worsened asthma control and increased bronchial hyperreactivity in the setting of higher fractional exhaled breath nitric oxide and blood eosinophils. These studies confirmed reduced expression of interferons and virus-specific pattern recognition receptors in both cohorts with atopy. Nevertheless, despite this defect in innate immunity, volunteers with allergic rhinitis/asthma had reduced rhinovirus concentrations in comparison to the controls. CONCLUSION These results confirm that the presence of an allergic inflammatory disorder of the airway is associated with reduced innate immune responsive to rhinovirus infection. Despite this, these volunteers with allergy have reduced viral loads, arguing for the presence of a compensatory mechanism to clear the infection. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02910401.
Collapse
Affiliation(s)
- Xin Feng
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, NHC Key Laboratory of Otorhinolaryngology (Shandong University), Jinan, Shandong, People's Republic of China
| | - Monica G Lawrence
- Department of Medicine, University of Virginia Health System, Charlottesville, Virginia; Department of Pediatrics, University of Virginia Health System, Charlottesville, Virginia
| | - Spencer C Payne
- Department of Otolaryngology, University of Virginia Health System, Charlottesville, Virginia
| | - Jose Mattos
- Department of Otolaryngology, University of Virginia Health System, Charlottesville, Virginia
| | - Elaine Etter
- Department of Medicine, University of Virginia Health System, Charlottesville, Virginia
| | - Julie A Negri
- Department of Medicine, University of Virginia Health System, Charlottesville, Virginia
| | - Deborah Murphy
- Department of Pediatrics, University of Virginia Health System, Charlottesville, Virginia
| | - Joshua L Kennedy
- Department of Pediatrics and Medicine, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, Arkansas
| | - John W Steinke
- Department of Medicine, University of Virginia Health System, Charlottesville, Virginia
| | - Larry Borish
- Department of Medicine, University of Virginia Health System, Charlottesville, Virginia; Department of Microbiology, University of Virginia Health System, Charlottesville, Virginia.
| |
Collapse
|
16
|
Watkinson RL, Looi K, Laing IA, Cianferoni A, Kicic A. Viral Induced Effects on a Vulnerable Epithelium; Lessons Learned From Paediatric Asthma and Eosinophilic Oesophagitis. Front Immunol 2021; 12:773600. [PMID: 34912343 PMCID: PMC8666438 DOI: 10.3389/fimmu.2021.773600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/05/2021] [Indexed: 01/07/2023] Open
Abstract
The epithelium is integral to the protection of many different biological systems and for the maintenance of biochemical homeostasis. Emerging evidence suggests that particular children have epithelial vulnerabilities leading to dysregulated barrier function and integrity, that resultantly contributes to disease pathogenesis. These epithelial vulnerabilities likely develop in utero or in early life due to various genetic, epigenetic and environmental factors. Although various epithelia are uniquely structured with specific function, prevalent allergic-type epithelial diseases in children potentially have common or parallel disease processes. These include inflammation and immune response dysregulation stemming from atypical epithelial barrier function and integrity. Two diseases where aetiology and pathogenesis are potentially linked to epithelial vulnerabilities include Paediatric Asthma and Eosinophilic Oesophagitis (EoE). For example, rhinovirus C (RV-C) is a known risk factor for paediatric asthma development and is known to disrupt respiratory epithelial barrier function causing acute inflammation. In addition, EoE, a prevalent atopic condition of the oesophageal epithelium, is characterised by similar innate immune and epithelial responses to viral injury. This review examines the current literature and identifies the gaps in the field defining viral-induced effects on a vulnerable respiratory epithelium and resulting chronic inflammation, drawing from knowledge generated in acute wheezing illness, paediatric asthma and EoE. Besides highlighting the importance of epithelial structure and barrier function in allergic disease pathogenesis regardless of specific epithelial sub-types, this review focuses on the importance of examining other parallel allergic-type disease processes that may uncover commonalities driving disease pathogenesis. This in turn may be beneficial in the development of common therapeutics for current clinical management and disease prevention in the future.
Collapse
Affiliation(s)
- Rebecca L Watkinson
- Division of Paediatrics, Medical School, The University of Western Australia, Nedlands, WA, Australia.,Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Kevin Looi
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA, Australia.,School of Public Health, Curtin University, Bentley, WA, Australia
| | - Ingrid A Laing
- Division of Paediatrics, Medical School, The University of Western Australia, Nedlands, WA, Australia.,Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Antonella Cianferoni
- Pediatrics Department, Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Anthony Kicic
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, Perth, WA, Australia.,School of Public Health, Curtin University, Bentley, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
17
|
Altman MC, Calatroni A, Ramratnam S, Jackson DJ, Presnell S, Rosasco MG, Gergen PJ, Bacharier LB, O'Connor GT, Sandel MT, Kattan M, Wood RA, Visness CM, Gern JE. Endotype of allergic asthma with airway obstruction in urban children. J Allergy Clin Immunol 2021; 148:1198-1209. [PMID: 33713771 PMCID: PMC8429519 DOI: 10.1016/j.jaci.2021.02.040] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 01/25/2021] [Accepted: 02/01/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Black and Hispanic children growing up in disadvantaged urban neighborhoods have the highest rates of asthma and related morbidity in the United States. OBJECTIVES This study sought to identify specific respiratory phenotypes of health and disease in this population, associations with early life exposures, and molecular patterns of gene expression in nasal epithelial cells that underlie clinical disease. METHODS The study population consisted of 442 high-risk urban children who had repeated assessments of wheezing, allergen-specific IgE, and lung function through 10 years of age. Phenotypes were identified by developing temporal trajectories for these data, and then compared to early life exposures and patterns of nasal epithelial gene expression at 11 years of age. RESULTS Of the 6 identified respiratory phenotypes, a high wheeze, high atopy, low lung function group had the greatest respiratory morbidity. In early life, this group had low exposure to common allergens and high exposure to ergosterol in house dust. While all high-atopy groups were associated with increased expression of a type-2 inflammation gene module in nasal epithelial samples, an epithelium IL-13 response module tracked closely with impaired lung function, and a MUC5AC hypersecretion module was uniquely upregulated in the high wheeze, high atopy, low lung function group. In contrast, a medium wheeze, low atopy group showed altered expression of modules of epithelial integrity, epithelial injury, and antioxidant pathways. CONCLUSIONS In the first decade of life, high-risk urban children develop distinct phenotypes of respiratory health versus disease that link early life environmental exposures to childhood allergic sensitization and asthma. Moreover, unique patterns of airway gene expression demonstrate how specific molecular pathways underlie distinct respiratory phenotypes, including allergic and nonallergic asthma.
Collapse
Affiliation(s)
- Matthew C Altman
- Immunology Division, Benaroya Research Institute Systems, Seattle, Wash; Department of Medicine, University of Washington, Seattle, Wash.
| | | | - Sima Ramratnam
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Daniel J Jackson
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Scott Presnell
- Immunology Division, Benaroya Research Institute Systems, Seattle, Wash
| | - Mario G Rosasco
- Immunology Division, Benaroya Research Institute Systems, Seattle, Wash
| | - Peter J Gergen
- National Institute of Allergy and Infectious Diseases, Rockville, Md
| | - Leonard B Bacharier
- Department of Pediatrics, Washington University School of Medicine and St Louis Children's Hospital, St Louis, Mo
| | - George T O'Connor
- Department of Medicine, Boston University School of Medicine, Boston, Mass
| | - Megan T Sandel
- Department of Medicine, Boston University School of Medicine, Boston, Mass
| | - Meyer Kattan
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY
| | - Robert A Wood
- Department of Pediatrics, Johns Hopkins University Medical Center, Baltimore, Md
| | | | - James E Gern
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| |
Collapse
|
18
|
Yang Z, Mitländer H, Vuorinen T, Finotto S. Mechanism of Rhinovirus Immunity and Asthma. Front Immunol 2021; 12:731846. [PMID: 34691038 PMCID: PMC8526928 DOI: 10.3389/fimmu.2021.731846] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/20/2021] [Indexed: 12/30/2022] Open
Abstract
The majority of asthma exacerbations in children are caused by Rhinovirus (RV), a positive sense single stranded RNA virus of the Picornavirus family. The host has developed virus defense mechanisms that are mediated by the upregulation of interferon-activated signaling. However, the virus evades the immune system by inducing immunosuppressive cytokines and surface molecules like programmed cell death protein 1 (PD-1) and its ligand (PD-L1) on immunocompetent cells. Initially, RV infects epithelial cells, which constitute a physiologic mucosal barrier. Upon virus entrance, the host cell immediately recognizes viral components like dsRNA, ssRNA, viral glycoproteins or CpG-DNA by host pattern recognition receptors (PRRs). Activation of toll like receptors (TLR) 3, 7 and 8 within the endosome and through MDA-5 and RIG-I in the cytosol leads to the production of interferon (IFN) type I and other antiviral agents. Every cell type expresses IFNAR1/IFNAR2 receptors thus allowing a generalized antiviral activity of IFN type I resulting in the inhibition of viral replication in infected cells and preventing viral spread to non-infected cells. Among immune evasion mechanisms of the virus, there is downregulation of IFN type I and its receptor as well as induction of the immunosuppressive cytokine TGF-β. TGF-β promotes viral replication and is associated with induction of the immunosuppression signature markers LAP3, IDO and PD-L1. This article reviews the recent advances on the regulation of interferon type I expression in association with RV infection in asthmatics and the immunosuppression induced by the virus.
Collapse
Affiliation(s)
- Zuqin Yang
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Hannah Mitländer
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Tytti Vuorinen
- Medical Microbiology, Turku University Hospital, Institut of Biomedicine, University of Turku, Turku, Finland
| | - Susetta Finotto
- Department of Molecular Pneumology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
19
|
Bonner K, Scotney E, Saglani S. Factors and mechanisms contributing to the development of preschool wheezing disorders. Expert Rev Respir Med 2021; 15:745-760. [PMID: 33881953 DOI: 10.1080/17476348.2021.1913057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Half of all children will experience an episode of wheezing by their sixth birthday and acute episodes of wheezing in preschool children account for the majority of all childhood hospital admissions for wheeze. Recurrent preschool wheezing associates with early loss of lung function and a life-long impact on lung health. AREAS COVERED We reviewed the literature on PubMed from August 2010-2020 focussing on factors associated with wheeze inception and persistence, paying specific attention to mechanistic studies that have investigated the impact of early life exposures in shaping immune responses in children with underlying susceptibility to wheezing. In particular, the role of early allergen sensitization, respiratory infections, and the impact of the environment on shaping the airway microbiome and resulting immune responses are discussed. EXPERT OPINION There is an abundance of associative data showing the role of in utero and postnatal factors influencing wheeze onset and persistence. However, mechanistic and stratified, biomarker-based interventional studies that confirm these associations are now needed if we are to impact the significant healthcare burden resulting from preschool wheezing disorders.
