1
|
Colomb F, McSorley HJ. Protein families secreted by nematodes to modulate host immunity. Curr Opin Microbiol 2025; 84:102582. [PMID: 39954371 DOI: 10.1016/j.mib.2025.102582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/14/2025] [Accepted: 01/24/2025] [Indexed: 02/17/2025]
Abstract
Parasitic nematodes release a wide variety of immunomodulatory proteins, which allow them to escape the host's immune-mediated killing or ejection mechanisms. This immunomodulation is mediated by nematode excretory/secretory (E/S) products, which contain multiple families of immunomodulatory proteins. Many of these families are conserved across different parasitic nematodes, while others are apparently unique to specific species. While some E/S products interact with host proteins, others have evolved to target host lipids, glycans, and metabolites. In this review, we will focus on three families of immunomodulatory proteins, which are particularly expanded in intestinal nematodes: the venom allergen-like proteins, the apyrases, and the complement control protein domain-containing proteins. These families of proteins suppress host immune responses, and evidence is gathering that these could be effective vaccine antigens against these intractable parasites.
Collapse
Affiliation(s)
- Florent Colomb
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland
| | - Henry J McSorley
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland.
| |
Collapse
|
2
|
Curren B, Ahmed T, Rashid RB, Sebina I, Al Amin Sikder M, Howard DR, Alorro M, Ullah MA, Bissell A, Rahman MM, Pearen MA, Ramm GA, Varelias A, Rose-John S, MacDonald KPA, Hoelzle R, Ó Cuív P, Spann KM, Dennis PG, Phipps S. A maternal high-fat diet predisposes to infant lung disease via increased neutrophil-mediated IL-6 trans-signaling. Cell Rep 2024; 43:114974. [PMID: 39535919 DOI: 10.1016/j.celrep.2024.114974] [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: 04/15/2024] [Revised: 09/24/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
A poor maternal diet during pregnancy predisposes the infant to severe lower respiratory tract infections (sLRIs), which, in turn, increases childhood asthma risk; however, the underlying mechanisms remain poorly understood. Here, we show that the offspring of high-fat diet (HFD)-fed mothers (HFD-reared pups) developed an sLRI following pneumovirus inoculation in early life and subsequent asthma in later life upon allergen exposure. Prior to infection, HFD-reared pups developed microbial dysbiosis and low-grade systemic inflammation (LGSI), characterized by hyperneutropoiesis in the liver and elevated inflammatory cytokine expression, most notably granulocyte-colony stimulating factor (G-CSF), interleukin-17A (IL-17A), IL-6 and soluble IL-6 receptor (sIL-6R) (indicative of IL-6 trans-signaling) in the circulation and multiple organs but most prominently the liver. Inhibition of IL-6 trans-signaling using sgp130Fc transgenic mice or via specific genetic deletion of IL-6Ra on neutrophils conferred protection against both diseases. Taken together, our findings suggest that a maternal HFD induces neonatal LGSI that predisposes to sLRI and subsequent asthma via neutrophil-mediated IL-6 trans-signaling.
Collapse
Affiliation(s)
- Bodie Curren
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD 4072, Australia
| | - Tufael Ahmed
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QLD 4000, Australia
| | - Ridwan B Rashid
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Ismail Sebina
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Md Al Amin Sikder
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD 4072, Australia
| | - Daniel R Howard
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD 4072, Australia
| | - Mariah Alorro
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Md Ashik Ullah
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Alec Bissell
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Muhammed Mahfuzur Rahman
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD 4072, Australia
| | - Michael A Pearen
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Grant A Ramm
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD 4072, Australia
| | - Antiopi Varelias
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QLD 4000, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, QLD 4072, Australia
| | - Stefan Rose-John
- Christian-Albrechts-Universität zu Kiel, Medical Faculty, Olshausenstraße 40, 24098 Kiel, Germany
| | - Kelli P A MacDonald
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD 4072, Australia
| | - Robert Hoelzle
- School of Environment, The University of Queensland, QLD 4072, Australia
| | - Páraic Ó Cuív
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QLD 4000, Australia
| | - Kirsten M Spann
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QLD 4000, Australia
| | - Paul G Dennis
- Australian Infectious Diseases Research Centre, The University of Queensland, QLD 4072, Australia; School of Environment, The University of Queensland, QLD 4072, Australia
| | - Simon Phipps
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD 4072, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QLD 4000, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, QLD 4072, Australia.
| |
Collapse
|
3
|
Ullah MA, Rittchen S, Li J, Curren BF, Namubiru P, Ahmed T, Howard DR, Rahman MM, Sikder MAA, Rashid RB, Collinson N, Lor M, Smythe ML, Phipps S. Dual therapy with corticosteroid ablates the beneficial effect of DP2 antagonism in chronic experimental asthma. Nat Commun 2024; 15:10253. [PMID: 39592603 PMCID: PMC11599388 DOI: 10.1038/s41467-024-54670-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 11/18/2024] [Indexed: 11/28/2024] Open
Abstract
Prostaglandin D2 (PGD2) signals via the DP1 and DP2 receptors. In Phase II trials, DP2 antagonism decreased airway inflammation and airway smooth muscle (ASM) area in moderate-to-severe asthma patients. However, in Phase III, DP2 antagonism failed to lower the rate of exacerbations, and DP2 as a target was shelved. Here, using a preclinical model of chronic experimental asthma, we demonstrate that rhinovirus-induced exacerbations increase PGD2 release, mucus production, transforming growth factor (TGF)-β1 and type-2 inflammation. DP2 antagonism or DP1 agonism ablates these phenotypes, increases epithelial EGF expression and decreases ASM area via increased IFN-γ. In contrast, dual DP1-DP2 antagonism or dual corticosteroid/DP2 antagonism, which attenuates endogenous PGD2, prevented ASM resolution. We demonstrate that DP2 antagonism resolves ASM remodelling via PGD2/DP1-mediated upregulation of IFN-γ expression, and that dual DP2 antagonism/corticosteroid therapy, as often occurred in the human trials, impairs the efficacy of DP2 antagonism by suppressing endogenous PGD2 and IFN-γ production.
Collapse
Affiliation(s)
- Md Ashik Ullah
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Sonja Rittchen
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, Graz, 8010, Austria
| | - Jia Li
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- Department of Laboratory Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20080, China
| | - Bodie F Curren
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Patricia Namubiru
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Tufael Ahmed
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Daniel R Howard
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Muhammed Mahfuzur Rahman
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Md Al Amin Sikder
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Ridwan B Rashid
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Natasha Collinson
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Mary Lor
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Mark L Smythe
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, 4072, Australia
| | - Simon Phipps
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia.
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, 4072, Australia.
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD, 4072, Australia.
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, 4000, Australia.
| |
Collapse
|
4
|
Radhouani M, Starkl P. Adjuvant-independent airway sensitization and infection mouse models leading to allergic asthma. FRONTIERS IN ALLERGY 2024; 5:1423938. [PMID: 39157265 PMCID: PMC11327155 DOI: 10.3389/falgy.2024.1423938] [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: 04/26/2024] [Accepted: 07/05/2024] [Indexed: 08/20/2024] Open
Abstract
Asthma is a chronic respiratory disease of global importance. Mouse models of allergic asthma have been instrumental in advancing research and novel therapeutic strategies for patients. The application of relevant allergens and physiological routes of exposure in such models has led to valuable insights into the complexities of asthma onset and development as well as key disease mechanisms. Furthermore, environmental microbial exposures and infections have been shown to play a fundamental part in asthma pathogenesis and alter disease outcome. In this review, we delve into physiological mouse models of allergic asthma and explore literature reports on most significant interplays between microbial infections and asthma development with relevance to human disease.
Collapse
Affiliation(s)
- Mariem Radhouani
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Philipp Starkl
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
5
|
Malinczak CA, Fonseca W, Hrycaj SM, Morris SB, Rasky AJ, Yagi K, Wellik DM, Ziegler SF, Zemans RL, Lukacs NW. Early-life pulmonary viral infection leads to long-term functional and lower airway structural changes in the lungs. Am J Physiol Lung Cell Mol Physiol 2024; 326:L280-L291. [PMID: 38290164 PMCID: PMC11281791 DOI: 10.1152/ajplung.00300.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/03/2024] [Accepted: 01/17/2024] [Indexed: 02/01/2024] Open
Abstract
Early-life respiratory virus infections have been correlated with enhanced development of childhood asthma. In particular, significant numbers of respiratory syncytial virus (RSV)-hospitalized infants go on to develop lung disease. It has been suggested that early-life viral infections may lead to altered lung development or repair that negatively impacts lung function later in life. Our data demonstrate that early-life RSV infection modifies lung structure, leading to decreased lung function. At 5 wk postneonatal RSV infection, significant defects are observed in baseline pulmonary function test (PFT) parameters consistent with decreased lung function as well as enlarged alveolar spaces. Lung function changes in the early-life RSV-infected group continue at 3 mo of age. The altered PFT and structural changes induced by early-life RSV were mitigated in TSLPR-/- mice that have previously been shown to have reduced immune cell accumulation associated with a persistent Th2 environment. Importantly, long-term effects were demonstrated using a secondary RSV infection 3 mo following the initial early-life RSV infection and led to significant additional defects in lung function, with severe mucus deposition within the airways, and consolidation of the alveolar spaces. These studies suggest that early-life respiratory viral infection leads to alterations in lung structure/repair that predispose to diminished lung function later in life.NEW & NOTEWORTHY These studies outline a novel finding that early-life respiratory virus infection can alter lung structure and function long-term. Importantly, the data also indicate that there are critical links between inflammatory responses and subsequent events that produce a more severe pathogenic response later in life. The findings provide additional data to support that early-life infections during lung development can alter the trajectory of airway function.
Collapse
Affiliation(s)
| | - Wendy Fonseca
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States
| | - Steven M Hrycaj
- Department of Internal Medicine, Pulmonary, University of Michigan, Ann Arbor, Michigan, United States
| | - Susan B Morris
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States
| | - Andrew J Rasky
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States
| | - Kazuma Yagi
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States
| | - Deneen M Wellik
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, Wisconsin, United States
| | - Steven F Ziegler
- Immunology Program, Benaroya Research Institute, Seattle, Washington, United States
| | - Rachel L Zemans
- Department of Internal Medicine, Pulmonary, University of Michigan, Ann Arbor, Michigan, United States
| | - Nicholas W Lukacs
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, United States
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
6
|
Lameire S, Hammad H. Lung epithelial cells: Upstream targets in type 2-high asthma. Eur J Immunol 2023; 53:e2250106. [PMID: 36781404 DOI: 10.1002/eji.202250106] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023]
Abstract
Over the last years, technological advances in the field of asthma have led to the identification of two disease endotypes, namely, type 2-high and type 2-low asthma, characterized by different pathophysiologic mechanisms at a cellular and molecular level. Although specific immune cells are important contributors to each of the recognized asthma endotype, the lung epithelium is now regarded as a crucial player able to orchestrate responses to inhaled environmental triggers such as allergens and microbes. The impact of the epithelium goes beyond its physical barrier. It is nowadays considered as a part of the innate immune system that can actively respond to insults. Activated epithelial cells, by producing a specific set of cytokines, trigger innate and adaptive immune cells to cause pathology. Here, we review how the epithelium contributes to the development of Th2 sensitization to allergens and asthma with a "type 2-high" signature, in both murine models and human studies of this asthma endotype. We also discuss epithelial responses to respiratory viruses, such as rhinovirus, respiratory syncytial virus, and SARS-CoV-2, and how these triggers influence not only asthma development but also asthma exacerbation. Finally, we also summarize the results of promising clinical trials using biologicals targeting epithelial-derived cytokines in asthmatic patients.
Collapse
Affiliation(s)
- Sahine Lameire
- Laboratory of Mucosal Immunology and Immunoregulation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Hamida Hammad
- Laboratory of Mucosal Immunology and Immunoregulation, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| |
Collapse
|
7
|
Curren B, Ahmed T, Howard DR, Ashik Ullah M, Sebina I, Rashid RB, Al Amin Sikder M, Namubiru P, Bissell A, Ngo S, Jackson DJ, Toussaint M, Edwards MR, Johnston SL, McSorley HJ, Phipps S. IL-33-induced neutrophilic inflammation and NETosis underlie rhinovirus-triggered exacerbations of asthma. Mucosal Immunol 2023; 16:671-684. [PMID: 37506849 DOI: 10.1016/j.mucimm.2023.07.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 06/04/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023]
Abstract
Rhinovirus-induced neutrophil extracellular traps (NETs) contribute to acute asthma exacerbations; however, the molecular factors that trigger NETosis in this context remain ill-defined. Here, we sought to implicate a role for IL-33, an epithelial cell-derived alarmin rapidly released in response to infection. In mice with chronic experimental asthma (CEA), but not naïve controls, rhinovirus inoculation induced an early (1 day post infection; dpi) inflammatory response dominated by neutrophils, neutrophil-associated cytokines (IL-1α, IL-1β, CXCL1), and NETosis, followed by a later, type-2 inflammatory phase (3-7 dpi), characterised by eosinophils, elevated IL-4 levels, and goblet cell hyperplasia. Notably, both phases were ablated by HpARI (Heligmosomoides polygyrus Alarmin Release Inhibitor), which blocks IL-33 release and signalling. Instillation of exogenous IL-33 recapitulated the rhinovirus-induced early phase, including the increased presence of NETs in the airway mucosa, in a PAD4-dependent manner. Ex vivo IL-33-stimulated neutrophils from mice with CEA, but not naïve mice, underwent NETosis and produced greater amounts of IL-1α/β, IL-4, and IL-5. In nasal samples from rhinovirus-infected people with asthma, but not healthy controls, IL-33 levels correlated with neutrophil elastase and dsDNA. Our findings suggest that IL-33 blockade ameliorates the severity of an asthma exacerbation by attenuating neutrophil recruitment and the downstream generation of NETs.