Collapse
Affiliation(s)
- Katie Bonner
- Inflammation, Repair & Development Section, National Heart & Lung Institute, Imperial College London, London, UK.,Department of Paediatric Respiratory Medicine, Royal Brompton Hospital, London, UK
| | - Elizabeth Scotney
- Inflammation, Repair & Development Section, National Heart & Lung Institute, Imperial College London, London, UK.,Department of Paediatric Respiratory Medicine, Royal Brompton Hospital, London, UK
| | - Sejal Saglani
- Inflammation, Repair & Development Section, National Heart & Lung Institute, Imperial College London, London, UK.,Department of Paediatric Respiratory Medicine, Royal Brompton Hospital, London, UK
| |
Collapse
|
20
|
Zhou J, Zhou XD, Xu R, Du XZ, Li Q, Li B, Zhang GY, Chen LX, Perelman JM, Kolosov VP. The Degradation of Airway Epithelial Tight Junctions in Asthma Under High Airway Pressure Is Probably Mediated by Piezo-1. Front Physiol 2021; 12:637790. [PMID: 33868003 PMCID: PMC8047413 DOI: 10.3389/fphys.2021.637790] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/11/2021] [Indexed: 12/02/2022] Open
Abstract
Full functioning of the airway physical barrier depends on cellular integrity, which is coordinated by a series of tight junction (TJ) proteins. Due to airway spasm, edema, and mucus obstruction, positive end-expiratory alveolar pressure (also termed auto-PEEP) is a common pathophysiological phenomenon, especially in acute asthma attack. However, the influence of auto-PEEP on small airway epithelial TJs is currently unclear. We performed studies to investigate the effect of extra pressure on small airway epithelial TJs and its mechanism. The results first confirmed that a novel mechanosensitive receptor, piezo-1, was highly expressed in the airway epithelium of asthmatic mice. Extra pressure induced the degradation of occludin, ZO-1 and claudin-18 in primary human small airway epithelial cells (HSAECs), resulting in a decrease in transepithelial electrical resistance (TER) and an increase in cell layer permeability. Through in vitro investigations, we observed that exogenous pressure stimulation could elevate the intracellular calcium concentration ([Ca2+]i) in HSAECs. Downregulation of piezo-1 with siRNA and pretreatment with BAPTA-AM or ALLN reduced the degradation of TJs and attenuated the impairment of TJ function induced by exogenous pressure. These findings indicate the critical role of piezo-1/[Ca2+]i/calpain signaling in the regulation of small airway TJs under extra pressure stimulation.
Collapse
Affiliation(s)
- Jia Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiang-Dong Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, China.,Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou, China
| | - Rui Xu
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xian-Zhi Du
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qi Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Bin Li
- Department of Respiratory and Critical Care Medicine, People's Hospital of Fengjie, Chongqing, China
| | - Guo-Yue Zhang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ling-Xiu Chen
- Department of Respiratory Medicine, Chongqing Three Gorges Central Hospital, Chongqing, China
| | - Juliy M Perelman
- Far Eastern Scientific Center of Physiology and Pathology of Respiration, Russian Academy of Medical Sciences, Blagoveshchensk, Russia
| | - Victor P Kolosov
- Far Eastern Scientific Center of Physiology and Pathology of Respiration, Russian Academy of Medical Sciences, Blagoveshchensk, Russia
| |
Collapse
|
21
|
Distinct Antiviral Properties of Two Different Bacterial Lysates. Can Respir J 2021; 2021:8826645. [PMID: 33613792 PMCID: PMC7878088 DOI: 10.1155/2021/8826645] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/20/2021] [Indexed: 12/23/2022] Open
Abstract
Oral bacterial lysates (OBLs) can reduce the frequency and severity of recurrent respiratory tract infections in children from viral and bacterial origins. OBL-induced early innate immune reaction was already shown, but the specific features of different OBLs have never been studied and compared. A study was conducted to assess in vitro the protective effects on rhinovirus- (RV-) infected human bronchial epithelial cells (BECs) of two slightly different OBLs: OM-85 and Pulmonarom. Furthermore, since immune cells represent the key arm for antiviral defence, the capacity of these OBLs to induce selected cytokine production in mouse bone marrow-derived DCs (BMDCs) was also evaluated. Although different OBLs may share some mechanisms to protect host cells from virus infection, some product-specific antimicrobial activities were observed on RV-infected human BECs and mouse BMDCs. These results are consistent with a product-specific response possibly triggered by different pathogen-associated molecular patterns (PAMPs) contained in OBLs.
Collapse
|
22
|
Tzani-Tzanopoulou P, Skliros D, Megremis S, Xepapadaki P, Andreakos E, Chanishvili N, Flemetakis E, Kaltsas G, Taka S, Lebessi E, Doudoulakakis A, Papadopoulos NG. Interactions of Bacteriophages and Bacteria at the Airway Mucosa: New Insights Into the Pathophysiology of Asthma. FRONTIERS IN ALLERGY 2021; 1:617240. [PMID: 35386933 PMCID: PMC8974763 DOI: 10.3389/falgy.2020.617240] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/29/2020] [Indexed: 12/16/2022] Open
Abstract
The airway epithelium is the primary site where inhaled and resident microbiota interacts between themselves and the host, potentially playing an important role on allergic asthma development and pathophysiology. With the advent of culture independent molecular techniques and high throughput technologies, the complex composition and diversity of bacterial communities of the airways has been well-documented and the notion of the lungs' sterility definitively rejected. Recent studies indicate that the microbial composition of the asthmatic airways across the spectrum of disease severity, differ significantly compared with healthy individuals. In parallel, a growing body of evidence suggests that bacterial viruses (bacteriophages or simply phages), regulating bacterial populations, are present in almost every niche of the human body and can also interact directly with the eukaryotic cells. The triptych of airway epithelial cells, bacterial symbionts and resident phages should be considered as a functional and interdependent unit with direct implications on the respiratory and overall homeostasis. While the role of epithelial cells in asthma pathophysiology is well-established, the tripartite interactions between epithelial cells, bacteria and phages should be scrutinized, both to better understand asthma as a system disorder and to explore potential interventions.
Collapse
Affiliation(s)
- Panagiota Tzani-Tzanopoulou
- Allergy and Clinical Immunology Unit, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Skliros
- Laboratory of Molecular Biology, Department of Biotechnology, School of Food, Biotechnology and Development, Agricultural University of Athens, Athens, Greece
| | - Spyridon Megremis
- Division of Evolution and Genomic Sciences, University of Manchester, Manchester, United Kingdom
| | - Paraskevi Xepapadaki
- Allergy and Clinical Immunology Unit, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Andreakos
- Center for Clinical, Experimental Surgery and Translational Research of the Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Nina Chanishvili
- Laboratory for Genetics of Microorganisms and Bacteriophages, Eliava Institute of Bacteriophage, Microbiology & Virology, Tbilisi, GA, United States
| | - Emmanouil Flemetakis
- Laboratory of Molecular Biology, Department of Biotechnology, School of Food, Biotechnology and Development, Agricultural University of Athens, Athens, Greece
| | - Grigoris Kaltsas
- Department of Electrical and Electronic Engineering, University of West Attica, Athens, Greece
| | - Styliani Taka
- Allergy and Clinical Immunology Unit, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelia Lebessi
- Department of Microbiology, P. & A. Kyriakou Children's Hospital, Athens, Greece
| | | | - Nikolaos G Papadopoulos
- Allergy and Clinical Immunology Unit, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece.,Division of Evolution and Genomic Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
23
|
The Airway Epithelium-A Central Player in Asthma Pathogenesis. Int J Mol Sci 2020; 21:ijms21238907. [PMID: 33255348 PMCID: PMC7727704 DOI: 10.3390/ijms21238907] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/20/2020] [Accepted: 11/21/2020] [Indexed: 12/11/2022] Open
Abstract
Asthma is a chronic inflammatory airway disease characterized by variable airflow obstruction in response to a wide range of exogenous stimuli. The airway epithelium is the first line of defense and plays an important role in initiating host defense and controlling immune responses. Indeed, increasing evidence indicates a range of abnormalities in various aspects of epithelial barrier function in asthma. A central part of this impairment is a disruption of the airway epithelial layer, allowing inhaled substances to pass more easily into the submucosa where they may interact with immune cells. Furthermore, many of the identified susceptibility genes for asthma are expressed in the airway epithelium. This review focuses on the biology of the airway epithelium in health and its pathobiology in asthma. We will specifically discuss external triggers such as allergens, viruses and alarmins and the effect of type 2 inflammatory responses on airway epithelial function in asthma. We will also discuss epigenetic mechanisms responding to external stimuli on the level of transcriptional and posttranscriptional regulation of gene expression, as well the airway epithelium as a potential treatment target in asthma.
Collapse
|
24
|
Novak N, Cabanillas B. Viruses and asthma: the role of common respiratory viruses in asthma and its potential meaning for SARS-CoV-2. Immunology 2020; 161:83-93. [PMID: 32687609 PMCID: PMC7405154 DOI: 10.1111/imm.13240] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/10/2020] [Accepted: 07/12/2020] [Indexed: 12/20/2022] Open
Abstract
Viral infections and atopic diseases are closely related and contribute to each other. The physiological deficiencies and immune mechanisms that underlie atopic diseases can result in a suboptimal defense against multiple viruses, and promote a suitable environment for their proliferation and dissemination. Viral infections, on the other hand, can induce per se several immunological mechanisms involved in allergic inflammation capable to promote the initiation or exacerbation of atopic diseases such as atopic asthma. In a world that is affected more and more by factors that significantly impact the prevalence of atopic diseases, coronavirus disease 2019 (COVID-19) induced by the novel coronavirus severe acute respiratory syndrome (SARS-CoV-2) is having an unprecedented impact with still unpredictable consequences. Therefore, it is of crucial importance to revise the available scientific literature regarding the association between common respiratory viruses and asthma, as well as the newly emerging data about the molecular mechanisms of SARS-CoV-2 infection and its possible relation with asthma, to better understand the interrelation between common viruses and asthma and its potential meaning on the current global pandemic of COVID-19.
Collapse
Affiliation(s)
- Natalija Novak
- Department of Dermatology and Allergy, University Hospital Bonn, Bonn, Germany
| | - Beatriz Cabanillas
- Department of Allergy, Research Institute Hospital 12 de Octubre, Madrid, Spain
| |
Collapse
|
25
|
Lejeune S, Deschildre A, Le Rouzic O, Engelmann I, Dessein R, Pichavant M, Gosset P. Childhood asthma heterogeneity at the era of precision medicine: Modulating the immune response or the microbiota for the management of asthma attack. Biochem Pharmacol 2020; 179:114046. [PMID: 32446884 PMCID: PMC7242211 DOI: 10.1016/j.bcp.2020.114046] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022]
Abstract
Exacerbations are a main characteristic of asthma. In childhood, the risk is increasing with severity. Exacerbations are a strong phenotypic marker, particularly of severe and therapy-resistant asthma. These early-life events may influence the evolution and be involved in lung function decline. In children, asthma attacks are facilitated by exposure to allergens and pollutants, but are mainly triggered by microbial agents. Multiple studies have assessed immune responses to viruses, and to a lesser extend bacteria, during asthma exacerbation. Research has identified impairment of innate immune responses in children, related to altered pathogen recognition, interferon release, or anti-viral response. Influence of this host-microbiota dialog on the adaptive immune response may be crucial, leading to the development of biased T helper (Th)2 inflammation. These dynamic interactions may impact the presentations of asthma attacks, and have long-term consequences. The aim of this review is to synthesize studies exploring immune mechanisms impairment against viruses and bacteria promoting asthma attacks in children. The potential influence of the nature of infectious agents and/or preexisting microbiota on the development of exacerbation is also addressed. We then discuss our understanding of how these diverse host-microbiota interactions in children may account for the heterogeneity of endotypes and clinical presentations. Finally, improving the knowledge of the pathophysiological processes induced by infections has led to offer new opportunities for the development of preventive or curative therapeutics for acute asthma. A better definition of asthma endotypes associated with precision medicine might lead to substantial progress in the management of severe childhood asthma.
Collapse
Affiliation(s)
- Stéphanie Lejeune
- CHU Lille, Univ. Lille, Pediatric Pulmonology and Allergy Department, Hôpital Jeanne de Flandre, F-59000 Lille, France; Univ. Lille, INSERM Unit 1019, CNRS UMR 9017, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille, F-59019 Lille Cedex, France
| | - Antoine Deschildre
- CHU Lille, Univ. Lille, Pediatric Pulmonology and Allergy Department, Hôpital Jeanne de Flandre, F-59000 Lille, France; Univ. Lille, INSERM Unit 1019, CNRS UMR 9017, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille, F-59019 Lille Cedex, France
| | - Olivier Le Rouzic
- Univ. Lille, INSERM Unit 1019, CNRS UMR 9017, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille, F-59019 Lille Cedex, France; CHU Lille, Univ. Lille, Department of Respiratory Diseases, F-59000 Lille Cedex, France
| | - Ilka Engelmann
- Univ. Lille, Virology Laboratory, EA3610, Institute of Microbiology, CHU Lille, F-59037 Lille Cedex, France
| | - Rodrigue Dessein
- Univ. Lille, INSERM Unit 1019, CNRS UMR 9017, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille, F-59019 Lille Cedex, France; Univ. Lille, Bacteriology Department, Institute of Microbiology, CHU Lille, F-59037 Lille Cedex, France
| | - Muriel Pichavant
- Univ. Lille, INSERM Unit 1019, CNRS UMR 9017, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille, F-59019 Lille Cedex, France
| | - Philippe Gosset
- Univ. Lille, INSERM Unit 1019, CNRS UMR 9017, CHU Lille, Institut Pasteur de Lille, Center for Infection and Immunity of Lille, F-59019 Lille Cedex, France.
| |
Collapse
|
26
|
Ling KM, Garratt LW, Gill EE, Lee AHY, Agudelo-Romero P, Sutanto EN, Iosifidis T, Rosenow T, Turvey SE, Lassmann T, Hancock REW, Kicic A, Stick SM. Rhinovirus Infection Drives Complex Host Airway Molecular Responses in Children With Cystic Fibrosis. Front Immunol 2020; 11:1327. [PMID: 32765492 PMCID: PMC7378398 DOI: 10.3389/fimmu.2020.01327] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 05/26/2020] [Indexed: 01/22/2023] Open
Abstract
Early-life viral infections are responsible for pulmonary exacerbations that can contribute to disease progression in young children with cystic fibrosis (CF). The most common respiratory viruses detected in the CF airway are human rhinoviruses (RV), and augmented airway inflammation in CF has been attributed to dysregulated airway epithelial responses although evidence has been conflicting. Here, we exposed airway epithelial cells from children with and without CF to RV in vitro. Using RNA-Seq, we profiled the transcriptomic differences of CF and non-CF airway epithelial cells at baseline and in response to RV. There were only modest differences between CF and non-CF cells at baseline. In response to RV, there were 1,442 and 896 differentially expressed genes in CF and non-CF airway epithelial cells, respectively. The core antiviral responses in CF and non-CF airway epithelial cells were mediated through interferon signaling although type 1 and 3 interferon signaling, when measured, were reduced in CF airway epithelial cells following viral challenge consistent with previous reports. The transcriptional responses in CF airway epithelial cells were more complex than in non-CF airway epithelial cells with diverse over-represented biological pathways, such as cytokine signaling and metabolic and biosynthetic pathways. Network analysis highlighted that the differentially expressed genes of CF airway epithelial cells' transcriptional responses were highly interconnected and formed a more complex network than observed in non-CF airway epithelial cells. We corroborate observations in fully differentiated air–liquid interface (ALI) cultures, identifying genes involved in IL-1 signaling and mucin glycosylation that are only dysregulated in the CF airway epithelial response to RV infection. These data provide novel insights into the CF airway epithelial cells' responses to RV infection and highlight potential pathways that could be targeted to improve antiviral and anti-inflammatory responses in CF.
Collapse
Affiliation(s)
- Kak-Ming Ling
- Paediatrics, Medical School, Faculty of Healthy and Medical Science, The University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia
| | - Luke W Garratt
- Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia.,School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Erin E Gill
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Amy H Y Lee
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Patricia Agudelo-Romero
- Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia
| | - Erika N Sutanto
- Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia
| | - Thomas Iosifidis
- Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia
| | - Tim Rosenow
- Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia
| | - Stuart E Turvey
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Timo Lassmann
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia
| | - Robert E W Hancock
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Anthony Kicic
- Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia.,School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia.,Occupation and Environment, School of Public Health, Curtin University, Perth, WA, Australia.,Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, WA, Australia
| | - Stephen M Stick
- Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia.,School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia.,Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
27
|
Cai Y, Varasteh S, van Putten JPM, Folkerts G, Braber S. Mannheimia haemolytica and lipopolysaccharide induce airway epithelial inflammatory responses in an extensively developed ex vivo calf model. Sci Rep 2020; 10:13042. [PMID: 32747652 PMCID: PMC7400546 DOI: 10.1038/s41598-020-69982-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 07/20/2020] [Indexed: 01/27/2023] Open
Abstract
Pulmonary infection is associated with inflammation and damage to the bronchial epithelium characterized by an increase in the release of inflammatory factors and a decrease in airway barrier function. Our objective is to optimize a method for the isolation and culture of primary bronchial epithelial cells (PBECs) and to provide an ex vivo model to study mechanisms of epithelial airway inflammation. PBECs were isolated and cultured from the airways of calves in a submerged cell culture and liquid-liquid interface system. A higher yield and cell viability were obtained after stripping the epithelium from the bronchial section compared to cutting the bronchial section in smaller pieces prior to digestion. Mannheimia haemolytica and lipopolysaccharide (LPS) as stimulants increased inflammatory responses (IL-8, IL-6 and TNF-α release), possibly, by the activation of "TLR-mediated MAPKs and NF-κB" signaling. Furthermore, M. haemolytica and LPS disrupted the bronchial epithelial layer as observed by a decreased transepithelial electrical resistance and zonula occludens-1 and E-cadherin expression. An optimized isolation and culture method for calf PBECs was developed, which cooperated with animal use Replacement, Reduction and Refinement (3R's) principle, and can also contribute to the increased knowledge and development of effective therapies for other animal and humans (childhood) respiratory diseases.
Collapse
Affiliation(s)
- Yang Cai
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Soheil Varasteh
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Jos P M van Putten
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - Gert Folkerts
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Saskia Braber
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
28
|
Montgomery ST, Frey DL, Mall MA, Stick SM, Kicic A. Rhinovirus Infection Is Associated With Airway Epithelial Cell Necrosis and Inflammation via Interleukin-1 in Young Children With Cystic Fibrosis. Front Immunol 2020; 11:596. [PMID: 32328066 PMCID: PMC7161373 DOI: 10.3389/fimmu.2020.00596] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/13/2020] [Indexed: 12/19/2022] Open
Abstract
Introduction: The responses of cystic fibrosis (CF) airway epithelial cells (AEC) to rhinovirus (RV) infection are likely to contribute to early pathobiology of lung disease with increased neutrophilic inflammation and lower apoptosis reported. Necrosis of AEC resulting in airway inflammation driven by IL-1 signaling is a characteristic finding in CF detectable in airways of young children. Being the most common early-life infection, RV-induced epithelial necrosis may contribute to early neutrophilic inflammation in CF via IL-1 signaling. As little is known about IL-1 and biology of CF lung disease, this study assessed cellular and pro-inflammatory responses of CF and non-CF AEC following RV infection, with the hypothesis that RV infection drives epithelial necrosis and IL-1 driven inflammation. Methods:Primary AEC obtained from children with (n = 6) and without CF (n = 6) were infected with RV (MOI 3) for 24 h and viable, necrotic and apoptotic events quantified via flow cytometry using a seven-step gating strategy (% total events). IL-1α, IL-1β, IL-1Ra, IL-8, CXCL10, CCL5, IFN-β, IL-28A, IL-28B, and IL-29 were also measured in cell culture supernatants (pg/mL). Results:RV infection reduced viable events in non-CF AEC (p < 0.05), increased necrotic events in non-CF and CF AEC (p < 0.05) and increased apoptotic events in non-CF AEC (p < 0.05). Infection induced IL-1α and IL-1β production in both phenotypes (p < 0.05) but only correlated with necrosis (IL-1α: r = 0.80; IL-1β: r = 0.77; p < 0.0001) in CF AEC. RV infection also increased IL-1Ra in non-CF and CF AEC (p < 0.05), although significantly more in non-CF AEC (p < 0.05). Finally, infection stimulated IL-8 production in non-CF and CF AEC (p < 0.05) and correlated with IL-1α (r = 0.63 & r = 0.74 respectively; p < 0.0001). Conclusions:This study found RV infection drives necrotic cell death in CF AEC. Furthermore, RV induced IL-1 strongly correlated with necrotic cell death in these cells. As IL-1R signaling drives airway neutrophilia and mucin production, these observations suggest RV infection early in life may exacerbate inflammation and mucin accumulation driving early CF lung disease. Since IL-1R can be targeted therapeutically with IL-1Ra, these data suggest a new anti-inflammatory therapeutic approach targeting downstream effects of IL-1R signaling to mitigate viral-induced, muco-inflammatory triggers of early lung disease.