Collapse
Affiliation(s)
- Bodie Curren
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; School of Biomedical Sciences, The University of Queensland, Queensland 4072, Australia
| | - Tufael Ahmed
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; School of Biomedical Sciences, Queensland University of Technology, Queensland 4000, Australia
| | - Daniel R Howard
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; School of Biomedical Sciences, The University of Queensland, Queensland 4072, Australia
| | - Md Ashik Ullah
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
| | - Ismail Sebina
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; School of Biomedical Sciences, The University of Queensland, Queensland 4072, Australia; School of Biomedical Sciences, Queensland University of Technology, Queensland 4000, Australia
| | - Ridwan B Rashid
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; School of Biomedical Sciences, The University of Queensland, Queensland 4072, Australia
| | - Md Al Amin Sikder
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; School of Biomedical Sciences, The University of Queensland, Queensland 4072, Australia
| | - Patricia Namubiru
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; School of Biomedical Sciences, The University of Queensland, Queensland 4072, Australia
| | - Alec Bissell
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
| | - Sylvia Ngo
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
| | - David J Jackson
- School of Immunology & Microbial Sciences, King's College London, London, UK; National Heart and Lung Institute, Imperial College London, London, UK
| | - Marie Toussaint
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Michael R Edwards
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Henry J McSorley
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Simon Phipps
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia; School of Biomedical Sciences, The University of Queensland, Queensland 4072, Australia; School of Biomedical Sciences, Queensland University of Technology, Queensland 4000, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, 4072 Queensland, Australia.
| |
Collapse
|
8
|
Spector C, De Sanctis CM, Panettieri RA, Koziol-White CJ. Rhinovirus induces airway remodeling: what are the physiological consequences? Respir Res 2023; 24:238. [PMID: 37773065 PMCID: PMC10540383 DOI: 10.1186/s12931-023-02529-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/01/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Rhinovirus infections commonly evoke asthma exacerbations in children and adults. Recurrent asthma exacerbations are associated with injury-repair responses in the airways that collectively contribute to airway remodeling. The physiological consequences of airway remodeling can manifest as irreversible airway obstruction and diminished responsiveness to bronchodilators. Structural cells of the airway, including epithelial cells, smooth muscle, fibroblasts, myofibroblasts, and adjacent lung vascular endothelial cells represent an understudied and emerging source of cellular and extracellular soluble mediators and matrix components that contribute to airway remodeling in a rhinovirus-evoked inflammatory environment. MAIN BODY While mechanistic pathways associated with rhinovirus-induced airway remodeling are still not fully characterized, infected airway epithelial cells robustly produce type 2 cytokines and chemokines, as well as pro-angiogenic and fibroblast activating factors that act in a paracrine manner on neighboring airway cells to stimulate remodeling responses. Morphological transformation of structural cells in response to rhinovirus promotes remodeling phenotypes including induction of mucus hypersecretion, epithelial-to-mesenchymal transition, and fibroblast-to-myofibroblast transdifferentiation. Rhinovirus exposure elicits airway hyperresponsiveness contributing to irreversible airway obstruction. This obstruction can occur as a consequence of sub-epithelial thickening mediated by smooth muscle migration and myofibroblast activity, or through independent mechanisms mediated by modulation of the β2 agonist receptor activation and its responsiveness to bronchodilators. Differential cellular responses emerge in response to rhinovirus infection that predispose asthmatic individuals to persistent signatures of airway remodeling, including exaggerated type 2 inflammation, enhanced extracellular matrix deposition, and robust production of pro-angiogenic mediators. CONCLUSIONS Few therapies address symptoms of rhinovirus-induced airway remodeling, though understanding the contribution of structural cells to these processes may elucidate future translational targets to alleviate symptoms of rhinovirus-induced exacerbations.
Collapse
Affiliation(s)
- Cassandra Spector
- Rutgers Institute for Translation Medicine and Science, New Brunswick, NJ, USA
| | - Camden M De Sanctis
- Rutgers Institute for Translation Medicine and Science, New Brunswick, NJ, USA
| | | | | |
Collapse
|
9
|
Dounce-Cuevas CA, Flores-Flores A, Bazán MS, Portales-Rivera V, Morelos-Ulíbarri AA, Bazán-Perkins B. Asthma and COVID-19: a controversial relationship. Virol J 2023; 20:207. [PMID: 37679779 PMCID: PMC10485988 DOI: 10.1186/s12985-023-02174-0] [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: 06/09/2023] [Accepted: 08/31/2023] [Indexed: 09/09/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection induces a spectrum of clinical manifestations that depend on the immune response of the patient, i.e., from an asymptomatic form to an inflammatory response with multiorgan deterioration. In some cases, severe cases of SARS-CoV-2 are characterized by an excessive, persistent release of inflammatory mediators known as a cytokine storm. This phenomenon arises from an ineffective T helper (Th)-1 response, which is unable to control the infection and leads to a reinforcement of innate immunity, causing tissue damage. The evolution of the disease produced by SARS-CoV2, known as COVID-19, has been of interest in several research fields. Asthma patients have been reported to present highly variable outcomes due to the heterogeneity of the disease. For example, the Th2 response in patients with allergic asthma is capable of decreasing Th1 activation in COVID-19, preventing the onset of a cytokine storm; additionally, IL-33 released by damaged epithelium in the context of COVID-19 potentiates either Th1 or T2-high responses, a process that contributes to poor outcomes. IL-13, a T2-high inflammatory cytokine, decreases the expression of angiotensin converting enzyme-2 (ACE2) receptor, hindering SARS-CoV-2 entry; finally, poor outcomes have been observed in COVID-19 patients with severe neutrophilic asthma. In other contexts, the COVID-19 lockdown has had interesting effects on asthma epidemiology. The incidence of asthma in the most populated states in Mexico, including Tamaulipas, which has the highest asthma incidence in the country, showed similar tendencies independent of how strict the lockdown measures were in each state. As described worldwide for various diseases, a decrease in asthma cases was observed during the COVID-19 lockdown. This decrease was associated with a drop in acute respiratory infection cases. The drop in cases of various diseases, such as diabetes, hypertension or depression, observed in 2020 was restored in 2022, but not for asthma and acute respiratory infections. There were slight increases in asthma cases when in-person classes resumed. In conclusion, although many factors were involved in asthma outcomes during the pandemic, it seems that acute respiratory infection is intimately linked to asthma cases. Social distancing during remote learning, particularly school lockdown, appears to be an important cause of the decrease in cases.
Collapse
Affiliation(s)
- Carlos A Dounce-Cuevas
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, 14380, Mexico City, Mexico
| | - Angélica Flores-Flores
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, 14380, Mexico City, Mexico
- Laboratorio de Inmunofarmacología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Mexico City, Mexico
| | - Mariana S Bazán
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, 14380, Mexico City, Mexico
| | - Victor Portales-Rivera
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, 14380, Mexico City, Mexico
| | | | - Blanca Bazán-Perkins
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, 14380, Mexico City, Mexico.
- Laboratorio de Inmunofarmacología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Mexico City, Mexico.
| |
Collapse
|
10
|
Urbani F, Cometa M, Martelli C, Santoli F, Rana R, Ursitti A, Bonato M, Baraldo S, Contoli M, Papi A. Update on virus-induced asthma exacerbations. Expert Rev Clin Immunol 2023; 19:1259-1272. [PMID: 37470413 DOI: 10.1080/1744666x.2023.2239504] [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: 03/07/2023] [Revised: 06/01/2023] [Accepted: 07/18/2023] [Indexed: 07/21/2023]
Abstract
INTRODUCTION Viral infections are common triggers for asthma exacerbation. Subjects with asthma are more susceptible to viral infections and develop more severe or long-lasting lower respiratory tract symptoms than healthy individuals owing to impaired immune responses. Of the many viruses associated with asthma exacerbation, rhinovirus (RV) is the most frequently identified virus in both adults and children. AREAS COVERED We reviewed epidemiological and clinical links and mechanistic studies on virus-associated asthma exacerbations. We included sections on severe acute respiratory syndrome coronavirus 2 (SARS-CoV2), the latest evidence of coronavirus disease 2019 (COVID-19) in asthma patients, and past and future searches for therapeutic and prevention targets. EXPERT OPINION Early treatment or prevention of viral infections might significantly reduce the rate of asthma exacerbation, which is one of the key points of disease management. Although it is hypothetically possible nowadays to interfere with every step of the infectious cycle of respiratory tract viruses, vaccination development has provided some of the most encouraging results. Future research should proceed toward the development of a wider spectrum of vaccines to achieve a better quality of life for patients with asthma and to reduce the economic burden on the healthcare system.
Collapse
Affiliation(s)
- Francesca Urbani
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Marianna Cometa
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Chiara Martelli
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Federica Santoli
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Roberto Rana
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Antonio Ursitti
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Matteo Bonato
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Simonetta Baraldo
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Padova, Italy
| | - Marco Contoli
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| | - Alberto Papi
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'anna University Hospital, Ferrara, Italy
| |
Collapse
|
11
|
Allam VSRR, Waern I, Taha S, Akula S, Wernersson S, Pejler G. Nafamostat has anti-asthmatic effects associated with suppressed pro-inflammatory gene expression, eosinophil infiltration and airway hyperreactivity. Front Immunol 2023; 14:1136780. [PMID: 37153590 PMCID: PMC10160450 DOI: 10.3389/fimmu.2023.1136780] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 04/07/2023] [Indexed: 05/09/2023] Open
Abstract
Introduction Asthma is characterized by an imbalance between proteases and their inhibitors. Hence, an attractive therapeutic option could be to interfere with asthma-associated proteases. Here we exploited this option by assessing the impact of nafamostat, a serine protease inhibitor known to neutralize mast cell tryptase. Methods Nafamostat was administered in a mouse model for asthma based on sensitization by house dust mite (HDM) extract, followed by the assessment of effects on airway hyperreactivity, inflammatory parameters and gene expression. Results We show that nafamostat efficiently suppressed the airway hyperreactivity in HDM-sensitized mice. This was accompanied by reduced infiltration of eosinophils and lymphocytes to the airways, and by lower levels of pro-inflammatory compounds within the airway lumen. Further, nafamostat had a dampening impact on goblet cell hyperplasia and smooth muscle layer thickening in the lungs of HDM-sensitized animals. To obtain deeper insight into the underlying mechanisms, a transcriptomic analysis was conducted. This revealed, as expected, that the HDM sensitization caused an upregulated expression of numerous pro-inflammatory genes. Further, the transcriptomic analysis showed that nafamostat suppressed the levels of multiple pro-inflammatory genes, with a particular impact on genes related to asthma. Discussion Taken together, this study provides extensive insight into the ameliorating effect of nafamostat on experimental asthma, and our findings can thereby provide a basis for the further evaluation of nafamostat as a potential therapeutic agent in human asthma.
Collapse
Affiliation(s)
- Venkata Sita Rama Raju Allam
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Ida Waern
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Sowsan Taha
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Srinivas Akula
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Sara Wernersson
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
- *Correspondence: Sara Wernersson, ; Gunnar Pejler,
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- *Correspondence: Sara Wernersson, ; Gunnar Pejler,
| |
Collapse
|
12
|
Bryant N, Muehling LM. T-cell responses in asthma exacerbations. Ann Allergy Asthma Immunol 2022; 129:709-718. [PMID: 35918022 PMCID: PMC9987567 DOI: 10.1016/j.anai.2022.07.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/22/2022] [Accepted: 07/22/2022] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Asthma is a chronic lung disease comprising multiple endotypes and characterized by periodic exacerbations. A diverse array of T cells has been found to contribute to all endotypes of asthma in pathogenic and regulatory roles. Here, we review the contributions of CD4+, CD8+, and unconventional T cells in allergic and nonallergic asthma. DATA SOURCES Review of published literature pertaining to conventional and unconventional T-cell types in asthma. STUDY SELECTIONS Recent peer-reviewed articles pertaining to T cells in asthma, with additional peer-reviewed studies for context. RESULTS Much research in asthma has focused on the roles of CD4+ TH cells. Roles for TH2 cells in promoting allergic asthma pathogenesis have been well-described, and the recent description of pathogenic TH2A cells provides additional insight into these responses. Other TH types, notably TH1 and TH17, have been linked to neutrophilic and steroid-resistant asthma phenotypes. Beyond CD4+ T cells, CD8+ Tc2 cells are also strongly associated with allergic asthma. An emerging area for study is unconventional T-cell types, including γδT, invariant natural killer T, and mucosal-associated invariant T cells. Although data in asthma remain limited for these cells, their ability to bridge innate and adaptive responses likely makes them key players in asthma. A number of asthma therapies target T-cell responses, and, although data are limited, they seem to modulate T-cell populations. CONCLUSION Given the diversity and heterogeneity of asthma and T-cell responses, there remain many rich avenues for research to better understand the pathogenesis of asthma. Despite the breadth of T cells in asthma, approved therapeutics remain limited to TH2 networks.
Collapse
Affiliation(s)
- Naomi Bryant
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Lyndsey M Muehling
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia.
| |
Collapse
|
13
|
Li L, Fuller SJ. Inhibiting the P2Y 13 receptor reduces IL-33 and HMGB1 lung concentrations and inflammatory cell infiltration in a murine model of asthma. Purinergic Signal 2022; 18:403-405. [PMID: 35294701 PMCID: PMC9832198 DOI: 10.1007/s11302-022-09859-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/09/2022] [Indexed: 01/15/2023] Open
Affiliation(s)
- Lanxin Li
- Department of Medicine, Sydney Medical School Nepean, University of Sydney, Nepean Hospital, Penrith, NSW 2750 Australia
| | - Stephen J. Fuller
- Department of Medicine, Sydney Medical School Nepean, University of Sydney, Nepean Hospital, Penrith, NSW 2750 Australia
| |
Collapse
|
14
|
Emerging Effects of IL-33 on COVID-19. Int J Mol Sci 2022; 23:ijms232113656. [PMID: 36362440 PMCID: PMC9658128 DOI: 10.3390/ijms232113656] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/27/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Since the start of COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), more than 6 million people have lost their lives worldwide directly or indirectly. Despite intensified efforts to clarify the immunopathology of COVID-19, the key factors and processes that trigger an inflammatory storm and lead to severe clinical outcomes in patients remain unclear. As an inflammatory storm factor, IL-33 is an alarmin cytokine, which plays an important role in cell damage or infection. Recent studies have shown that serum IL-33 is upregulated in COVID-19 patients and is strongly associated with poor outcomes. Increased IL-33 levels in severe infections may result from an inflammatory storm caused by strong interactions between activated immune cells. However, the effects of IL-33 in COVID-19 and the underlying mechanisms remain to be fully elucidated. In this review, we systematically discuss the biological properties of IL-33 under pathophysiological conditions and its regulation of immune cells, including neutrophils, innate lymphocytes (ILCs), dendritic cells, macrophages, CD4+ T cells, Th17/Treg cells, and CD8+ T cells, in COVID-19 phagocytosis. The aim of this review is to explore the potential value of the IL-33/immune cell pathway as a new target for early diagnosis, monitoring of severe cases, and clinical treatment of COVID-19.
Collapse
|
15
|
Phipps S, Howard DR, Werder RB. Eosinophils apply a handbrake to plasmacytoid dendritic cell antiviral immunity in asthma. J Allergy Clin Immunol 2022; 150:589-591. [PMID: 35917933 DOI: 10.1016/j.jaci.2022.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/12/2022] [Indexed: 11/18/2022]
Affiliation(s)
- Simon Phipps
- Queensland Institute of Medical Research Berghofer Medical Research Institute, Herston, Australia; School of Biomedical Sciences, The University of Queensland, Herston, Australia; School of Biomedical Sciences, Queensland University of Technology, Herston, Australia.
| | - Daniel Ryan Howard
- Queensland Institute of Medical Research Berghofer Medical Research Institute, Herston, Australia; School of Biomedical Sciences, The University of Queensland, Herston, Australia
| | - Rhiannon Bree Werder
- Queensland Institute of Medical Research Berghofer Medical Research Institute, Herston, Australia; Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, Mass; Pulmonary Center, Boston University School of Medicine, Boston, Mass
| |
Collapse
|
16
|
Du X, Yuan L, Yao Y, Yang Y, Zhou K, Wu X, Wang L, Qin L, Li W, Xiang Y, Qu X, Liu H, Qin X, Yang M, Liu C. ITGB4 Deficiency in Airway Epithelium Aggravates RSV Infection and Increases HDM Sensitivity. Front Immunol 2022; 13:912095. [PMID: 35958591 PMCID: PMC9357881 DOI: 10.3389/fimmu.2022.912095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/13/2022] [Indexed: 11/16/2022] Open
Abstract
Background The heterogeneity of RSV-infected pathology phenotype in early life is strongly associate with increased susceptibility of asthma in later life. However, the inner mechanism of this heterogeneity is still obscure. ITGB4 is a down-regulated adhesion molecular in the airway epithelia of asthma patients which may participate in the regulation of RSV infection related intracellular pathways. Object This study was designed to observe the involvement of ITGB4 in the process of RSV infection and the effect of ITGB4 deficiency on anti-RSV responses of airway epithelia. Results RSV infection caused a transient decrease of ITGB4 expression both in vitro and in vivo. Besides, ITGB4 deficiency induced not only exacerbated RSV infection, but also enhanced HDM sensitivity in later life. Moreover, IFN III (IFN-λ) was significantly suppressed during RSV infection in ITGB4 deficient airway epithelial cells. Furthermore, the suppression of IFN-λ were regulated by IRF-1 through the phosphorylation of EGFR in airway epithelial cells after RSV infection. Conclusion These results demonstrated the involvement of ITGB4 deficiency in the development of enhance RSV infection in early life and the increased HDM sensitivity in later life by down-regulation of IFN-λ through EGFR/IRF-1 pathway in airway epithelial cells.
Collapse
Affiliation(s)
- Xizi Du
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Lin Yuan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Ye Yao
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Yu Yang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Kai Zhou
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Xinyu Wu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Leyuan Wang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Ling Qin
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
| | - Wenkai Li
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| | - Yang Xiang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Xiangping Qu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Huijun Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Xiaoqun Qin
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Ming Yang
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI), University of Newcastle, New Lambton Heights, NSW, Australia
| | - Chi Liu
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
- Research Center of China-Africa Infectious Diseases, Xiangya School of Medicine Central South University, Changsha, China
- *Correspondence: Chi Liu,
| |
Collapse
|
17
|
Schaunaman N, Dimasuay KG, Cervantes D, Li L, Numata M, Kraft M, Chu HW. Tollip Inhibits IL-33 Release and Inflammation in Influenza A Virus-Infected Mouse Airways. J Innate Immun 2022; 15:67-77. [PMID: 35760043 PMCID: PMC10643888 DOI: 10.1159/000525315] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 05/25/2022] [Indexed: 11/19/2022] Open
Abstract
Respiratory influenza A virus (IAV) infection continues to pose significant challenges in healthcare of human diseases including asthma. IAV infection in mice was shown to increase IL-33, a key cytokine in driving airway inflammation in asthma, but how IL-33 is regulated during viral infection remains unclear. We previously found that a genetic mutation in Toll-interacting protein (Tollip) was linked to less airway epithelial Tollip expression, increased neutrophil chemokines, and lower lung function in asthma patients. As Tollip is involved in maintaining mitochondrial function, and mitochondrial stress may contribute to extracellular ATP release and IL-33 secretion, we hypothesized that Tollip downregulates IL-33 secretion via inhibiting ATP release during IAV infection. Wild-type and Tollip knockout (KO) mice were infected with IAV and treated with either an ATP converter apyrase or an IL-33 decoy receptor soluble ST2 (sST2). KO mice significantly lost more body weight and had increased extracellular ATP, IL-33 release, and neutrophilic inflammation. Apyrase treatment reduced extracellular ATP levels, IL-33 release, and neutrophilic inflammation in Tollip KO mice. Excessive lung neutrophilic inflammation in IAV-infected Tollip KO mice was reduced by sST2, which was coupled with less IL-33 release. Our data suggest that Tollip inhibits IAV infection, potentially by inhibiting extracellular ATP release and reducing IL-33 activation and lung inflammation. In addition, sST2 may serve as a potential therapeutic approach to mitigate respiratory viral infection in human subjects with Tollip deficiency.
Collapse
Affiliation(s)
| | | | - Diana Cervantes
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Liwu Li
- Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Mari Numata
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - Monica Kraft
- Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona, USA
| | - Hong Wei Chu
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| |
Collapse
|
18
|
Abstract
A principal purpose of type 2 immunity was thought to be defense against large parasites, but it also functions in the restoration of homeostasis, such as toxin clearance following snake bites. In other cases, like allergy, the type 2 T helper (Th2) cytokines and cells present in the environment are detrimental and cause diseases. In recent years, the recognition of cell heterogeneity within Th2-associated cell populations has revealed specific functions of cells with a particular phenotype or gene signature. In addition, here we discuss the recent data regarding heterogeneity of type 2 immunity-related cells, as well as their newly identified role in a variety of processes ranging from involvement in respiratory viral infections [especially in the context of the recent COVID-19 (coronavirus disease 2019) pandemic] to control of cancer development or of metabolic homeostasis.
Collapse
Affiliation(s)
- Hamida Hammad
- Laboratory of Mucosal Immunology and Immunoregulation, VIB-UGent Center for Inflammation Research, Ghent, Belgium; .,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Nincy Debeuf
- Laboratory of Mucosal Immunology and Immunoregulation, VIB-UGent Center for Inflammation Research, Ghent, Belgium; .,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Helena Aegerter
- Laboratory of Mucosal Immunology and Immunoregulation, VIB-UGent Center for Inflammation Research, Ghent, Belgium; .,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Andrew S Brown
- Laboratory of Mucosal Immunology and Immunoregulation, VIB-UGent Center for Inflammation Research, Ghent, Belgium; .,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Mucosal Immunology and Immunoregulation, VIB-UGent Center for Inflammation Research, Ghent, Belgium; .,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
19
|
Eddens T, Parks OB, Williams JV. Neonatal Immune Responses to Respiratory Viruses. Front Immunol 2022; 13:863149. [PMID: 35493465 PMCID: PMC9047724 DOI: 10.3389/fimmu.2022.863149] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/23/2022] [Indexed: 11/30/2022] Open
Abstract
Respiratory tract infections are a leading cause of morbidity and mortality in newborns, infants, and young children. These early life infections present a formidable immunologic challenge with a number of possibly conflicting goals: simultaneously eliminate the acute pathogen, preserve the primary gas-exchange function of the lung parenchyma in a developing lung, and limit long-term sequelae of both the infection and the inflammatory response. The latter has been most well studied in the context of childhood asthma, where multiple epidemiologic studies have linked early life viral infection with subsequent bronchospasm. This review will focus on the clinical relevance of respiratory syncytial virus (RSV), human metapneumovirus (HMPV), and rhinovirus (RV) and examine the protective and pathogenic host responses within the neonate.
Collapse
Affiliation(s)
- Taylor Eddens
- Pediatric Scientist Development Program, University of Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
- Division of Allergy/Immunology, University of Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| | - Olivia B. Parks
- Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA, United States
| | - John V. Williams
- Division of Pediatric Infectious Diseases, University of Pittsburgh Medical Center (UPMC) Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
20
|
Cameron A, Dhariwal J, Upton N, Ranz Jimenez I, Paulsen M, Wong E, Trujillo‐Torralbo M, del Rosario A, Jackson DJ, Edwards MR, Johnston SL, Walton RP. Type I conventional dendritic cells relate to disease severity in virus-induced asthma exacerbations. Clin Exp Allergy 2022; 52:550-560. [PMID: 35212067 PMCID: PMC9310571 DOI: 10.1111/cea.14116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/31/2022] [Accepted: 02/10/2022] [Indexed: 01/07/2023]
Abstract
RATIONALE Rhinoviruses are the major precipitant of asthma exacerbations and individuals with asthma experience more severe/prolonged rhinovirus infections. Concurrent viral infection and allergen exposure synergistically increase exacerbation risk. Although dendritic cells orchestrate immune responses to both virus and allergen, little is known about their role in viral asthma exacerbations. OBJECTIVES To characterize dendritic cell populations present in the lower airways, and to assess whether their numbers are altered in asthma compared to healthy subjects prior to infection and during rhinovirus-16 infection. METHODS Moderately-severe atopic asthmatic patients and healthy controls were experimentally infected with rhinovirus-16. Bronchoalveolar lavage was collected at baseline, day 3 and day 8 post infection and dendritic cells isolated using fluorescence activated cell sorting. MEASUREMENTS AND MAIN RESULTS Numbers of type I conventional dendritic cells, which cross prime CD8+ T helper cells and produce innate interferons, were significantly reduced in the lower airways of asthma patients compared to healthy controls at baseline. This reduction was associated serum IgE at baseline and with reduced numbers of CD8+ T helper cells and with increased viral replication, airway eosinophils and reduced lung function during infection. IgE receptor expression on lower airway plasmacytoid dendritic cells was significantly increased in asthma, consistent with a reduced capacity to produce innate interferons. CONCLUSIONS Reduced numbers of anti-viral type I conventional dendritic cells in asthma are associated with adverse outcomes during rhinovirus infection. This, with increased FcεR1α expression on lower airway plasmacytoid DCs could mediate the more permissive respiratory viral infection observed in asthma patients.