Collapse
Affiliation(s)
- Samuel T Montgomery
- Faculty of Health and Medical Sciences, School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Dario L Frey
- Department of Translational Pulmonology, Translational Lung Research Center Heidelberg, University of Heidelberg, Heidelberg, Germany.,German Center for Lung Research, Heidelberg, Germany
| | - Marcus A Mall
- German Center for Lung Research, Heidelberg, Germany.,Department of Pediatric Pulmonology, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Stephen M Stick
- Faculty of Health and Medical Sciences, School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia.,Telethon Kids Institute, The University of Western Australia, Crawley, WA, Australia.,Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, WA, Australia
| | - Anthony Kicic
- Faculty of Health and Medical Sciences, School of Biomedical Sciences, The University of Western Australia, Crawley, WA, Australia.,Telethon Kids Institute, The University of Western Australia, Crawley, WA, Australia.,Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, WA, Australia.,School of Public Health, Curtin University, Bentley, WA, Australia.,Telethon Kids Institute, The University of Western Australia, Crawley, WA, Australia.,St John of God Hospital, Subiaco, WA, Australia
| | | |
Collapse
|
29
|
Iosifidis T, Sutanto EN, Buckley AG, Coleman L, Gill EE, Lee AH, Ling KM, Hillas J, Looi K, Garratt LW, Martinovich KM, Shaw NC, Montgomery ST, Kicic-Starcevich E, Karpievitch YV, Le Souëf P, Laing IA, Vijayasekaran S, Lannigan FJ, Rigby PJ, Hancock RE, Knight DA, Stick SM, Kicic A. Aberrant cell migration contributes to defective airway epithelial repair in childhood wheeze. JCI Insight 2020; 5:133125. [PMID: 32208383 DOI: 10.1172/jci.insight.133125] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 03/04/2020] [Indexed: 12/13/2022] Open
Abstract
Abnormal wound repair has been observed in the airway epithelium of patients with chronic respiratory diseases, including asthma. Therapies focusing on repairing vulnerable airways, particularly in early life, present a potentially novel treatment strategy. We report defective lower airway epithelial cell repair to strongly associate with common pre-school-aged and school-aged wheezing phenotypes, characterized by aberrant migration patterns and reduced integrin α5β1 expression. Next generation sequencing identified the PI3K/Akt pathway as the top upstream transcriptional regulator of integrin α5β1, where Akt activation enhanced repair and integrin α5β1 expression in primary cultures from children with wheeze. Conversely, inhibition of PI3K/Akt signaling in primary cultures from children without wheeze reduced α5β1 expression and attenuated repair. Importantly, the FDA-approved drug celecoxib - and its non-COX2-inhibiting analogue, dimethyl-celecoxib - stimulated the PI3K/Akt-integrin α5β1 axis and restored airway epithelial repair in cells from children with wheeze. When compared with published clinical data sets, the identified transcriptomic signature was also associated with viral-induced wheeze exacerbations highlighting the clinical potential of such therapy. Collectively, these results identify airway epithelial restitution via targeting the PI3K-integrin α5β1 axis as a potentially novel therapeutic avenue for childhood wheeze and asthma. We propose that the next step in the therapeutic development process should be a proof-of-concept clinical trial, since relevant animal models to test the crucial underlying premise are unavailable.
Collapse
Affiliation(s)
- Thomas Iosifidis
- Division of Pediatrics and.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine, University of Western Australia, Nedlands, Western Australia, Australia.,Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Erika N Sutanto
- Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Alysia G Buckley
- Centre of Microscopy, Characterisation and Analysis, University of Western Australia, Nedlands, Western Australia, Australia
| | - Laura Coleman
- Division of Pediatrics and.,Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Erin E Gill
- Center for Microbial Diseases Research, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amy H Lee
- Center for Microbial Diseases Research, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kak-Ming Ling
- Division of Pediatrics and.,Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Jessica Hillas
- Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Kevin Looi
- Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Luke W Garratt
- Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Kelly M Martinovich
- Division of Pediatrics and.,Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Nicole C Shaw
- Division of Pediatrics and.,Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Samuel T Montgomery
- Division of Pediatrics and.,Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | | | - Yuliya V Karpievitch
- Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Peter Le Souëf
- Division of Pediatrics and.,Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | - Ingrid A Laing
- Division of Pediatrics and.,Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia
| | | | - Francis J Lannigan
- School of Medicine, Notre Dame University, Fremantle, Western Australia, Australia
| | - Paul J Rigby
- Centre of Microscopy, Characterisation and Analysis, University of Western Australia, Nedlands, Western Australia, Australia
| | - Robert Ew Hancock
- Center for Microbial Diseases Research, University of British Columbia, Vancouver, British Columbia, Canada.,Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Stephen M Stick
- Division of Pediatrics and.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine, University of Western Australia, Nedlands, Western Australia, Australia.,Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia.,Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, Western Australia, Australia
| | - Anthony Kicic
- Division of Pediatrics and.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine, University of Western Australia, Nedlands, Western Australia, Australia.,Telethon Kids Institute Respiratory Research Centre, Perth, Western Australia, Australia.,Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, Western Australia, Australia.,School of Public Health, Curtin University, Bentley, Western Australia, Australia
| | | | | |
Collapse
|
30
|
He N, Liu L, Ding J, Sun Y, Xing H, Wang S. MiR-222-3p ameliorates glucocorticoid-induced inhibition of airway epithelial cell repair through down-regulating GILZ expression. J Recept Signal Transduct Res 2020; 40:301-312. [PMID: 32202184 DOI: 10.1080/10799893.2020.1742739] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
GILZ expression is induced by glucocorticoids (GCs) and is involved in the mechanism of airway epithelial cell repair in patients with asthma. The present study aimed to investigate the role of miR-222-3p/GILZ pathway in treatment of airway epithelial cell repair by GCs. 9HTE cells were treated by 10 µmol/L dexamethasone (Dex) for 6, 12, and 24 hours (h). MiR-222-3p mimic and GILZ were used for cell transfection. Cell vitality, migration, and invasion were detected by methyl-thiazolyl tetrazolium (MTT), wound healing, and Transwell. The targeting relationship between miR-222-3p and GILZ was predicted by TargetScan and further confirmed by dual-luciferase reporter assay. The expressions of relative mRNAs or proteins were detected by Western blot and quantitative polymerase chain reaction (qPCR). The results showed that Dex treatment up-regulated the GILZ expression level but inhibited the levels of p-Raf1, p-MEK1/2, p-ERK1/2, and miR-222-3p of the cells, moreover, it also inhibited cell activity, migration, and invasion in a time-dependent manner. MiR-222-3p specifically targeted GILZ. MiR-222-3p mimic ameliorated the cell viability, migration, and invasion reduced by Dex treatment, increased the expression levels of p-Raf1 and p-MEK1/2, p-ERK1/2, and partially reversed the effects of GILZ overexpression on the above indexes. Moreover, GILZ showed the opposite effects to miR-222-3p. MiR-222-3p activated MAPK signaling pathway through inhibiting the GILZ expression, thus promoting the cell viability, migration, and invasion previously reduced by Dex.
Collapse
Affiliation(s)
- Ning He
- Department of Allergy, Yantai Yuhuangding Hospital, Yantai, China
| | - Liping Liu
- Department of Allergy, Yantai Yuhuangding Hospital, Yantai, China
| | - Juan Ding
- Department of Allergy, Yantai Yuhuangding Hospital, Yantai, China
| | - Yuemei Sun
- Department of Allergy, Yantai Yuhuangding Hospital, Yantai, China
| | - Haiyan Xing
- Department of Allergy, Yantai Yuhuangding Hospital, Yantai, China
| | - Shuyun Wang
- Department of Allergy, Yantai Yuhuangding Hospital, Yantai, China
| |
Collapse
|
31
|
Roberts G. A complicated relationship between peanut environmental exposure and the development of allergic sensitization to peanuts. Clin Exp Allergy 2020; 48:488-489. [PMID: 29701920 DOI: 10.1111/cea.13145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- G Roberts
- Faculty of Medicine, Clinical and Experimental Sciences and Human Development and Health, University of Southampton, Southampton, UK.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK.,The David Hide Asthma and Allergy Research Centre, St Mary's Hospital, Isle of Wight, UK
| |
Collapse
|
32
|
Persson C. In vivo observations provide insight into roles of eosinophils and epithelial cells in asthma. Eur Respir J 2019; 54:13993003.00470-2019. [PMID: 31248957 DOI: 10.1183/13993003.00470-2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 06/18/2019] [Indexed: 01/10/2023]
Abstract
Observations in vivo in patients, supported by guinea-pig in vivo data, take centre stage in this perspective. Its objective is to highlight dichotomies between asthma features observed in vivo and accepted views involving cell/molecular biology research paradigms. For example, increased bronchial epithelial permeability is now considered a major paradigm and trait of asthma, yet, absorption of inhaled tracers has not been increased in vivo in asthma. Such maintained barrier function in exudative asthma reflects in vivo asymmetry of the epithelial lining as barrier between outside and inside world of molecules and cells. In desquamatory asthma, maintained epithelial tightness may be explained by in vivo demonstrations of exceedingly patchy epithelial loss, prompt creation of plasma-derived provisional barriers, and high-speed epithelial regeneration. Acknowledged protein/peptide secretion by epithelial cells in vitro is contrasted here with a dominant, unidirectional movement in vivo of plasma-derived proteins/peptides (including antimicrobial peptides) to the surface of an intact epithelial lining. Furthermore, longstanding claims that epithelium-produced adenosine is a mediator of asthma are eroded by observations in vivo in asthmatics. Notions concerning activation/fate of mucosal tissue eosinophils illustrate additional distinctions between accepted views and in vivo patient observations. Finally, in vitro-based paradigms preaching defect epithelial regeneration and increased permeability in pathogenesis of asthma are contrasted with experimental in vivo observations of exaggerated epithelial regeneration, which is multipathogenic in its own right. In conclusion, unexpected and challenging in vivo observations in recent decades underpin novel insights into mucosal mechanisms in asthma.
Collapse
Affiliation(s)
- Carl Persson
- Laboratory Medicine University Hospital, Lund, Sweden
| |
Collapse
|
33
|
Chen X, Qiu C. Respiratory tract mucous membrane microecology and asthma. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:495. [PMID: 31700931 PMCID: PMC6803190 DOI: 10.21037/atm.2019.09.06] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
According to the world health organization, the increasing incidence of asthma is placing a heavy burden on the social economy. Its high rate of disability and mortality has become a serious social and public health problem. Asthma is a heterogeneous disease in which genetic polymorphism interacts with environmental factors. Because the pathogenesis of asthma is not completely clear, there is no specific treatment. In 2010, 16S rRNA gene sequencing showed that lungs have many different microbial communities in both healthy and sick states. These microbial communities and respiratory mucosa constitute the respiratory mucosal microecology. When the respiratory mucosal microecology changes, it can play a key role in the occurrence and development of asthma and other respiratory diseases by regulating the immune mechanism. This paper reviews the latest research results in this field, and tries to explore the effects of changes in respiratory mucosal microecology on the pathogenesis of asthma, so as to provide new methods for early diagnosis, treatment and prevention of asthma.