Collapse
Affiliation(s)
- Aoife Cameron
- National Heart and Lung InstituteLondonUK,MRC Asthma UK Centre in Allergic Mechanisms of AsthmaLondonUK
| | - Jaideep Dhariwal
- National Heart and Lung InstituteLondonUK,MRC Asthma UK Centre in Allergic Mechanisms of AsthmaLondonUK
| | - Nadine Upton
- MRC Asthma UK Centre in Allergic Mechanisms of AsthmaLondonUK,School of Immunology & Microbial SciencesKing’s College LondonLondonUK
| | - Ismael Ranz Jimenez
- MRC Asthma UK Centre in Allergic Mechanisms of AsthmaLondonUK,School of Immunology & Microbial SciencesKing’s College LondonLondonUK
| | - Malte Paulsen
- St. Mary’s Flow Cytometry Core FacilityLondonUK,Novo Nordisk Foundation Center for Stem Cell MedicineFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Ernie Wong
- National Heart and Lung InstituteLondonUK,MRC Asthma UK Centre in Allergic Mechanisms of AsthmaLondonUK
| | | | - Ajerico del Rosario
- National Heart and Lung InstituteLondonUK,MRC Asthma UK Centre in Allergic Mechanisms of AsthmaLondonUK
| | - David J. Jackson
- MRC Asthma UK Centre in Allergic Mechanisms of AsthmaLondonUK,School of Immunology & Microbial SciencesKing’s College LondonLondonUK,Guy's and St Thomas’ NHS TrustLondonUK
| | - Michael R. Edwards
- National Heart and Lung InstituteLondonUK,MRC Asthma UK Centre in Allergic Mechanisms of AsthmaLondonUK
| | - Sebastian L. Johnston
- National Heart and Lung InstituteLondonUK,MRC Asthma UK Centre in Allergic Mechanisms of AsthmaLondonUK
| | - Ross P. Walton
- National Heart and Lung InstituteLondonUK,MRC Asthma UK Centre in Allergic Mechanisms of AsthmaLondonUK
| | | |
Collapse
|
21
|
Hazan G, Fox C, Eiden E, Anderson N, Friger M, Haspel J. Effect of the COVID-19 Lockdown on Asthma Biological Rhythms. J Biol Rhythms 2022; 37:152-163. [PMID: 35319293 DOI: 10.1177/07487304221081730] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Asthma has a striking temporal character, in which time-of-day, patient age, and season each influence disease activity. The extent to which rhythms in asthma activity reflect exposure to specific disease triggers remains unclear. In this study, we examined how virus mitigation strategies enacted during the COVID-19 pandemic ("lockdown measures") affected rhythms in asthma clinical activity in children. To this end, we retrospectively analyzed asthma clinical presentations in children aged <18 years to our regional academic medical center, comparing 4 years of medical records prior to COVID-19 lockdown measures with the 12 months immediately after the institution of such measures. We correlated these data to positive viral test results, febrile seizures, and allergic clinical surrogates (allergic reaction visits and Emergency Department [ED] antihistamine prescriptions, respectively) over the same time frame. In the 12 months following the institution of the COVID-19 lockdown, positivity rates for common respiratory viruses dropped by 70.2% and ED visits for asthma among children dropped by 62% compared to pre-COVID years. Lockdown suppressed seasonal variation in positive viral tests and asthma ED visits, while diurnal rhythms in asthma visits were unchanged. Asthma seasonality correlated most strongly with rhinovirus positivity both before and after the institution of COVID lockdown measures. Altogether, our data support a causal role for viruses in driving seasonal variability in asthma exacerbations in children.
Collapse
Affiliation(s)
- Guy Hazan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA.,Division of Pediatric Allergy and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Carolyn Fox
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Elise Eiden
- Institute for Informatics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Neil Anderson
- Division of Laboratory and Genomic Medicine, Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael Friger
- Department of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Jeffrey Haspel
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
22
|
Manna S, McAuley J, Jacobson J, Nguyen CD, Ullah MA, Sebina I, Williamson V, Mulholland EK, Wijburg O, Phipps S, Satzke C. Synergism and Antagonism of Bacterial-Viral Coinfection in the Upper Respiratory Tract. mSphere 2022; 7:e0098421. [PMID: 35044807 PMCID: PMC8769199 DOI: 10.1128/msphere.00984-21] [Citation(s) in RCA: 18] [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: 12/17/2021] [Accepted: 12/28/2021] [Indexed: 01/03/2023] Open
Abstract
Streptococcus pneumoniae (the pneumococcus) is a leading cause of pneumonia in children under 5 years of age. Coinfection by pneumococci and respiratory viruses enhances disease severity. Little is known about pneumococcal coinfections with respiratory syncytial virus (RSV). Here, we developed a novel infant mouse model of coinfection using pneumonia virus of mice (PVM), a murine analogue of RSV, to examine the dynamics of coinfection in the upper respiratory tract, an anatomical niche that is essential for host-to-host transmission and progression to disease. Coinfection increased damage to the nasal tissue and increased production of the chemokine CCL3. Nasopharyngeal pneumococcal density and shedding in nasal secretions were increased by coinfection. In contrast, coinfection reduced PVM loads in the nasopharynx, an effect that was independent of pneumococcal strain and the order of infection. We showed that this "antagonistic" effect was absent using either ethanol-killed pneumococci or a pneumococcal mutant deficient in capsule production and incapable of nasopharyngeal carriage. Colonization with a pneumococcal strain naturally unable to produce capsule also reduced viral loads. The pneumococcus-mediated reduction in PVM loads was caused by accelerated viral clearance from the nasopharynx. Although these synergistic and antagonistic effects occurred with both wild-type pneumococcal strains used in this study, the magnitude of the effects was strain dependent. Lastly, we showed that pneumococci can also antagonize influenza virus. Taken together, our study has uncovered multiple novel facets of bacterial-viral coinfection. Our findings have important public health implications, including for bacterial and viral vaccination strategies in young children. IMPORTANCE Respiratory bacterial-viral coinfections (such as pneumococci and influenza virus) are often synergistic, resulting in enhanced disease severity. Although colonization of the nasopharynx is the precursor to disease and transmission, little is known about bacterial-viral interactions that occur within this niche. In this study, we developed a novel mouse model to examine pneumococcal-viral interactions in the nasopharynx with pneumonia virus of mice (PVM) and influenza. We found that PVM infection benefits pneumococci by increasing their numbers in the nasopharynx and shedding of these bacteria in respiratory secretions. In contrast, we discovered that pneumococci decrease PVM numbers by accelerating viral clearance. We also report a similar effect of pneumococci on influenza. By showing that coinfections lead to both synergistic and antagonistic outcomes, our findings challenge the existing dogma in the field. Our work has important applications and implications for bacterial and viral vaccines that target these microbes.
Collapse
Affiliation(s)
- Sam Manna
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Julie McAuley
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jonathan Jacobson
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - Cattram D. Nguyen
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
| | - Md. Ashik Ullah
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Ismail Sebina
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Victoria Williamson
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | - E. Kim Mulholland
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Odilia Wijburg
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Simon Phipps
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Catherine Satzke
- Infection and Immunity, Murdoch Children’s Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
23
|
Byrne AJ, Saglani S, Snelgrove RJ. An Alarmin Role for P2Y 13 Receptor during Viral-driven Asthma Exacerbations. Am J Respir Crit Care Med 2022; 205:263-265. [PMID: 34936856 PMCID: PMC8886992 DOI: 10.1164/rccm.202111-2571ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Affiliation(s)
- Adam J Byrne
- National Heart and Lung Institute Imperial College London London, United Kingdom
| | - Sejal Saglani
- National Heart and Lung Institute Imperial College London London, United Kingdom
| | - Robert J Snelgrove
- National Heart and Lung Institute Imperial College London London, United Kingdom
| |
Collapse
|
24
|
Wang CJ, Cheng SL, Kuo SH. Asthma and COVID-19 Associations: Focus on IgE-Related Immune Pathology. Life (Basel) 2022; 12:life12020153. [PMID: 35207441 PMCID: PMC8874771 DOI: 10.3390/life12020153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/15/2022] [Accepted: 01/19/2022] [Indexed: 12/15/2022] Open
Abstract
Management of patients with asthma during the coronavirus disease 2019 (COVID-19) pandemic is a concern, especially since asthma predisposes patients to respiratory problems. Interestingly, asthma characterized by type 2 inflammation, also known as T-helper type 2-high endotype, displays a cellular and molecular profile that may confer protective effects against COVID-19. The results of experimental and clinical studies have established the actions of immunoglobulin E (IgE) in inducing airway hyperreactivity and weakening an interferon-mediated antiviral response following respiratory viral infection. Robust evidence supports the beneficial effect of the anti-IgE biologic treatment omalizumab on reducing respiratory virus-induced asthma exacerbations and reducing the frequency, duration, and severity of respiratory viral illness in patients with asthma. Indeed, accumulating reports of patients with severe asthma treated with omalizumab during the pandemic have reassuringly shown that continuing omalizumab treatment during COVID-19 is safe, and in fact may help prevent the severe course of COVID-19. Accordingly, guidance issued by the Global Initiative for Asthma recommends that all patients with asthma continue taking their prescribed asthma medications, including biologic therapy, during the COVID-19 pandemic. The impact of biologic treatments on patients with asthma and COVID-19 will be better understood as more evidence emerges.
Collapse
Affiliation(s)
- Chung-Jen Wang
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City 22056, Taiwan; (C.-J.W.); (S.-L.C.)
| | - Shih-Lung Cheng
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City 22056, Taiwan; (C.-J.W.); (S.-L.C.)
- Department of Chemical Engineering and Materials Science, Yuab Ze University, Taoyuan City 32003, Taiwan
| | - Sow-Hsong Kuo
- Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei City 22056, Taiwan; (C.-J.W.); (S.-L.C.)
- Correspondence:
| |
Collapse
|
25
|
Makrinioti H, Bush A, Gern J, Johnston SL, Papadopoulos N, Feleszko W, Camargo CA, Hasegawa K, Jartti T. The Role of Interferons in Driving Susceptibility to Asthma Following Bronchiolitis: Controversies and Research Gaps. Front Immunol 2021; 12:761660. [PMID: 34925333 PMCID: PMC8677668 DOI: 10.3389/fimmu.2021.761660] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/12/2021] [Indexed: 12/15/2022] Open
Abstract
Bronchiolitis is the most common cause of hospitalization in infancy and is associated with a higher risk for the development of childhood asthma. However, not all children hospitalized with bronchiolitis will develop asthma. The mechanisms underlying asthma development following bronchiolitis hospitalization are complex. Immune responses to respiratory viruses may underlie both bronchiolitis severity and long-term sequela (such as asthma). Interferons (IFNs) are important components of innate immune responses to respiratory viruses and could influence both asthma development and asthma exacerbations. However, the nature of the relationship between interferon production and wheezing illnesses is controversial. For example, low peripheral blood IFN responses at birth have been linked with recurrent wheeze and asthma development. In contrast, there is evidence that severe illnesses (e.g., hospitalization for bronchiolitis) are associated with increased IFN responses during acute infection (bronchiolitis hospitalization) and a higher risk for subsequent asthma diagnosis. Furthermore, mechanistic studies suggest that bronchial epithelial cells from asthmatic children have impaired IFN responses to respiratory viruses, which may enable increased viral replication followed by exaggerated secondary IFN responses. This review aims to discuss controversies around the role of IFNs as drivers of susceptibility to asthma development following bronchiolitis hospitalization. Past evidence from both mechanistic and cohort studies are discussed. We will highlight knowledge gaps that can inform future research study design.
Collapse
Affiliation(s)
- Heidi Makrinioti
- West Middlesex University Hospital, Chelsea, and Westminster Foundation Trust, London, United Kingdom.,Imperial Centre for Paediatrics and Child Health, Imperial College, London, United Kingdom
| | - Andrew Bush
- Imperial Centre for Paediatrics and Child Health, Imperial College, London, United Kingdom.,National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - James Gern
- Department of Paediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Wisconsin, WI, United States
| | | | - Nikolaos Papadopoulos
- Allergy Department, 2nd Paediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece.,Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
| | - Wojciech Feleszko
- Department of Paediatric Pneumology and Allergy, The Medical University of Warsaw, Warsaw, Poland
| | - Carlos A Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Tuomas Jartti
- Department of Paediatrics, Turku University Hospital and Turku University, Turku, Finland.,Research Unit for Pediatrics, Pediatric Neurology, Pediatric Surgery, Child Psychiatry, Dermatology, Clinical Genetics, Obstetrics and Gynecology, Otorhinolaryngology and Ophthalmology (PEDEGO), Medical Research Center, University of Oulu, Oulu, Finland.,Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
26
|
Schetters STT, Schuijs MJ. Pulmonary Eosinophils at the Center of the Allergic Space-Time Continuum. Front Immunol 2021; 12:772004. [PMID: 34868033 PMCID: PMC8634472 DOI: 10.3389/fimmu.2021.772004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/27/2021] [Indexed: 01/01/2023] Open
Abstract
Eosinophils are typically a minority population of circulating granulocytes being released from the bone-marrow as terminally differentiated cells. Besides their function in the defense against parasites and in promoting allergic airway inflammation, regulatory functions have now been attributed to eosinophils in various organs. Although eosinophils are involved in the inflammatory response to allergens, it remains unclear whether they are drivers of the asthma pathology or merely recruited effector cells. Recent findings highlight the homeostatic and pro-resolving capacity of eosinophils and raise the question at what point in time their function is regulated. Similarly, eosinophils from different physical locations display phenotypic and functional diversity. However, it remains unclear whether eosinophil plasticity remains as they develop and travel from the bone marrow to the tissue, in homeostasis or during inflammation. In the tissue, eosinophils of different ages and origin along the inflammatory trajectory may exhibit functional diversity as circumstances change. Herein, we outline the inflammatory time line of allergic airway inflammation from acute, late, adaptive to chronic processes. We summarize the function of the eosinophils in regards to their resident localization and time of recruitment to the lung, in all stages of the inflammatory response. In all, we argue that immunological differences in eosinophils are a function of time and space as the allergic inflammatory response is initiated and resolved.