Collapse
Affiliation(s)
- Xingyuan Chen
- Department of Respiratory and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen 518020, China
| | - Chen Qiu
- Department of Respiratory and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen 518020, China
| |
Collapse
|
34
|
Baraldo S, Contoli M, Bonato M, Snijders D, Biondini D, Bazzan E, Cosio MG, Barbato A, Papi A, Saetta M. Deficient Immune Response to Viral Infections in Children Predicts Later Asthma Persistence. Am J Respir Crit Care Med 2019; 197:673-675. [PMID: 28862881 DOI: 10.1164/rccm.201706-1249le] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
| | | | | | | | | | | | - Manuel G Cosio
- 1 University of Padova Padova, Italy.,3 McGill University Montreal, Canada
| | | | | | | |
Collapse
|
35
|
Ling KM, Hillas J, Lavender MA, Wrobel JP, Musk M, Stick SM, Kicic A. Azithromycin reduces airway inflammation induced by human rhinovirus in lung allograft recipients. Respirology 2019; 24:1212-1219. [PMID: 30989728 DOI: 10.1111/resp.13550] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 03/05/2019] [Accepted: 03/13/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND OBJECTIVE Human rhinovirus (RV) is a common upper and lower respiratory pathogen in lung allograft recipients causing respiratory tract exacerbation and contributing towards allograft dysfunction and long-term lung decline. In this study, we tested the hypothesis that RV could infect both the small and large airways, resulting in significant inflammation. METHODS Matched large and small airway epithelial cells (AEC) were obtained from five lung allograft recipients. Primary cultures were established, and monolayers were infected with RV1b over time with varying viral titre. Cell viability, receptor expression, viral copy number, apoptotic induction and inflammatory cytokine production were also assessed at each region. Finally, the effect of azithromycin on viral replication, induction of apoptosis and inflammation was investigated. RESULTS RV infection caused significant cytotoxicity in both large AEC (LAEC) and small AEC (SAEC), and induced a similar apoptotic response in both regions. There was a significant increase in receptor expression in the LAEC only post viral infection. Viral replication was elevated in both LAEC and SAEC, but was not significantly different. Prophylactic treatment of azithromycin reduced viral replication and dampened the production of inflammatory cytokines post-infection. CONCLUSION Our data illustrate that RV infection is capable of infecting upper and lower AEC, driving cell death and inflammation. Prophylactic treatment with azithromycin was found to mitigate some of the detrimental responses. Findings provide further support for the prophylactic prescription of azithromycin to minimize the impact of RV infection.
Collapse
Affiliation(s)
- Kak-Ming Ling
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Perth, WA, Australia
| | - Jessica Hillas
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Perth, WA, Australia
| | - Melanie A Lavender
- Advanced Lung Disease Unit, Fiona Stanley Hospital, Perth, WA, Australia
| | - Jeremy P Wrobel
- Advanced Lung Disease Unit, Fiona Stanley Hospital, Perth, WA, Australia.,Department of Medicine, University of Notre Dame, Fremantle, WA, Australia
| | - Michael Musk
- Advanced Lung Disease Unit, Fiona Stanley Hospital, Perth, WA, Australia
| | - Stephen M Stick
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Perth, WA, Australia.,School of Biomedical Science, The University of Western Australia, Perth, WA, Australia.,Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Perth, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Perth, WA, Australia
| | - Anthony Kicic
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Perth, WA, Australia.,Occupation and Environment, School of Public Health, Curtin University, Perth, WA, Australia.,School of Biomedical Science, The University of Western Australia, Perth, WA, Australia.,Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Perth, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
36
|
Increased susceptibility of airway epithelial cells from ataxia-telangiectasia to S. pneumoniae infection due to oxidative damage and impaired innate immunity. Sci Rep 2019; 9:2627. [PMID: 30796268 PMCID: PMC6385340 DOI: 10.1038/s41598-019-38901-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 12/19/2018] [Indexed: 12/25/2022] Open
Abstract
Respiratory disease is a major cause of morbidity and mortality in patients with ataxia-telangiectasia (A-T) who are prone to recurrent sinopulmonary infections, bronchiectasis, pulmonary fibrosis, and pulmonary failure. Upper airway infections are common in patients and S. pneumoniae is associated with these infections. We demonstrate here that the upper airway microbiome in patients with A-T is different from that to healthy controls, with S. pneumoniae detected largely in patients only. Patient-specific airway epithelial cells and differentiated air-liquid interface cultures derived from these were hypersensitive to infection which was at least in part due to oxidative damage since it was partially reversed by catalase. We also observed increased levels of the pro-inflammatory cytokines IL-8 and TNF-α (inflammasome-independent) and a decreased level of the inflammasome-dependent cytokine IL-β in patient cells. Further investigation revealed that the ASC-Caspase 1 signalling pathway was defective in A-T airway epithelial cells. These data suggest that the heightened susceptibility of these cells to S. pneumoniae infection is due to both increased oxidative damage and a defect in inflammasome activation, and has implications for lung disease in these patients.
Collapse
|
37
|
Trend S, Chang BJ, O'Dea M, Stick SM, Kicic A. Use of a Primary Epithelial Cell Screening Tool to Investigate Phage Therapy in Cystic Fibrosis. Front Pharmacol 2018; 9:1330. [PMID: 30546305 PMCID: PMC6280614 DOI: 10.3389/fphar.2018.01330] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 10/29/2018] [Indexed: 01/11/2023] Open
Abstract
Antimicrobial-resistant microbes are an increasing threat to human health. In cystic fibrosis (CF), airway infections with Pseudomonas aeruginosa remain a key driver of lung damage. With few new antibiotics on the development horizon, alternative therapeutic approaches are needed against antimicrobial-resistant pathogens. Phage therapy, or the use of viruses that infect bacteria, is one proposed novel therapy to treat bacterial infections. However, the airways are complex microenvironments with unique characteristics that may affect the success of novel therapies. Here, three phages of P. aeruginosa (E79, F116, and one novel clinically derived isolate, designated P5) were screened for activity against 21 P. aeruginosa strains isolated from children with CF. Of these, phage E79 showed broad antibacterial activity (91% of tested strains sensitive) and was selected for further assessment. E79 genomic DNA was extracted, sequenced, and confirmed to contain no bacterial pathogenicity genes. High titre phage preparations were then purified using ion-exchange column chromatography and depleted of bacterial endotoxin. Primary airway epithelial cells derived from children with CF (n = 8, age range 0.2–5.5 years, 5 males) or healthy non-CF controls (n = 8, age range 2.5–4.0 years, 4 males) were then exposed to purified phage for 48 h. Levels of inflammatory IL-1β, IL-6, and IL-8 cytokine production were measured in culture supernatant by immunoassays and the extent of cellular apoptosis was measured using a ssDNA kit. Cytokine and apoptosis levels were compared between E79-stimulated and unstimulated controls, and, encouragingly, purified preparations of E79 did not stimulate any significant inflammatory cytokine responses or induce apoptosis in primary epithelial cells derived from children with or without CF. Collectively, this study demonstrates the feasibility of utilizing pre-clinical in vitro culture models to screen therapeutic candidates, and the potential of E79 as a therapeutic phage candidate in CF.
Collapse
Affiliation(s)
- Stephanie Trend
- Telethon Kids Institute, Perth, WA, Australia.,Division of Paediatrics, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Barbara J Chang
- The Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Mark O'Dea
- School of Veterinary and Life Sciences, Murdoch University, Perth, WA, Australia
| | - Stephen M Stick
- Telethon Kids Institute, Perth, WA, Australia.,Division of Paediatrics, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia.,Department of Respiratory Medicine, Perth Children's Hospital, Perth, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia and Harry Perkins Institute of Medical Research, Perth, WA, Australia
| | - Anthony Kicic
- Telethon Kids Institute, Perth, WA, Australia.,Division of Paediatrics, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia.,Department of Respiratory Medicine, Perth Children's Hospital, Perth, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia and Harry Perkins Institute of Medical Research, Perth, WA, Australia.,Occupation and the Environment, School of Public Health, Curtin University, Perth, WA, Australia
| | | | | | | |
Collapse
|
38
|
Roberts G. A next-generation anti-IgE monoclonal? Clin Exp Allergy 2018; 46:1370-1371. [PMID: 27790844 DOI: 10.1111/cea.12832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- G Roberts
- Clinical and Experimental Sciences and Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK. .,NIHR Southampton Respiratory Biomedical Research Unit, University Hospital Southampton NHS Foundation Trust, Southampton, UK. .,The David Hide Asthma and Allergy Research Centre, St Mary's Hospital, Isle of Wight, UK.
| |
Collapse
|
39
|
Persson C. Airways exudation of plasma macromolecules: Innate defense, epithelial regeneration, and asthma. J Allergy Clin Immunol 2018; 143:1271-1286. [PMID: 30170125 PMCID: PMC7112321 DOI: 10.1016/j.jaci.2018.07.037] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 06/30/2018] [Accepted: 07/13/2018] [Indexed: 01/09/2023]
Abstract
This review discusses in vivo airway aspects of plasma exudation in relation to current views on epithelial permeability and epithelial regeneration in health and disease. Microvascular-epithelial exudation of bulk plasma proteins characteristically occurs in asthmatic patients, being especially pronounced in those with severe and exacerbating asthma. Healthy human and guinea pig airways challenged by noninjurious histamine-leukotriene–type autacoids also respond through prompt mucosal exudation of nonsieved plasma macromolecules. Contrary to current beliefs, epithelial permeability in the opposite direction (ie, absorption of inhaled molecules) has not been increased in patients with asthma and allergic rhinitis or in acutely exuding healthy airways. A slightly increased subepithelial hydrostatic pressure produces such unidirectional outward perviousness to macromolecules. Lack of increased absorption permeability in asthmatic patients can further be reconciled with occurrence of epithelial shedding, leaving small patches of denuded basement membrane. Counteracting escalating barrier breaks, plasma exudation promptly covers the denuded patches. Here it creates and sustains a biologically active barrier involving a neutrophil-rich, fibrin-fibronectin net. Furthermore, in the plasma-derived milieu, all epithelial cell types bordering the denuded patch dedifferentiate and migrate from all sides to cover the denuded basement membrane. However, this speedy epithelial regeneration can come at a cost. Guinea pig in vivo studies demonstrate that patches of epithelial denudation regeneration are exudation hot spots evoking asthma-like features, including recruitment/activation of granulocytes, proliferation of fibrocytes/smooth muscle cells, and basement membrane thickening. In conclusion, nonsieved plasma macromolecules can operate on the intact airway mucosa as potent components of first-line innate immunity responses. Exuded plasma also takes center stage in epithelial regeneration. When exaggerated, epithelial regeneration can contribute to the inception and development of asthma.