Collapse
Affiliation(s)
- Sjoerd T T Schetters
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Martijn J Schuijs
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium.,Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
27
|
Werder RB, Ullah MA, Rahman MM, Simpson J, Lynch JP, Collinson N, Rittchen S, Rashid RB, Sikder MAA, Handoko HY, Curren BF, Sebina I, Hartel G, Bissell A, Ngo S, Yarlagadda T, Hasnain SZ, Lu W, Sohal SS, Martin M, Bowler S, Burr LD, Martinez LO, Robaye B, Spann K, Ferreira MAR, Phipps S. Targeting the P2Y13 Receptor Suppresses IL-33 and HMGB1 Release and Ameliorates Experimental Asthma. Am J Respir Crit Care Med 2021; 205:300-312. [PMID: 34860143 DOI: 10.1164/rccm.202009-3686oc] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE The alarmins IL-33 and HMGB1 (high mobility group box 1) contribute to type-2 inflammation and asthma pathogenesis. OBJECTIVES To determine whether P2Y13 receptor (P2Y13-R), a purinergic G protein-coupled receptor (GPCR) and risk allele for asthma, regulates the release of IL-33 and HMGB1. METHODS Bronchial biopsies were obtained from healthy and asthmatic subjects. Primary human airway epithelial cells (AECs), primary mouse (m)AECs, or C57Bl/6 mice were inoculated with various aeroallergens or respiratory viruses, and the nuclear-to-cytoplasmic translocation and release of alarmins measured by immunohistochemistry and ELISA. The role of P2Y13-R in AEC function and in the onset, progression, and an exacerbation of experimental asthma, was assessed using pharmacological antagonists and P2Y13-R gene-deleted mice. MEASUREMENTS AND MAIN RESULTS Aeroallergen-exposure induced the extracellular release of ADP and ATP, nucleotides that activate P2Y13-R. ATP, ADP, aeroallergen (house dust mite, cockroach or Alternaria) or virus exposure induced the nuclear-to-cytoplasmic translocation and subsequent release of IL-33 and HMGB1, and this response was ablated by genetic deletion or pharmacological antagonism of P2Y13. In mice, prophylactic or therapeutic P2Y13-R blockade attenuated asthma onset, and critically, ablated the severity of a rhinovirus-associated exacerbation in a high-fidelity experimental model of chronic asthma. Moreover, P2Y13-R antagonism derepressed antiviral immunity, increasing IFN-λ production and decreasing viral copies in the lung. CONCLUSIONS We identify P2Y13-R as a novel gatekeeper of the nuclear alarmins IL-33 and HMGB1, and demonstrate that the targeting of this GPCR via genetic deletion or treatment with a small-molecule antagonist protects against the onset and exacerbations of experimental asthma.
Collapse
Affiliation(s)
- Rhiannon B Werder
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia.,The University of Queensland, 1974, Faculty of Medicine, Brisbane, Queensland, Australia.,Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, Massachusetts, United States.,Boston University School of Medicine, 12259, The Pulmonary Center and Department of Medicine, Boston, Massachusetts, United States
| | - Md Ashik Ullah
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia
| | - Muhammed Mahfuzur Rahman
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia.,The University of Queensland, 1974, Faculty of Medicine, Brisbane, Queensland, Australia
| | - Jennifer Simpson
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia.,The University of Queensland, 1974, Faculty of Medicine, Brisbane, Queensland, Australia.,National Institute of Allergy and Infectious Diseases, 35037, Barrier Immunity Section, Laboratory of Viral Diseases, Bethesda, Maryland, United States
| | - Jason P Lynch
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia.,Harvard Medical School, 1811, Department of Microbiology, Boston, Massachusetts, United States
| | - Natasha Collinson
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia
| | - Sonja Rittchen
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia.,Medical University of Graz, 31475, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Graz, Steiermark, Austria
| | - Ridwan B Rashid
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia.,The University of Queensland, 1974, Faculty of Medicine, Brisbane, Queensland, Australia
| | - Md Al Amin Sikder
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia.,The University of Queensland, 1974, Faculty of Medicine, Brisbane, Queensland, Australia
| | - Herlina Y Handoko
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia
| | - Bodie F Curren
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia.,The University of Queensland, 1974, Faculty of Medicine, Brisbane, Queensland, Australia
| | - Ismail Sebina
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia
| | - Gunter Hartel
- QIMR Berghofer, 56362, Brisbane, Queensland, Australia
| | - Alec Bissell
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia
| | - Sylvia Ngo
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia
| | - Tejasri Yarlagadda
- Queensland University of Technology Faculty of Health, 110544, Kelvin Grove, Queensland, Australia
| | - Sumaira Z Hasnain
- Mater Medical Research Institute, 200098, Brisbane, Queensland, Australia
| | - Wenying Lu
- University of Tasmania, 3925, Respiratory Translational Research Group, Launceston , Tasmania, Australia
| | - Sukhwinder S Sohal
- University of Tasmania , Respiratory Translational Research Group, Launceston , Tasmania, Australia
| | - Megan Martin
- Mater Health Services, Respiratory Medicine, South Brisbane, Queensland, Australia
| | - Simon Bowler
- Mater Health Services, Respiratory Medicine, South Brisbane, Queensland, Australia
| | - Lucy D Burr
- UQ School of Medicine, Brisbane, Queensland, Australia
| | - Laurent O Martinez
- University of Toulouse, 137668, Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
| | - Bernard Robaye
- Université Libre de Bruxelles, 26659, IRIBHM, Bruxelles, Belgium
| | - Kirsten Spann
- Queensland University of Technology, 1969, School of Biomedical Sciences, Brisbane, Queensland, Australia
| | - Manuel A R Ferreira
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia
| | - Simon Phipps
- QIMR Berghofer Medical Research Institute, 56362, Respiratory Immunology Laboratory, Brisbane, Queensland, Australia.,The University of Queensland, 1974, Faculty of Medicine, Brisbane, Queensland, Australia.,The University of Queensland, 1974, Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia;
| |
Collapse
|
28
|
Sánchez-Marteles M, Rubio-Gracia J, Peña-Fresneda N, Garcés-Horna V, Gracia-Tello B, Martínez-Lostao L, Crespo-Aznárez S, Pérez-Calvo JI, Giménez-López I. Early Measurement of Blood sST2 Is a Good Predictor of Death and Poor Outcomes in Patients Admitted for COVID-19 Infection. J Clin Med 2021; 10:3534. [PMID: 34441830 PMCID: PMC8396994 DOI: 10.3390/jcm10163534] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/08/2021] [Accepted: 08/08/2021] [Indexed: 01/08/2023] Open
Abstract
Although several biomarkers have shown correlation to prognosis in COVID-19 patients, their clinical value is limited because of lack of specificity, suboptimal sensibility or poor dynamic behavior. We hypothesized that circulating soluble ST2 (sST2) could be associated to a worse outcome in COVID-19. In total, 152 patients admitted for confirmed COVID-19 were included in a prospective non-interventional, observational study. Blood samples were drawn at admission, 48-72 h later and at discharge. sST2 concentrations and routine blood laboratory were analyzed. Primary endpoints were admission at intensive care unit (ICU) and mortality. Median age was 57.5 years [Standard Deviation (SD: 12.8)], 60.4% males. 10% of patients (n = 15) were derived to ICU and/or died during admission. Median (IQR) sST2 serum concentration (ng/mL) rose to 53.1 (30.9) at admission, peaked at 48-72 h (79.5(64)) and returned to admission levels at discharge (44.9[36.7]). A concentration of sST2 above 58.9 ng/mL was identified patients progressing to ICU admission or death. Results remained significant after multivariable analysis. The area under the receiver operating characteristics curve (AUC) of sST2 for endpoints was 0.776 (p = 0.001). In patients admitted for COVID-19 infection, early measurement of sST2 was able to identify patients at risk of severe complications or death.
Collapse
Affiliation(s)
- Marta Sánchez-Marteles
- Department of Internal Medicine, Hospital Clínico Universitario, Lozano Blesa, 50009 Zaragoza, Spain; (J.R.-G.); (V.G.-H.); (B.G.-T.); (S.C.-A.); (J.I.P.-C.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (N.P.-F.); (L.M.-L.); (I.G.-L.)
| | - Jorge Rubio-Gracia
- Department of Internal Medicine, Hospital Clínico Universitario, Lozano Blesa, 50009 Zaragoza, Spain; (J.R.-G.); (V.G.-H.); (B.G.-T.); (S.C.-A.); (J.I.P.-C.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (N.P.-F.); (L.M.-L.); (I.G.-L.)
| | - Natacha Peña-Fresneda
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (N.P.-F.); (L.M.-L.); (I.G.-L.)
- Facultad de Medicina, University of Zaragoza, 50009 Zaragoza, Spain
| | - Vanesa Garcés-Horna
- Department of Internal Medicine, Hospital Clínico Universitario, Lozano Blesa, 50009 Zaragoza, Spain; (J.R.-G.); (V.G.-H.); (B.G.-T.); (S.C.-A.); (J.I.P.-C.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (N.P.-F.); (L.M.-L.); (I.G.-L.)
| | - Borja Gracia-Tello
- Department of Internal Medicine, Hospital Clínico Universitario, Lozano Blesa, 50009 Zaragoza, Spain; (J.R.-G.); (V.G.-H.); (B.G.-T.); (S.C.-A.); (J.I.P.-C.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (N.P.-F.); (L.M.-L.); (I.G.-L.)
| | - Luis Martínez-Lostao
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (N.P.-F.); (L.M.-L.); (I.G.-L.)
- Facultad de Medicina, University of Zaragoza, 50009 Zaragoza, Spain
- Department of Immunology, Hospital Clínico Universitario, Lozano Blesa, 50009 Zaragoza, Spain
| | - Silvia Crespo-Aznárez
- Department of Internal Medicine, Hospital Clínico Universitario, Lozano Blesa, 50009 Zaragoza, Spain; (J.R.-G.); (V.G.-H.); (B.G.-T.); (S.C.-A.); (J.I.P.-C.)
| | - Juan Ignacio Pérez-Calvo
- Department of Internal Medicine, Hospital Clínico Universitario, Lozano Blesa, 50009 Zaragoza, Spain; (J.R.-G.); (V.G.-H.); (B.G.-T.); (S.C.-A.); (J.I.P.-C.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (N.P.-F.); (L.M.-L.); (I.G.-L.)
- Facultad de Medicina, University of Zaragoza, 50009 Zaragoza, Spain
| | - Ignacio Giménez-López
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain; (N.P.-F.); (L.M.-L.); (I.G.-L.)
- Facultad de Medicina, University of Zaragoza, 50009 Zaragoza, Spain
- Instituto Aragonés de Ciencias de la Salud (IACS), 50009 Zaragoza, Spain
| |
Collapse
|
29
|
Raita Y, Pérez-Losada M, Freishtat RJ, Harmon B, Mansbach JM, Piedra PA, Zhu Z, Camargo CA, Hasegawa K. Integrated omics endotyping of infants with respiratory syncytial virus bronchiolitis and risk of childhood asthma. Nat Commun 2021; 12:3601. [PMID: 34127671 PMCID: PMC8203688 DOI: 10.1038/s41467-021-23859-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 05/17/2021] [Indexed: 02/04/2023] Open
Abstract
Respiratory syncytial virus (RSV) bronchiolitis is not only the leading cause of hospitalization in U.S. infants, but also a major risk factor for asthma development. While emerging evidence suggests clinical heterogeneity within RSV bronchiolitis, little is known about its biologically-distinct endotypes. Here, we integrated clinical, virus, airway microbiome (species-level), transcriptome, and metabolome data of 221 infants hospitalized with RSV bronchiolitis in a multicentre prospective cohort study. We identified four biologically- and clinically-meaningful endotypes: A) clinicalclassicmicrobiomeM. nonliquefaciensinflammationIFN-intermediate, B) clinicalatopicmicrobiomeS. pneumoniae/M. catarrhalisinflammationIFN-high, C) clinicalseveremicrobiomemixedinflammationIFN-low, and D) clinicalnon-atopicmicrobiomeM.catarrhalisinflammationIL-6. Particularly, compared with endotype A infants, endotype B infants-who are characterized by a high proportion of IgE sensitization and rhinovirus coinfection, S. pneumoniae/M. catarrhalis codominance, and high IFN-α and -γ response-had a significantly higher risk for developing asthma (9% vs. 38%; OR, 6.00: 95%CI, 2.08-21.9; P = 0.002). Our findings provide an evidence base for the early identification of high-risk children during a critical period of airway development.