Collapse
Affiliation(s)
- Carl Persson
- Department of Laboratory Medicine, University Hospital of Lund, Lund, Sweden.
| |
Collapse
|
40
|
The Cell Research Trends of Asthma: A Stem Frequency Analysis of the Literature. JOURNAL OF HEALTHCARE ENGINEERING 2018; 2018:9363820. [PMID: 30210753 PMCID: PMC6126072 DOI: 10.1155/2018/9363820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 03/26/2018] [Accepted: 07/22/2018] [Indexed: 02/06/2023]
Abstract
Objective This study summarized asthma literature indexed in the Medical Literature Analysis and Retrieval System Online (MEDLINE) and explored the history and present trends of asthma cell research by stem frequency ranking to forecast the prospect of future work. Methods Literature was obtained from MEDLINE for the past 30 years and divided into three groups by decade as the retrieval time. The frequency of stemmed words in each group was calculated using Python with Apache Spark and the Natural Language Tool Kit for ranking. The unique stems or shared stems of 3 decades were summarized. Results A total of 1331, 4393, and 7215 records were retrieved from 3 decades chronologically, and the stem ranking of the top 50 were listed by frequency. The number of stems shared with 3 decades was 26 and with the first and last 2 decades was 5 and 13. Conclusions The number of cell research studies of asthma has increased rapidly, and scholars have paid more attentions on experimental research, especially on mechanistic research. Eosinophils, mast cells, and T cells are the hot spots of immunocyte research, while epithelia and smooth muscle cells are the hot spots of structural cell research. The research trend is closely linked with the development of experimental technology, including animal models. Early studies featured basic research, but immunity research has dominated in recent decades. The distinct definition of asthma phenotypes associated with genetic characteristics, immunity research, and the introduction of new cells will be the hot spots in future work.
Collapse
|
41
|
Stenberg Hammar K, Niespodziana K, van Hage M, Kere J, Valenta R, Hedlin G, Söderhäll C. Reduced CDHR3 expression in children wheezing with rhinovirus. Pediatr Allergy Immunol 2018; 29:200-206. [PMID: 29314338 DOI: 10.1111/pai.12858] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/15/2017] [Indexed: 01/12/2023]
Abstract
BACKGROUND Rhinovirus-induced wheezing in young children has been associated with increased asthma risk at school age. Recently, the transmembrane protein cadherin-related family member 3 (CDHR3) was identified as the RV-C receptor and the genetic variant rs6967330 (p.Cys529Tyr) was reported to be associated with enhanced RV-C binding and increased replication in vitro. The aim of this study was to examine rs6967330 genotypes and mRNA expression of CDHR3 in relation to presence of rhinovirus and clinical symptoms in children with acute wheezing and compare to a group of age-matched healthy children. METHODS rs6967330;G>A was genotyped (n = 216), and CDHR3 mRNA expression was measured in peripheral blood leukocytes (n = 69) from a subgroup of children wheezing with RV infection acute and at a follow-up visit 2-3 months later, and in healthy controls. Standardized TaqMan assays were used. RESULTS The risk allele rs6967330-A was over-represented in the wheezing group (P < .001). Reduced mRNA levels of CDHR3 were found in children with acute wheezing as compared to the control group (P = .001). Children with the rs6967330 genotypes AA/AG showed the largest differences in CDHR3 expression between acute and follow-up visit (P < .04). CONCLUSIONS Preschool children with RV-induced wheezing were shown to have reduced CDHR3 mRNA levels, which might result in an increased permeability of the epithelial layers of the airways and thereby an increased vulnerability. Thus, measuring CDHR3 mRNA levels might help to identify a more severe phenotype of wheezing preschool children.
Collapse
Affiliation(s)
- Katarina Stenberg Hammar
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden.,Centre of Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Katarzyna Niespodziana
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Marianne van Hage
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden.,Molecular Neurology Research Program, University of Helsinki and Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Rudolf Valenta
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Gunilla Hedlin
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden.,Centre of Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Cilla Söderhäll
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Centre of Allergy Research, Karolinska Institutet, Stockholm, Sweden.,Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
42
|
Looi K, Buckley AG, Rigby PJ, Garratt LW, Iosifidis T, Zosky GR, Larcombe AN, Lannigan FJ, Ling KM, Martinovich KM, Kicic-Starcevich E, Shaw NC, Sutanto EN, Knight DA, Kicic A, Stick SM. Effects of human rhinovirus on epithelial barrier integrity and function in children with asthma. Clin Exp Allergy 2018; 48:513-524. [PMID: 29350877 DOI: 10.1111/cea.13097] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/13/2017] [Accepted: 11/21/2017] [Indexed: 11/29/2022]
Abstract
BACKGROUND Bronchial epithelial tight junctions (TJ) have been extensively assessed in healthy airway epithelium. However, no studies have yet assessed the effect of human rhinovirus (HRV) infection on the expression and resultant barrier function in epithelial tight junctions (TJ) in childhood asthma. OBJECTIVES To investigate the impact of HRV infection on airway epithelial TJ expression and barrier function in airway epithelial cells (AECs) of children with and without asthma. Furthermore, to test the hypothesis that barrier integrity and function is compromised to a greater extent by HRV in AECs from asthmatic children. METHODS Primary AECs were obtained from children with and without asthma, differentiated into air-liquid interface (ALI) cultures and infected with rhinovirus. Expression of claudin-1, occludin and zonula occluden-1 (ZO-1) was assessed via qPCR, immunocytochemistry (ICC), in-cell western (ICW) and confocal microscopy. Barrier function was assessed by transepithelial electrical resistance (TER; RT ) and permeability to fluorescent dextran. RESULTS Basal TJ gene expression of claudin-1 and occludin was significantly upregulated in asthmatic children compared to non-asthmatics; however, no difference was seen with ZO-1. Interestingly, claudin-1, occludin and ZO-1 protein expression was significantly reduced in AEC of asthmatic children compared to non-asthmatic controls suggesting possible post-transcriptional inherent differences. HRV infection resulted in a transient dissociation of TJ and airway barrier integrity in non-asthmatic children. Although similar dissociation of TJ was observed in asthmatic children, a significant and sustained reduction in TJ expression concurrent with both a significant decrease in TER and an increase in permeability in asthmatic children was observed. CONCLUSION This study demonstrates novel intrinsic differences in TJ gene and protein expression between AEC of children with and without asthma. Furthermore, it correlates directly the relationship between HRV infection and the resultant dissociation of epithelial TJ that causes a continued altered barrier function in children with asthma.
Collapse
Affiliation(s)
- K Looi
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - A G Buckley
- Centre for Microscopy, Characterisation and Analysis (CMCA), University of Western Australia, Crawley, WA, Australia
| | - P J Rigby
- Centre for Microscopy, Characterisation and Analysis (CMCA), University of Western Australia, Crawley, WA, Australia
| | - L W Garratt
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - T Iosifidis
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Nedlands, WA, Australia
| | - G R Zosky
- School of Medicine, Faculty of Health, University of Tasmania, Hohart, TAS, Australia
| | - A N Larcombe
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Occupation and Environment, School of Public Health, Curtin University, Perth, WA, Australia
| | - F J Lannigan
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,School of Medicine, Notre Dame University, Fremantle, WA, Australia
| | - K-M Ling
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - K M Martinovich
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - E Kicic-Starcevich
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia
| | - N C Shaw
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - E N Sutanto
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia
| | - D A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Priority Research Centre for Asthma and Respiratory Disease, Hunter Medical Research Institute, Newcastle, NSW, Australia.,Department of Anaesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - A Kicic
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Nedlands, WA, Australia.,Occupation and Environment, School of Public Health, Curtin University, Perth, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia
| | - S M Stick
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Nedlands, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia
| |
Collapse
|
43
|
Moskwa S, Piotrowski W, Marczak J, Pawełczyk M, Lewandowska-Polak A, Jarzębska M, Brauncajs M, Głobińska A, Górski P, Papadopoulos NG, Edwards MR, Johnston SL, Kowalski ML. Innate Immune Response to Viral Infections in Primary Bronchial Epithelial Cells is Modified by the Atopic Status of Asthmatic Patients. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2018; 10:144-154. [PMID: 29411555 PMCID: PMC5809763 DOI: 10.4168/aair.2018.10.2.144] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Revised: 09/10/2017] [Accepted: 09/24/2017] [Indexed: 12/18/2022]
Abstract
Purpose In order to gain an insight into determinants of reported variability in immune responses to respiratory viruses in human bronchial epithelial cells (HBECs) from asthmatics, the responses of HBEC to viral infections were evaluated in HBECs from phenotypically heterogeneous groups of asthmatics and in healthy controls. Methods HBECs were obtained during bronchoscopy from 10 patients with asthma (6 atopic and 4 non-atopic) and from healthy controls (n=9) and grown as undifferentiated cultures. HBECs were infected with parainfluenza virus (PIV)-3 (MOI 0.1) and rhinovirus (RV)-1B (MOI 0.1), or treated with medium alone. The cell supernatants were harvested at 8, 24, and 48 hours. IFN-α, CXCL10 (IP-10), and RANTES (CCL5) were analyzed by using Cytometric Bead Array (CBA), and interferon (IFN)-β and IFN-λ1 by ELISA. Gene expression of IFNs, chemokines, and IFN-regulatory factors (IRF-3 and IRF-7) was determined by using quantitative PCR. Results PIV3 and RV1B infections increased IFN-λ1 mRNA expression in HBECs from asthmatics and healthy controls to a similar extent, and virus-induced IFN-λ1 expression correlated positively with IRF-7 expression. Following PIV3 infection, IP-10 protein release and mRNA expression were significantly higher in asthmatics compared to healthy controls (median 36.03-fold). No differences in the release or expression of RANTES, IFN-λ1 protein and mRNA, or IFN-α and IFN-β mRNA between asthmatics and healthy controls were observed. However, when asthmatics were divided according to their atopic status, HBECs from atopic asthmatics (n=6) generated significantly more IFN-λ1 protein and demonstrated higher IFN-α, IFN-β, and IRF-7 mRNA expressions in response to PIV3 compared to non-atopic asthmatics (n=4) and healthy controls (n=9). In response to RV1B infection, IFN-β mRNA expression was lower (12.39-fold at 24 hours and 19.37-fold at 48 hours) in non-atopic asthmatics compared to atopic asthmatics. Conclusions The immune response of HBECs to virus infections may not be deficient in asthmatics, but seems to be modified by atopic status.
Collapse
Affiliation(s)
- Sylwia Moskwa
- Department of Immunology, Rheumatology and Allergy; Healthy Ageing Research Centre, Medical University of Lodz, Lodz, Poland.,Department of Microbiology and Laboratory Medical Immunology, Medical University of Lodz, Lodz, Poland
| | - Wojciech Piotrowski
- Department of Pneumonology and Allergy, Medical University of Lodz, Lodz, Poland
| | - Jerzy Marczak
- Department of Pneumonology and Allergy, Medical University of Lodz, Lodz, Poland
| | - Małgorzata Pawełczyk
- Department of Immunology, Rheumatology and Allergy; Healthy Ageing Research Centre, Medical University of Lodz, Lodz, Poland
| | - Anna Lewandowska-Polak
- Department of Immunology, Rheumatology and Allergy; Healthy Ageing Research Centre, Medical University of Lodz, Lodz, Poland.,Department of Rheumatology, Medical University of Lodz, Lodz, Poland
| | - Marzanna Jarzębska
- Department of Immunology, Rheumatology and Allergy; Healthy Ageing Research Centre, Medical University of Lodz, Lodz, Poland
| | - Małgorzata Brauncajs
- Department of Immunology, Rheumatology and Allergy; Healthy Ageing Research Centre, Medical University of Lodz, Lodz, Poland
| | - Anna Głobińska
- Department of Immunology, Rheumatology and Allergy; Healthy Ageing Research Centre, Medical University of Lodz, Lodz, Poland
| | - Paweł Górski
- Department of Pneumonology and Allergy, Medical University of Lodz, Lodz, Poland
| | - Nikolaos G Papadopoulos
- Allergy Research Centre, 2nd Pediatric Clinic, National Kapodistrian, University of Athens, Athens, Greece
| | - Michael R Edwards
- National Heart and Lung Institute, Imperial College London, London, UK; Asthma UK Centre in Allergic Mechanisms of Asthma
| | - Sebastian L Johnston
- National Heart and Lung Institute, Imperial College London, London, UK; Asthma UK Centre in Allergic Mechanisms of Asthma
| | - Marek L Kowalski
- Department of Immunology, Rheumatology and Allergy; Healthy Ageing Research Centre, Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
44
|
Martinovich KM, Iosifidis T, Buckley AG, Looi K, Ling KM, Sutanto EN, Kicic-Starcevich E, Garratt LW, Shaw NC, Montgomery S, Lannigan FJ, Knight DA, Kicic A, Stick SM. Conditionally reprogrammed primary airway epithelial cells maintain morphology, lineage and disease specific functional characteristics. Sci Rep 2017; 7:17971. [PMID: 29269735 PMCID: PMC5740081 DOI: 10.1038/s41598-017-17952-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 12/04/2017] [Indexed: 01/19/2023] Open
Abstract
Current limitations to primary cell expansion led us to test whether airway epithelial cells derived from healthy children and those with asthma and cystic fibrosis (CF), co-cultured with an irradiated fibroblast feeder cell in F-medium containing 10 µM ROCK inhibitor could maintain their lineage during expansion and whether this is influenced by underlying disease status. Here, we show that conditionally reprogrammed airway epithelial cells (CRAECs) can be established from both healthy and diseased phenotypes. CRAECs can be expanded, cryopreserved and maintain phenotypes over at least 5 passages. Population doublings of CRAEC cultures were significantly greater than standard cultures, but maintained their lineage characteristics. CRAECs from all phenotypes were also capable of fully differentiating at air-liquid interface (ALI) and maintained disease specific characteristics including; defective CFTR channel function cultures and the inability to repair wounds. Our findings indicate that CRAECs derived from children maintain lineage, phenotypic and importantly disease-specific functional characteristics over a specified passage range.
Collapse
Affiliation(s)
- Kelly M Martinovich
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Thomas Iosifidis
- School of Paediatrics and Child Health, The University of Western Australia, Crawley, Western Australia, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Alysia G Buckley
- Centre of Microscopy, Characterisation and Analysis, The University of Western Australia, Crawley, Western Australia, Australia
| | - Kevin Looi
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Kak-Ming Ling
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Erika N Sutanto
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Elizabeth Kicic-Starcevich
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Luke W Garratt
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Nicole C Shaw
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Samuel Montgomery
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia
| | - Francis J Lannigan
- School of Paediatrics and Child Health, The University of Western Australia, Crawley, Western Australia, Australia
| | - Darryl A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,Priority Research Centre for Asthma and Respiratory Disease, Hunter Medical Research Institute, Newcastle, New South Wales, Australia.,Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, Canada
| | - Anthony Kicic
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia. .,School of Paediatrics and Child Health, The University of Western Australia, Crawley, Western Australia, Australia. .,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, Western Australia, Australia. .,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia. .,Occupation and Environment, School of Public Health, Curtin University, Perth, Western Australia, Australia.
| | - Stephen M Stick
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Crawley, Western Australia, Australia.,School of Paediatrics and Child Health, The University of Western Australia, Crawley, Western Australia, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, Western Australia, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, Western Australia, Australia
| |
Collapse
|
45
|
Edwards MR, Strong K, Cameron A, Walton RP, Jackson DJ, Johnston SL. Viral infections in allergy and immunology: How allergic inflammation influences viral infections and illness. J Allergy Clin Immunol 2017; 140:909-920. [PMID: 28987220 PMCID: PMC7173222 DOI: 10.1016/j.jaci.2017.07.025] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 07/20/2017] [Accepted: 07/31/2017] [Indexed: 12/20/2022]
Abstract
Viral respiratory tract infections are associated with asthma inception in early life and asthma exacerbations in older children and adults. Although how viruses influence asthma inception is poorly understood, much research has focused on the host response to respiratory viruses and how viruses can promote; or how the host response is affected by subsequent allergen sensitization and exposure. This review focuses on the innate interferon-mediated host response to respiratory viruses and discusses and summarizes the available evidence that this response is impaired or suboptimal. In addition, the ability of respiratory viruses to act in a synergistic or additive manner with TH2 pathways will be discussed. In this review we argue that these 2 outcomes are likely linked and discuss the available evidence that shows reciprocal negative regulation between innate interferons and TH2 mediators. With the renewed interest in anti-TH2 biologics, we propose a rationale for why they are particularly successful in controlling asthma exacerbations and suggest ways in which future clinical studies could be used to find direct evidence for this hypothesis.
Collapse
Affiliation(s)
- Michael R Edwards
- COPD & Asthma Section, National Heart Lung Institute, Imperial College London, London, United Kingdom; MRC & Asthma UK Centre for Allergic Mechanisms of Asthma, London, United Kingdom.
| | - Katherine Strong
- COPD & Asthma Section, National Heart Lung Institute, Imperial College London, London, United Kingdom; MRC & Asthma UK Centre for Allergic Mechanisms of Asthma, London, United Kingdom
| | - Aoife Cameron
- COPD & Asthma Section, National Heart Lung Institute, Imperial College London, London, United Kingdom; MRC & Asthma UK Centre for Allergic Mechanisms of Asthma, London, United Kingdom
| | - Ross P Walton
- COPD & Asthma Section, National Heart Lung Institute, Imperial College London, London, United Kingdom; MRC & Asthma UK Centre for Allergic Mechanisms of Asthma, London, United Kingdom
| | - David J Jackson
- COPD & Asthma Section, National Heart Lung Institute, Imperial College London, London, United Kingdom; MRC & Asthma UK Centre for Allergic Mechanisms of Asthma, London, United Kingdom; Guy's & St Thomas's Hospital London, London, United Kingdom
| | - Sebastian L Johnston
- COPD & Asthma Section, National Heart Lung Institute, Imperial College London, London, United Kingdom; MRC & Asthma UK Centre for Allergic Mechanisms of Asthma, London, United Kingdom
| |
Collapse
|
46
|
Loxham M, Davies DE. Phenotypic and genetic aspects of epithelial barrier function in asthmatic patients. J Allergy Clin Immunol 2017; 139:1736-1751. [PMID: 28583446 PMCID: PMC5457128 DOI: 10.1016/j.jaci.2017.04.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/13/2017] [Accepted: 04/14/2017] [Indexed: 12/22/2022]
Abstract
The bronchial epithelium is continuously exposed to a multitude of noxious challenges in inhaled air. Cellular contact with most damaging agents is reduced by the action of the mucociliary apparatus and by formation of a physical barrier that controls passage of ions and macromolecules. In conjunction with these defensive barrier functions, immunomodulatory cross-talk between the bronchial epithelium and tissue-resident immune cells controls the tissue microenvironment and barrier homeostasis. This is achieved by expression of an array of sensors that detect a wide variety of viral, bacterial, and nonmicrobial (toxins and irritants) agents, resulting in production of many different soluble and cell-surface molecules that signal to cells of the immune system. The ability of the bronchial epithelium to control the balance of inhibitory and activating signals is essential for orchestrating appropriate inflammatory and immune responses and for temporally modulating these responses to limit tissue injury and control the resolution of inflammation during tissue repair. In asthmatic patients abnormalities in many aspects of epithelial barrier function have been identified. We postulate that such abnormalities play a causal role in immune dysregulation in the airways by translating gene-environment interactions that underpin disease pathogenesis and exacerbation.
Collapse
Affiliation(s)
- Matthew Loxham
- Clinical and Experimental Sciences and the Southampton NIHR Respiratory Biomedical Research Unit, University of Southampton Faculty of Medicine, Sir Henry Wellcome Laboratories, University Hospital Southampton, Southampton, United Kingdom
| | - Donna E Davies
- Clinical and Experimental Sciences and the Southampton NIHR Respiratory Biomedical Research Unit, University of Southampton Faculty of Medicine, Sir Henry Wellcome Laboratories, University Hospital Southampton, Southampton, United Kingdom.
| |
Collapse
|
47
|
Fayon M, Lacoste-Rodrigues A, Barat P, Helbling JC, Nacka F, Berger P, Moisan MP, Corcuff JB. Nasal airway epithelial cell IL-6 and FKBP51 gene expression and steroid sensitivity in asthmatic children. PLoS One 2017; 12:e0177051. [PMID: 28493984 PMCID: PMC5426685 DOI: 10.1371/journal.pone.0177051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 04/21/2017] [Indexed: 12/11/2022] Open
Abstract
Background Many asthmatic patients exhibit uncontrolled asthma despite high-dose inhaled corticosteroids (ICS). Airway epithelial cells (AEC) have distinct activation profiles that can influence ICS response. Objectives A pilot study to identify gene expression markers of AEC dysfunction and markers of corticosteroid sensitivity in asthmatic and non-asthmatic control children, for comparison with published reports in adults. Methods AEC were obtained by nasal brushings and primary submerged cultures, and incubated in control conditions or in the presence of 10 ng/ml TNFalpha, 10-8M dexamethasone, or both. RT-PCR-based expression of FKBP51 (a steroid hormone receptor signalling regulator), NF-kB, IL-6, LIF (an IL-6 family neurotrophic cytokine), serpinB2 (which inhibits plasminogen activation and promotes fibrin deposition) and porin (a marker of mitochondrial mass) were determined. Results 6 patients without asthma (median age 11yr; min-max: 7–13), 8 with controlled asthma (11yr, 7–13; median daily fluticasone dose = 100 μg), and 4 with uncontrolled asthma (12yr, 7–14; 1000 μg fluticasone daily) were included. Baseline expression of LIF mRNA was significantly increased in uncontrolled vs controlled asthmatic children. TNFalpha significantly increased LIF expression in uncontrolled asthma. A similar trend was observed regarding IL-6. Dexamethasone significantly upregulated FKBP51 expression in all groups but the response was blunted in asthmatic children. No significant upregulation was identified regarding NF-kB, serpinB2 and porin. Conclusion LIF and FKBP51 expression in epithelial cells were the most interesting markers of AEC dysfunction/response to corticosteroid treatment.