Collapse
Affiliation(s)
- Yoshihiko Raita
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Marcos Pérez-Losada
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, The George Washington University, Washington, DC, USA
- CIBIO-InBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, Universidade do Porto, Campus Agrário de Vairão, Vairão, Portugal
| | - Robert J Freishtat
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC, USA
- Division of Emergency Medicine, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Brennan Harmon
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC, USA
| | - Jonathan M Mansbach
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pedro A Piedra
- Departments of Molecular Virology and Microbiology and Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Zhaozhong Zhu
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Carlos A Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
30
|
Liang Y, Ge Y, Sun J. IL-33 in COVID-19: friend or foe? Cell Mol Immunol 2021; 18:1602-1604. [PMID: 33972738 PMCID: PMC8108013 DOI: 10.1038/s41423-021-00685-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 04/10/2021] [Indexed: 01/30/2023] Open
Affiliation(s)
- Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA. .,Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.
| | - Yiyue Ge
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA. .,Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
31
|
The basic immunology of asthma. Cell 2021; 184:1469-1485. [PMID: 33711259 DOI: 10.1016/j.cell.2021.02.016] [Citation(s) in RCA: 503] [Impact Index Per Article: 125.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/21/2021] [Accepted: 02/04/2021] [Indexed: 12/21/2022]
Abstract
In many asthmatics, chronic airway inflammation is driven by IL-4-, IL-5-, and IL-13-producing Th2 cells or ILC2s. Type 2 cytokines promote hallmark features of the disease such as eosinophilia, mucus hypersecretion, bronchial hyperresponsiveness (BHR), IgE production, and susceptibility to exacerbations. However, only half the asthmatics have this "type 2-high" signature, and "type 2-low" asthma is more associated with obesity, presence of neutrophils, and unresponsiveness to corticosteroids, the mainstay asthma therapy. Here, we review the underlying immunological basis of various asthma endotypes by discussing results obtained from animal studies as well as results generated in clinical studies targeting specific immune pathways.
Collapse
|
32
|
DP1 prostanoid receptor activation increases the severity of an acute lower respiratory viral infection in mice via TNF-α-induced immunopathology. Mucosal Immunol 2021; 14:963-972. [PMID: 33879829 PMCID: PMC8057290 DOI: 10.1038/s41385-021-00405-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 03/17/2021] [Accepted: 04/07/2021] [Indexed: 02/04/2023]
Abstract
Respiratory syncytial virus (RSV) bronchiolitis is a leading cause of infant hospitalization and mortality. We previously identified that prostaglandin D2 (PGD2), released following RSV infection of primary human airway epithelial cells or pneumonia virus of mice (PVM) infection of neonatal mice, elicits pro- or antiviral innate immune responses as a consequence of D-type prostanoid receptor 2 (DP2) or DP1 activation, respectively. Here, we sought to determine whether treatment with the DP1 agonist BW245c decreases the severity of bronchiolitis in PVM-infected neonatal mice. Consistent with previous findings, BW245c treatment increased IFN-λ production and decreased viral load in week 1 of the infection. However, unexpectedly, BW245c treatment increased mortality in week 2 of the infection. This increased morbidity was associated with viral spread to the parenchyma, an increased cellular infiltrate of TNF-α-producing cells (neutrophils, monocytes, and CD4+ T cells), and the heightened production of the pro-inflammatory cytokines TNF-α, IL-6, and IL-1β. These phenotypes, as well as the increased mortality, were significantly attenuated following the administration of anti-TNF-α to PVM-infected, BW245c-treated mice. In summary, pharmacological activation of the DP1 receptor in PVM-infected neonatal mice boosts antiviral innate and adaptive immunity, however, this is ultimately detrimental, as a consequence of increased TNF-α-induced morbidity and mortality.
Collapse
|
33
|
Respiratory syncytial virus upregulates IL-33 expression in mouse model of virus-induced inflammation exacerbation in OVA-sensitized mice and in asthmatic subjects. Cytokine 2020; 138:155349. [PMID: 33132030 DOI: 10.1016/j.cyto.2020.155349] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND Bronchial asthma (BA) is a chronic disease of the airways. The great majority of BA exacerbations are associated with respiratory viral infections. Recent findings point out a possible role of proinflammatory cytokine interleukin-33 (IL-33) in the development of atopic diseases. Although, little is known about the role of IL-33 in virus-induced BA exacerbations. METHODS We used mouse models of RSV (respiratory syncytial virus)-induced inflammation exacerbation in OVA-sensitized mice and RSV infection alone in adult animals to characterize expression of il33 in the mouse lungs. Moreover, we studied the influence of il33 knockdown with intranasally administrated siRNA on the development of RSV-induced inflammation exacerbation. In addition, we evaluated the expression of IL33 in the ex vivo stimulated PBMCs from allergic asthma patients and healthy subjects with and without confirmed acute respiratory viral infection. RESULTS Using mouse models, we found that infection with RSV drives enhanced il33 mRNA expression in the mouse lung. Treatment with anti-il33 siRNA diminishes airway inflammation in the lungs (we found a decrease in the number of inflammatory cells in the lungs and in the severity of histopathological alterations) of mice with RSV-induced inflammation exacerbation, but do not influence viral load. Elevated level of the IL33 mRNA was detected in ex vivo stimulated blood lymphocytes of allergic asthmatics infected with respiratory viruses. RSV and rhinovirus were the most detected viruses in volunteers with symptoms of respiratory infection. CONCLUSION The present study provides additional evidence of the crucial role of the IL-33 in pathogenesis of RSV infection and virus-induced allergic bronchial asthma exacerbations.
Collapse
|
34
|
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: 27] [Impact Index Per Article: 5.4] [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
|
35
|
What Makes the Lung Unique – Tissue-Specific Immunity in the Respiratory Tract. EUROPEAN MEDICAL JOURNAL 2020. [DOI: 10.33590/emj/20-00089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The immune system constitutes a critical mechanism of the human body to preserve health and mitigate disease. In the lung, immunity is seen as a critical driver in many respiratory diseases, in particular in those characterised by aberrant inflammation, such as chronic obstructive pulmonary disease, fibrosis, and asthma. In this review, the specialised set of immune cells and lung tissue-specific regulators, including key cytokines such as granulocyte-macrophage colony-stimulating factor and transforming growth factor β, that control immune responses in the respiratory tract will be discussed. Furthermore, the current understanding of the impact of key environmental components such as the role of oxygen and lung microbiota on lung immunity will be highlighted. The goal is to identify the unique aspects of lung immune biology to facilitate insights into the aetiology of common lung inflammatory diseases and to provide the basis for a deeper mechanistic understanding of the underlying immune processes. Finally, key future avenues of research such as using more comprehensive quantitative approaches for elucidating molecular disease mechanisms as well as the potential to exploit tissue-specific regulators of immunity for therapy of lung inflammatory disorders will be discussed.
Collapse
|
36
|
Simpson J, Loh Z, Ullah MA, Lynch JP, Werder RB, Collinson N, Zhang V, Dondelinger Y, Bertrand MJM, Everard ML, Blyth CC, Hartel G, Van Oosterhout AJ, Gough PJ, Bertin J, Upham JW, Spann KM, Phipps S. Respiratory Syncytial Virus Infection Promotes Necroptosis and HMGB1 Release by Airway Epithelial Cells. Am J Respir Crit Care Med 2020; 201:1358-1371. [PMID: 32105156 DOI: 10.1164/rccm.201906-1149oc] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Rationale: Respiratory syncytial virus (RSV) bronchiolitis causes significant infant mortality. Bronchiolitis is characterized by airway epithelial cell (AEC) death; however, the mode of death remains unknown.Objectives: To determine whether necroptosis contributes to RSV bronchiolitis pathogenesis via HMGB1 (high mobility group box 1) release.Methods: Nasopharyngeal samples were collected from children presenting to the hospital with acute respiratory infection. Primary human AECs and neonatal mice were inoculated with RSV and murine Pneumovirus, respectively. Necroptosis was determined via viability assays and immunohistochemistry for RIPK1 (receptor-interacting protein kinase-1), MLKL (mixed lineage kinase domain-like pseudokinase) protein, and caspase-3. Necroptosis was blocked using pharmacological inhibitors and RIPK1 kinase-dead knockin mice.Measurements and Main Results: HMGB1 levels were elevated in nasopharyngeal samples of children with acute RSV infection. RSV-induced epithelial cell death was associated with increased phosphorylated RIPK1 and phosphorylated MLKL but not active caspase-3 expression. Inhibition of RIPK1 or MLKL attenuated RSV-induced HMGB1 translocation and release, and lowered viral load. MLKL inhibition increased active caspase-3 expression in a caspase-8/9-dependent manner. In susceptible mice, Pneumovirus infection upregulated RIPK1 and MLKL expression in the airway epithelium at 8 to 10 days after infection, coinciding with AEC sloughing, HMGB1 release, and neutrophilic inflammation. Genetic or pharmacological inhibition of RIPK1 or MLKL attenuated these pathologies, lowered viral load, and prevented type 2 inflammation and airway remodeling. Necroptosis inhibition in early life ameliorated asthma progression induced by viral or allergen challenge in later life.Conclusions: Pneumovirus infection induces AEC necroptosis. Inhibition of necroptosis may be a viable strategy to limit the severity of viral bronchiolitis and break its nexus with asthma.
Collapse
Affiliation(s)
- Jennifer Simpson
- QIMR Berghofer Medical Research Institute, Herston, Australia.,School of Biomedical Science, University of Queensland, Brisbane, Queensland, Australia
| | - Zhixuan Loh
- School of Biomedical Science, University of Queensland, Brisbane, Queensland, Australia
| | - Md Ashik Ullah
- QIMR Berghofer Medical Research Institute, Herston, Australia.,School of Biomedical Science, University of Queensland, Brisbane, Queensland, Australia
| | - Jason P Lynch
- QIMR Berghofer Medical Research Institute, Herston, Australia.,School of Biomedical Science, University of Queensland, Brisbane, Queensland, Australia
| | - Rhiannon B Werder
- QIMR Berghofer Medical Research Institute, Herston, Australia.,School of Biomedical Science, University of Queensland, Brisbane, Queensland, Australia
| | | | - Vivian Zhang
- QIMR Berghofer Medical Research Institute, Herston, Australia.,School of Biomedical Science, University of Queensland, Brisbane, Queensland, Australia
| | - Yves Dondelinger
- VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Mathieu J M Bertrand
- VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | | | - Christopher C Blyth
- School of Medicine and.,Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia.,Department of Infectious Diseases, Perth Children's Hospital, Perth, Western Australia, Australia.,Department of Microbiology, PathWest Laboratory Medicine WA, QEII Medical Centre, Perth, Western Australia, Australia
| | - Gunter Hartel
- QIMR Berghofer Medical Research Institute, Herston, Australia
| | | | | | | | - John W Upham
- University of Queensland Diamantina Institute, Brisbane, Queensland, Australia.,Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia; and
| | - Kirsten M Spann
- Queensland University of Technology, Brisbane, Queensland, Australia
| | - Simon Phipps
- QIMR Berghofer Medical Research Institute, Herston, Australia.,School of Biomedical Science, University of Queensland, Brisbane, Queensland, Australia.,Australian Infectious Diseases Research Centre, Brisbane, Queensland, Australia; and
| |
Collapse
|
37
|
Loh Z, Simpson J, Ullah A, Zhang V, Gan WJ, Lynch JP, Werder RB, Sikder AA, Lane K, Sim CB, Porrello E, Mazzone SB, Sly PD, Steptoe RJ, Spann KM, Sukkar MB, Upham JW, Phipps S. HMGB1 amplifies ILC2-induced type-2 inflammation and airway smooth muscle remodelling. PLoS Pathog 2020; 16:e1008651. [PMID: 32658914 PMCID: PMC7377495 DOI: 10.1371/journal.ppat.1008651] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/23/2020] [Accepted: 05/24/2020] [Indexed: 12/16/2022] Open
Abstract
Type-2 immunity elicits tissue repair and homeostasis, however dysregulated type-2 responses cause aberrant tissue remodelling, as observed in asthma. Severe respiratory viral infections in infancy predispose to later asthma, however, the processes that mediate tissue damage-induced type-2 inflammation and the origins of airway remodelling remain ill-defined. Here, using a preclinical mouse model of viral bronchiolitis, we find that increased epithelial and mesenchymal high-mobility group box 1 (HMGB1) expression is associated with increased numbers of IL-13-producing type-2 innate lymphoid cell (ILC2s) and the expansion of the airway smooth muscle (ASM) layer. Anti-HMGB1 ablated lung ILC2 numbers and ASM growth in vivo, and inhibited ILC2-mediated ASM cell proliferation in a co-culture model. Furthermore, we identified that HMGB1/RAGE (receptor for advanced glycation endproducts) signalling mediates an ILC2-intrinsic IL-13 auto-amplification loop. In summary, therapeutic targeting of the HMGB1/RAGE signalling axis may act as a novel asthma preventative by dampening ILC2-mediated type-2 inflammation and associated ASM remodelling. Asthma can start at any time in life, although most often begins in early childhood. Wheezy viral bronchiolitis is a major independent risk factor for subsequent asthma. However, key knowledge gaps exist in relation to the sequelae of severe viral bronchiolitis and the pathogenic processes that promote type-2 inflammation and airway wall remodelling, cardinal features of asthma. Our study addresses this gap by identifying high-mobility group box 1 as a pathogenic cytokine that contributes to group 2 innate lymphoid cell-induced airway smooth muscle growth.