Collapse
Affiliation(s)
- Michael Fayon
- Université de Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Bordeaux, France
- CHU de Bordeaux, Centre d’Investigation Clinique (CIC 1401), Bordeaux, France
- * E-mail:
| | - Aurelie Lacoste-Rodrigues
- Université de Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Bordeaux, France
- CHU de Bordeaux, Centre d’Investigation Clinique (CIC 1401), Bordeaux, France
| | - Pascal Barat
- CHU de Bordeaux, Centre d’Investigation Clinique (CIC 1401), Bordeaux, France
- Université de Bordeaux, Nutrition and Integrative Neurobiology, Bordeaux, France
| | - Jean-Christophe Helbling
- Université de Bordeaux, Nutrition and Integrative Neurobiology, Bordeaux, France
- INRA, UMR1286, Nutrition and Integrative Neurobiology, Bordeaux, France
| | - Fabienne Nacka
- CHU de Bordeaux, Centre d’Investigation Clinique (CIC 1401), Bordeaux, France
| | - Patrick Berger
- Université de Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Bordeaux, France
- CHU de Bordeaux, Centre d’Investigation Clinique (CIC 1401), Bordeaux, France
| | - Marie-Pierre Moisan
- Université de Bordeaux, Nutrition and Integrative Neurobiology, Bordeaux, France
- INRA, UMR1286, Nutrition and Integrative Neurobiology, Bordeaux, France
| | - Jean-Benoit Corcuff
- Université de Bordeaux, Nutrition and Integrative Neurobiology, Bordeaux, France
- INRA, UMR1286, Nutrition and Integrative Neurobiology, Bordeaux, France
| |
Collapse
|
48
|
Troy NM, Bosco A. Respiratory viral infections and host responses; insights from genomics. Respir Res 2016; 17:156. [PMID: 27871304 PMCID: PMC5117516 DOI: 10.1186/s12931-016-0474-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 11/10/2016] [Indexed: 01/23/2023] Open
Abstract
Respiratory viral infections are a leading cause of disease and mortality. The severity of these illnesses can vary markedly from mild or asymptomatic upper airway infections to severe wheezing, bronchiolitis or pneumonia. In this article, we review the viral sensing pathways and organizing principles that govern the innate immune response to infection. Then, we reconstruct the molecular networks that differentiate symptomatic from asymptomatic respiratory viral infections, and identify the underlying molecular drivers of these networks. Finally, we discuss unique aspects of the biology and pathogenesis of infections with respiratory syncytial virus, rhinovirus and influenza, drawing on insights from genomics.
Collapse
Affiliation(s)
- Niamh M Troy
- Telethon Kids Institute, The University of Western Australia, Subiaco, Australia
| | - Anthony Bosco
- Telethon Kids Institute, The University of Western Australia, Subiaco, Australia.
| |
Collapse
|
49
|
Looi K, Troy NM, Garratt LW, Iosifidis T, Bosco A, Buckley AG, Ling KM, Martinovich KM, Kicic-Starcevich E, Shaw NC, Sutanto EN, Zosky GR, Rigby PJ, Larcombe AN, Knight DA, Kicic A, Stick SM. Effect of human rhinovirus infection on airway epithelium tight junction protein disassembly and transepithelial permeability. Exp Lung Res 2016; 42:380-395. [PMID: 27726456 DOI: 10.1080/01902148.2016.1235237] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
RATIONALE No studies have assessed the effects of human rhinovirus (HRV) infection on epithelial tight junctions (TJs) and resultant barrier function. AIM OF THE STUDY To correlate viral infection with TJ disassembly, epithelial barrier integrity, and function. MATERIALS AND METHODS Human airway epithelial cells were infected with HRV minor serotype 1B (HRV-1B) at various 50% tissue culture infectivity doses (TCID50) over 72 hours. HRV replication was assessed by quantitative-polymerase chain reaction (qPCR) while cell viability and apoptosis were assessed by proliferation and apoptotic assays, respectively. Protein expression of claudin-1, occludin, and zonula occludens protein-1 (ZO-1) was assessed using In-Cell™ Western assays. Transepithelial permeability assays were performed to assess effects on barrier functionality. RT2 Profiler focused qPCR arrays and pathway analysis evaluating associations between human TJ and antiviral response were performed to identify potential interactions and pathways between genes of interests. RESULTS HRV-1B infection affected viability that was both time and TCID50 dependent. Significant increases in apoptosis and viral replication post-infection correlated with viral titer. Viral infection significantly decreased claudin-1 protein expression at the lower TCID50, while a significant decrease in all three TJ protein expressions occurred at higher TCID50. Decrease in protein expression was concomitant with significant increases in epithelial permeability of fluorescein isothiocynate labeled-dextran 4 and 20 kDa. Analysis of focused qPCR arrays demonstrated a significant decrease in ZO-1 gene expression. Furthermore, network analysis between human TJ and antiviral response genes revealed possible interactions and regulation of TJ genes via interleukin (IL)-15 in response to HRV-1B infection. CONCLUSION HRV-1B infection directly alters human airway epithelial TJ expression leading to increased epithelial permeability potentially via an antiviral response of IL-15.
Collapse
Affiliation(s)
- Kevin Looi
- a School of Paediatrics and Child Health , The University of Western Australia , Nedlands , Western Australia , Australia
| | - Niamh M Troy
- b Telethon Kids Institute, Centre for Health Research , The University of Western Australia , Crawley , Western Australia , Australia
| | - Luke W Garratt
- a School of Paediatrics and Child Health , The University of Western Australia , Nedlands , Western Australia , Australia.,b Telethon Kids Institute, Centre for Health Research , The University of Western Australia , Crawley , Western Australia , Australia
| | - Thomas Iosifidis
- a School of Paediatrics and Child Health , The University of Western Australia , Nedlands , Western Australia , Australia.,c Centre for Cell Therapy and Regenerative Medicine , School of Medicine and Pharmacology, The University of Western Australia , Nedlands , Western Australia , Australia
| | - Anthony Bosco
- b Telethon Kids Institute, Centre for Health Research , The University of Western Australia , Crawley , Western Australia , Australia
| | - Alysia G Buckley
- d Centre for Microscopy, Characterisation and Analysis , The University of Western Australia , Crawley , Western Australia , Australia
| | - Kak-Ming Ling
- b Telethon Kids Institute, Centre for Health Research , The University of Western Australia , Crawley , Western Australia , Australia
| | - Kelly M Martinovich
- b Telethon Kids Institute, Centre for Health Research , The University of Western Australia , Crawley , Western Australia , Australia
| | - Elizabeth Kicic-Starcevich
- b Telethon Kids Institute, Centre for Health Research , The University of Western Australia , Crawley , Western Australia , Australia
| | - Nicole C Shaw
- b Telethon Kids Institute, Centre for Health Research , The University of Western Australia , Crawley , Western Australia , Australia
| | - Erika N Sutanto
- b Telethon Kids Institute, Centre for Health Research , The University of Western Australia , Crawley , Western Australia , Australia.,e Department of Respiratory Medicine , Princess Margaret Hospital for Children , Perth , Western Australia , Australia
| | - Graeme R Zosky
- f School of Medicine, Faculty of Health , University of Tasmania , Hobart , Tasmania , Australia
| | - Paul J Rigby
- d Centre for Microscopy, Characterisation and Analysis , The University of Western Australia , Crawley , Western Australia , Australia
| | - Alexander N Larcombe
- b Telethon Kids Institute, Centre for Health Research , The University of Western Australia , Crawley , Western Australia , Australia
| | - Darryl A Knight
- g School of Biomedical Sciences and Pharmacy , University of Newcastle , Callaghan , New South Wales , Australia.,h Priority Research Centre for Asthma and Respiratory Disease , Hunter Medical Research Institute , Newcastle , New South Wales , Australia.,i Department of Anesthesiology , Pharmacology and Therapeutics, University of British Columbia , Vancouver , Canada
| | - Anthony Kicic
- a School of Paediatrics and Child Health , The University of Western Australia , Nedlands , Western Australia , Australia.,b Telethon Kids Institute, Centre for Health Research , The University of Western Australia , Crawley , Western Australia , Australia.,c Centre for Cell Therapy and Regenerative Medicine , School of Medicine and Pharmacology, The University of Western Australia , Nedlands , Western Australia , Australia.,e Department of Respiratory Medicine , Princess Margaret Hospital for Children , Perth , Western Australia , Australia
| | - Stephen M Stick
- a School of Paediatrics and Child Health , The University of Western Australia , Nedlands , Western Australia , Australia.,b Telethon Kids Institute, Centre for Health Research , The University of Western Australia , Crawley , Western Australia , Australia.,c Centre for Cell Therapy and Regenerative Medicine , School of Medicine and Pharmacology, The University of Western Australia , Nedlands , Western Australia , Australia.,e Department of Respiratory Medicine , Princess Margaret Hospital for Children , Perth , Western Australia , Australia
| |
Collapse
|
50
|
Abstract
This review highlights the important articles published in the area of asthma research from January 2015 to July 2016. In basic science, significant advances have been made in understanding the link between the innate immune response and type II acquired immune responses in asthma and the role of the airway epithelium. Novel information continues to emerge with regard to the pathogenesis and heterogeneity of severe asthma. There have been important translational clinical trials in the areas of childhood asthma, treatment of allergy to improve asthma outcomes, and improving drug delivery to optimize the management of asthma. In addition, there are increasing data concerning the application of biological agents to the management of severe asthma. This body of work discusses the most notable advances in the understanding and management of asthma.
Collapse
Affiliation(s)
- Su-Ling Loo
- Priority Research Centre for Healthy Lungs, University of Newcastle, Hunter Medical Research Institute, New Lambton, NSW, 2305, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, University of Newcastle, Hunter Medical Research Institute, New Lambton, NSW, 2305, Australia
| |
Collapse
|