Collapse
Affiliation(s)
- Zhixuan Loh
- School of Biomedical Sciences, The University of Queensland, Queensland, Australia
- Institute for Molecular Bioscience, The University of Queensland, Queensland, Australia
| | - Jennifer Simpson
- School of Biomedical Sciences, The University of Queensland, Queensland, Australia
- QIMR Berghofer Medical Research Institute, Queensland, Australia
| | - Ashik Ullah
- School of Biomedical Sciences, The University of Queensland, Queensland, Australia
- QIMR Berghofer Medical Research Institute, Queensland, Australia
| | - Vivian Zhang
- QIMR Berghofer Medical Research Institute, Queensland, Australia
| | - Wan J. Gan
- School of Biomedical Sciences, The University of Queensland, Queensland, Australia
| | - Jason P. Lynch
- School of Biomedical Sciences, The University of Queensland, Queensland, Australia
- QIMR Berghofer Medical Research Institute, Queensland, Australia
| | - Rhiannon B. Werder
- School of Biomedical Sciences, The University of Queensland, Queensland, Australia
- QIMR Berghofer Medical Research Institute, Queensland, Australia
| | - Al Amin Sikder
- School of Biomedical Sciences, The University of Queensland, Queensland, Australia
- QIMR Berghofer Medical Research Institute, Queensland, Australia
| | - Katie Lane
- School of Biomedical Sciences, The University of Queensland, Queensland, Australia
| | - Choon Boon Sim
- Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
| | - Enzo Porrello
- Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
| | - Stuart B. Mazzone
- School of Biomedical Sciences, The University of Queensland, Queensland, Australia
- Department of Anatomy and Neuroscience, University of Melbourne, Victoria, Australia
| | - Peter D. Sly
- Children’s Health and Environment Program, Child Health Research Centre, University of Queensland, Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Queensland, Australia
| | - Raymond J. Steptoe
- UQ Diamantina Institute, The University of Queensland, Queensland, Australia
| | - Kirsten M. Spann
- School of Biomedical Sciences, Queensland University of Technology, Queensland, Australia
| | - Maria B. Sukkar
- Graduate School of Health, Faculty of Health, University of Technology Sydney, Ultimo, NSW, Australia
| | - John W. Upham
- UQ Diamantina Institute, The University of Queensland, Queensland, Australia
| | - Simon Phipps
- School of Biomedical Sciences, The University of Queensland, Queensland, Australia
- QIMR Berghofer Medical Research Institute, Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Queensland, Australia
- * E-mail:
| |
Collapse
|
38
|
Zhu JJ, Stenfeldt C, Bishop EA, Canter JA, Eschbaumer M, Rodriguez LL, Arzt J. Mechanisms of Maintenance of Foot-and-Mouth Disease Virus Persistence Inferred From Genes Differentially Expressed in Nasopharyngeal Epithelia of Virus Carriers and Non-carriers. Front Vet Sci 2020; 7:340. [PMID: 32637426 PMCID: PMC7318773 DOI: 10.3389/fvets.2020.00340] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/15/2020] [Indexed: 12/13/2022] Open
Abstract
Foot-and-mouth disease virus (FMDV) causes persistent infection of nasopharyngeal epithelial cells in ~50% of infected ruminants. The mechanisms involved are not clear. This study provides a continued investigation of differentially expressed genes (DEG) identified in a previously published transcriptomic study analyzing micro-dissected epithelial samples from FMDV carriers and non-carriers. Pathway analysis of DEG indicated that immune cell trafficking, cell death and hematological system could be affected by the differential gene expression. Further examination of the DEG identified five downregulated (chemerin, CCL23, CXCL15, CXCL16, and CXCL17) and one upregulated (CCL2) chemokines in carriers compared to non-carriers. The differential expression could reduce the recruitment of neutrophils, antigen-experienced T cells and dendritic cells and increase the migration of macrophages and NK cells to the epithelia in carriers, which was supported by DEG expressed in these immune cells. Downregulated chemokine expression could be mainly due to the inhibition of canonical NFκB signaling based on DEG in the signaling pathways and transcription factor binding sites predicted from the proximal promoters. Additionally, upregulated CD69, IL33, and NID1 and downregulated CASP3, IL17RA, NCR3LG1, TP53BP1, TRAF3, and TRAF6 in carriers could inhibit the Th17 response, NK cell cytotoxicity and apoptosis. Based on our findings, we hypothesize that (1) under-expression of chemokines that recruit neutrophils, antigen-experienced T cells and dendritic cells, (2) blocking NK cell binding to target cells and (3) suppression of apoptosis induced by death receptor signaling, viral RNA, and cell-mediated cytotoxicity in the epithelia compromised virus clearance and allowed FMDV to persist. These hypothesized mechanisms provide novel information for further investigation of persistent FMDV infection.
Collapse
Affiliation(s)
- James J Zhu
- USDA-ARS, Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Orient, NY, United States
| | - Carolina Stenfeldt
- USDA-ARS, Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Orient, NY, United States.,Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, United States
| | - Elizabeth A Bishop
- USDA-ARS, Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Orient, NY, United States
| | - Jessica A Canter
- USDA-ARS, Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Orient, NY, United States.,Plum Island Animal Disease Center, Oak Ridge Institute for Science and Education (ORISE), Orient, NY, United States
| | - Michael Eschbaumer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Greifswald, Germany
| | - Luis L Rodriguez
- USDA-ARS, Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Orient, NY, United States
| | - Jonathan Arzt
- USDA-ARS, Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Orient, NY, United States
| |
Collapse
|
39
|
Lynch JP, Werder RB, Curren BF, Sikder MAA, Ullah A, Sebina I, Rashid RB, Zhang V, Upham JW, Hill GR, Steptoe RJ, Phipps S. Long-lived regulatory T cells generated during severe bronchiolitis in infancy influence later progression to asthma. Mucosal Immunol 2020; 13:652-664. [PMID: 32066837 DOI: 10.1038/s41385-020-0268-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 01/22/2020] [Accepted: 01/26/2020] [Indexed: 02/04/2023]
Abstract
The type-2 inflammatory response that promotes asthma pathophysiology occurs in the absence of sufficient immunoregulation. Impaired regulatory T cell (Treg) function also predisposes to severe viral bronchiolitis in infancy, a major risk factor for asthma. Hence, we hypothesized that long-lived, aberrantly programmed Tregs causally link viral bronchiolitis with later asthma. Here we found that transient plasmacytoid dendritic cell (pDC) depletion during viral infection in early-life, which causes the expansion of aberrant Tregs, predisposes to allergen-induced or virus-induced asthma in later-life, and is associated with altered airway epithelial cell (AEC) responses and the expansion of impaired, long-lived Tregs. Critically, the adoptive transfer of aberrant Tregs (unlike healthy Tregs) to asthma-susceptible mice failed to prevent the development of viral-induced or allergen-induced asthma. Lack of protection was associated with increased airway epithelial cytoplasmic-HMGB1 (high-mobility group box 1), a pro-type-2 inflammatory alarmin, and granulocytic inflammation. Aberrant Tregs expressed lower levels of CD39, an ectonucleotidase that hydrolyzes extracellular ATP, a known inducer of alarmin release. Using cultured mouse AECs, we identify that healthy Tregs suppress allergen-induced HMGB1 translocation whereas this ability is markedly impaired in aberrant Tregs. Thus, defective Treg programming in infancy has durable consequences that underlie the association between bronchiolitis and subsequent asthma.
Collapse
Affiliation(s)
- Jason P Lynch
- Respiratory Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, 4006, Australia.,School of Biomedical Sciences, University of Queensland, Queensland, 4072, Australia
| | - Rhiannon B Werder
- Respiratory Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, 4006, Australia.,School of Biomedical Sciences, University of Queensland, Queensland, 4072, Australia
| | - Bodie F Curren
- Respiratory Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, 4006, Australia.,School of Biomedical Sciences, University of Queensland, Queensland, 4072, Australia
| | - Md Al Amin Sikder
- Respiratory Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, 4006, Australia.,School of Biomedical Sciences, University of Queensland, Queensland, 4072, Australia
| | - Ashik Ullah
- Respiratory Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, 4006, Australia
| | - Ismail Sebina
- Respiratory Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, 4006, Australia
| | - Ridwan B Rashid
- Respiratory Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, 4006, Australia.,School of Biomedical Sciences, University of Queensland, Queensland, 4072, Australia
| | - Vivian Zhang
- Respiratory Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, 4006, Australia.,School of Biomedical Sciences, University of Queensland, Queensland, 4072, Australia
| | - John W Upham
- UQ Diamantina Institute, The University of Queensland, Queensland, 4102, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, Queensland, 4072, Australia
| | - Geoff R Hill
- Fred Hutchinson Cancer Research Center, Seattle, WA, 1100, USA
| | - Raymond J Steptoe
- UQ Diamantina Institute, The University of Queensland, Queensland, 4102, Australia
| | - Simon Phipps
- Respiratory Immunology, QIMR Berghofer Medical Research Institute, Herston, Queensland, 4006, Australia. .,Australian Infectious Diseases Research Centre, The University of Queensland, Queensland, 4072, Australia.
| |
Collapse
|
40
|
Matsumoto K, Iikura K, Morita H, Saito H. Barrier dysfunction in the atopic march-how does atopic dermatitis lead to asthma in children? J Allergy Clin Immunol 2020; 145:1551-1553. [PMID: 32344057 DOI: 10.1016/j.jaci.2020.04.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/03/2020] [Accepted: 04/17/2020] [Indexed: 11/17/2022]
Affiliation(s)
- Kenji Matsumoto
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.
| | - Katsuhito Iikura
- Department of Pediatrics, National Hospital Organization Nishisaitama-chuo National Hospital, Saitama, Japan
| | - Hideaki Morita
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Hirohisa Saito
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| |
Collapse
|
41
|
Muehling LM, Heymann PW, Wright PW, Eccles JD, Agrawal R, Carper HT, Murphy DD, Workman LJ, Word CR, Ratcliffe SJ, Capaldo BJ, Platts-Mills TAE, Turner RB, Kwok WW, Woodfolk JA. Human T H1 and T H2 cells targeting rhinovirus and allergen coordinately promote allergic asthma. J Allergy Clin Immunol 2020; 146:555-570. [PMID: 32320734 DOI: 10.1016/j.jaci.2020.03.037] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/03/2020] [Accepted: 03/27/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Allergic asthmatic subjects are uniquely susceptible to acute wheezing episodes provoked by rhinovirus. However, the underlying immune mechanisms and interaction between rhinovirus and allergy remain enigmatic, and current paradigms are controversial. OBJECTIVE We sought to perform a comprehensive analysis of type 1 and type 2 innate and adaptive responses in allergic asthmatic subjects infected with rhinovirus. METHODS Circulating virus-specific TH1 cells and allergen-specific TH2 cells were precisely monitored before and after rhinovirus challenge in allergic asthmatic subjects (total IgE, 133-4692 IU/mL; n = 28) and healthy nonallergic controls (n = 12) using peptide/MHCII tetramers. T cells were sampled for up to 11 weeks to capture steady-state and postinfection phases. T-cell responses were analyzed in parallel with 18 cytokines in the nose, upper and lower airway symptoms, and lung function. The influence of in vivo IgE blockade was also examined. RESULTS In uninfected asthmatic subjects, higher numbers of circulating virus-specific PD-1+ TH1 cells, but not allergen-specific TH2 cells, were linked to worse lung function. Rhinovirus infection induced an amplified antiviral TH1 response in asthmatic subjects versus controls, with synchronized allergen-specific TH2 expansion, and production of type 1 and 2 cytokines in the nose. In contrast, TH2 responses were absent in infected asthmatic subjects who had normal lung function, and in those receiving anti-IgE. Across all subjects, early induction of a minimal set of nasal cytokines that discriminated high responders (G-CSF, IFN-γ, TNF-α) correlated with both egress of circulating virus-specific TH1 cells and worse symptoms. CONCLUSIONS Rhinovirus induces robust TH1 responses in allergic asthmatic subjects that may promote disease, even after the infection resolves.
Collapse
Affiliation(s)
- Lyndsey M Muehling
- Department of Medicine, University of Virginia School of Medicine, Charlottesville; Department of Microbiology, University of Virginia School of Medicine, Charlottesville
| | - Peter W Heymann
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville
| | - Paul W Wright
- Department of Medicine, University of Virginia School of Medicine, Charlottesville
| | - Jacob D Eccles
- Department of Medicine, University of Virginia School of Medicine, Charlottesville; Department of Microbiology, University of Virginia School of Medicine, Charlottesville
| | - Rachana Agrawal
- Department of Medicine, University of Virginia School of Medicine, Charlottesville
| | - Holliday T Carper
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville
| | - Deborah D Murphy
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville
| | - Lisa J Workman
- Department of Medicine, University of Virginia School of Medicine, Charlottesville
| | - Carolyn R Word
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville
| | - Sarah J Ratcliffe
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville
| | - Brian J Capaldo
- Department of Microbiology, University of Virginia School of Medicine, Charlottesville
| | | | - Ronald B Turner
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville
| | | | - Judith A Woodfolk
- Department of Medicine, University of Virginia School of Medicine, Charlottesville; Department of Microbiology, University of Virginia School of Medicine, Charlottesville.
| |
Collapse
|
42
|
Zhang X, Zhang X, Zhang N, Wang X, Sun L, Chen N, Zhao S, He Q. Airway microbiome, host immune response and recurrent wheezing in infants with severe respiratory syncytial virus bronchiolitis. Pediatr Allergy Immunol 2020; 31:281-289. [PMID: 31788862 DOI: 10.1111/pai.13183] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 11/03/2019] [Accepted: 11/25/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Early interactions between respiratory viruses and microbiota might modulate host immune responses and subsequently contribute to later development of recurrent wheezing and asthma in childhood. We aimed to study the possible association between respiratory microbiome, host immune response, and the development of recurrent wheezing in infants with severe respiratory syncytial virus (RSV) bronchiolitis. METHODS Seventy-four infants who were hospitalized at Beijing Children's Hospital during an initial episode of severe RSV bronchiolitis at 6 months of age or less were included and followed up until the age of 3 years. Sputum samples were collected, and their microbiota profiles, LPS, and cytokines were analyzed by 16S rRNA-based sequencing, ELISA, and multiplex immunoassay, respectively. RESULTS Twenty-six (35.1%) infants developed recurrent wheezing by the age of 3 years, and 48 (64.9%) did not. The relative abundance of Haemophilus, Moraxella, and Klebsiella was higher in infants who later developed recurrent wheezing than in those who did not (LDA score >3.5). Airway levels of LPS (P = .003), CXCL8 (P = .004), CCL5 (P = .029), IL-6 (P = .004), and IL-13 (P < .001) were significantly higher in infants who later developed recurrent wheezing than in those who did not. Moreover, high airway abundance of Haemophilus was associated with CXCL8 (r = 0.246, P = .037) level, and that of Moraxella was associated with IL-6 level (r = 0.236, P = .046) and IL-10 level (r = 0.266, P = .024). CONCLUSION Our study suggests that higher abundance of Haemophilus and Moraxella in airway microbiome might modulate airway inflammation during severe RSV bronchiolitis in infancy, potentially contributing to the development of subsequent recurrent wheezing in later childhood.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Department of Medical Microbiology, Capital Medical University, Beijing, China.,Department of Respiratory Medicine, Beijing Children's Hospital, Beijing, China
| | - Xiang Zhang
- Department of Respiratory Medicine, Beijing Children's Hospital, Beijing, China
| | - Nan Zhang
- Department of Medical Microbiology, Capital Medical University, Beijing, China
| | - Xinglan Wang
- Department of Respiratory Medicine, Beijing Children's Hospital, Beijing, China
| | - Lin Sun
- Department of Medical Microbiology, Capital Medical University, Beijing, China
| | - Ning Chen
- Department of Medical Microbiology, Capital Medical University, Beijing, China
| | - Shunying Zhao
- Department of Respiratory Medicine, Beijing Children's Hospital, Beijing, China
| | - Qiushui He
- Department of Medical Microbiology, Capital Medical University, Beijing, China.,Department of Medical Microbiology and Immunology, University of Turku, Turku, Finland
| |
Collapse
|
43
|
Gilles S, Blume C, Wimmer M, Damialis A, Meulenbroek L, Gökkaya M, Bergougnan C, Eisenbart S, Sundell N, Lindh M, Andersson L, Dahl Å, Chaker A, Kolek F, Wagner S, Neumann AU, Akdis CA, Garssen J, Westin J, Land B, Davies DE, Traidl‐Hoffmann C. Pollen exposure weakens innate defense against respiratory viruses. Allergy 2020; 75:576-587. [PMID: 31512243 DOI: 10.1111/all.14047] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 06/13/2019] [Accepted: 06/24/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND Hundreds of plant species release their pollen into the air every year during early spring. During that period, pollen allergic as well as non-allergic patients frequently present to doctors with severe respiratory tract infections. Our objective was therefore to assess whether pollen may interfere with antiviral immunity. METHODS We combined data from real-life human exposure cohorts, a mouse model and human cell culture to test our hypothesis. RESULTS Pollen significantly diminished interferon-λ and pro-inflammatory chemokine responses of airway epithelia to rhinovirus and viral mimics and decreased nuclear translocation of interferon regulatory factors. In mice infected with respiratory syncytial virus, co-exposure to pollen caused attenuated antiviral gene expression and increased pulmonary viral titers. In non-allergic human volunteers, nasal symptoms were positively correlated with airborne birch pollen abundance, and nasal birch pollen challenge led to downregulation of type I and -III interferons in nasal mucosa. In a large patient cohort, numbers of rhinoviruspositive cases were correlated with airborne birch pollen concentrations. CONCLUSION The ability of pollen to suppress innate antiviral immunity, independent of allergy, suggests that high-risk population groups should avoid extensive outdoor activities when pollen and respiratory virus seasons coincide.
Collapse
Affiliation(s)
- Stefanie Gilles
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
| | - Cornelia Blume
- Faculty of Medicine Academic Unit of Clinical and Experimental Sciences University of Southampton Southampton UK
- Southampton NIHR Respiratory Biomedical Research Unit University Hospital Southampton Southampton UK
| | - Maria Wimmer
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
- Division of Pharmacology Department of Pharmaceutical Sciences Faculty of Science Utrecht University Utrecht The Netherlands
| | - Athanasios Damialis
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
| | - Laura Meulenbroek
- Division of Pharmacology Department of Pharmaceutical Sciences Faculty of Science Utrecht University Utrecht The Netherlands
- Department of Immunology Nutricia Research Utrecht The Netherlands
| | - Mehmet Gökkaya
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
| | - Carolin Bergougnan
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
| | - Selina Eisenbart
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
| | - Nicklas Sundell
- Department of Infectious Diseases/Clinical Virology University of Gothenburg Gothenburg Sweden
| | - Magnus Lindh
- Department of Infectious Diseases/Clinical Virology University of Gothenburg Gothenburg Sweden
| | - Lars‐Magnus Andersson
- Department of Infectious Diseases/Clinical Virology University of Gothenburg Gothenburg Sweden
| | - Åslög Dahl
- Department of Biological and Environmental Sciences Faculty of Sciences University of Gothenburg Gothenburg Sweden
| | - Adam Chaker
- ENT Department Klinikum Rechts der Isar Technical University of Munich Munich Germany
| | - Franziska Kolek
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
| | - Sabrina Wagner
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
| | - Avidan U. Neumann
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University Zurich Davos Switzerland
- Christine‐Kühne‐Center for Allergy Research and Education (CK‐Care) Davos Switzerland
| | - Johan Garssen
- Division of Pharmacology Department of Pharmaceutical Sciences Faculty of Science Utrecht University Utrecht The Netherlands
- Department of Immunology Nutricia Research Utrecht The Netherlands
| | - Johan Westin
- Department of Infectious Diseases/Clinical Virology University of Gothenburg Gothenburg Sweden
| | - Belinda Land
- Department of Immunology Nutricia Research Utrecht The Netherlands
- Laboratory of Translational Immunology The Wilhelmina Children's Hospital University Medical Center Utrecht Utrecht The Netherlands
| | - Donna E. Davies
- Faculty of Medicine Academic Unit of Clinical and Experimental Sciences University of Southampton Southampton UK
- Southampton NIHR Respiratory Biomedical Research Unit University Hospital Southampton Southampton UK
| | - Claudia Traidl‐Hoffmann
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
- Christine‐Kühne‐Center for Allergy Research and Education (CK‐Care) Davos Switzerland
| |
Collapse
|
44
|
Ullah MA, Vicente CT, Collinson N, Curren B, Sikder MAA, Sebina I, Simpson J, Varelias A, Lindquist JA, Ferreira MAR, Phipps S. PAG1 limits allergen-induced type 2 inflammation in the murine lung. Allergy 2020; 75:336-345. [PMID: 31321783 DOI: 10.1111/all.13991] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/30/2019] [Accepted: 06/24/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Phosphoprotein associated with glycosphingolipid-enriched microdomains 1 (PAG1) is a transmembrane adaptor protein that affects immune receptor signaling in T and B cells. Evidence from genome-wide association studies of asthma suggests that genetic variants that regulate the expression of PAG1 are associated with asthma risk. However, it is not known whether PAG1 expression is causally related to asthma pathophysiology. Here, we investigated the role of PAG1 in a preclinical mouse model of house dust mite (HDM)-induced allergic sensitization and allergic airway inflammation. METHODS Pag1-deficient (Pag1-/- ) and wild-type (WT) mice were sensitized or sensitized/challenged to HDM, and hallmark features of allergic inflammation were assessed. The contribution of T cells was assessed through depletion (anti-CD4 antibody) and adoptive transfer studies. RESULTS Type 2 inflammation (eosinophilia, eotaxin-2 expression, IL-4/IL-5/IL-13 production, mucus production) in the airways and lungs was significantly increased in HDM sensitized/challenged Pag1-/- mice compared to WT mice. The predisposition to allergic sensitization was associated with increased airway epithelial high-mobility group box 1 (HMGB1) translocation and release, increased type 2 innate lymphoid cells (ILC2s) and monocyte-derived dendritic cell numbers in the mediastinal lymph nodes, and increased T-helper type 2 (TH 2)-cell differentiation. CD4+ T-cell depletion studies or the adoptive transfer of WT OVA-specific CD4+ T cells to WT or Pag1-/- recipients demonstrated that the heightened propensity for TH 2-cell differentiation was both T cell intrinsic and extrinsic. CONCLUSION PAG1 deficiency increased airway epithelial activation, ILC2 expansion, and TH 2 differentiation. As a consequence, PAG1 deficiency predisposed toward allergic sensitization and increased the severity of experimental asthma.
Collapse
Affiliation(s)
- Md Ashik Ullah
- QIMR Berghofer Medical Research Institute Brisbane Qld Australia
- Faculty of Medicine University of Queensland Brisbane Qld Australia
| | - Cristina T. Vicente
- QIMR Berghofer Medical Research Institute Brisbane Qld Australia
- Faculty of Medicine University of Queensland Brisbane Qld Australia
| | | | - Bodie Curren
- QIMR Berghofer Medical Research Institute Brisbane Qld Australia
- Faculty of Medicine University of Queensland Brisbane Qld Australia
| | - Md Al Amin Sikder
- QIMR Berghofer Medical Research Institute Brisbane Qld Australia
- Faculty of Medicine University of Queensland Brisbane Qld Australia
| | - Ismail Sebina
- QIMR Berghofer Medical Research Institute Brisbane Qld Australia
| | - Jennifer Simpson
- QIMR Berghofer Medical Research Institute Brisbane Qld Australia
- Faculty of Medicine University of Queensland Brisbane Qld Australia
| | - Antiopi Varelias
- QIMR Berghofer Medical Research Institute Brisbane Qld Australia
- Faculty of Medicine University of Queensland Brisbane Qld Australia
| | - Jonathan A. Lindquist
- Clinic for Nephrology and Hypertension, Diabetology and Endocrinology Otto‐von‐Guericke University Magdeburg Germany
| | | | - Simon Phipps
- QIMR Berghofer Medical Research Institute Brisbane Qld Australia
- Faculty of Medicine University of Queensland Brisbane Qld Australia
- Australian Infectious Diseases Research Centre University of Queensland Brisbane Qld Australia
| |
Collapse
|
45
|
Lee H, Chen VCH, Yang YH, Kuo TY, Lin TC, Wu SI, Kao KL, Weng JC, Kelsen BA, Liang SHY. Decreased Risk of Influenza in Child and Adolescent Patients with Attention-Deficit Hyperactivity Disorder Following Methylphenidate Treatment: A Nationwide Cohort Study in Taiwan. Neuropsychiatr Dis Treat 2020; 16:1309-1319. [PMID: 32547034 PMCID: PMC7247598 DOI: 10.2147/ndt.s242519] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/22/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Young individuals with attention-deficit hyperactivity disorder (ADHD) may have an elevated risk of influenza because of the difficulty in complying with the behavioral procedures that help protect against influenza. Moreover, the effects of sufficient methylphenidate treatment on influenza have received little attention. OBJECTIVE This study evaluated the association between ADHD medication usage and influenza and assessed the effect of duration of ADHD treatment on the risk of influenza using a nationwide population-based database. METHODS This study investigated methylphenidate usage and the risk of influenza among children and adolescents with ADHD. We identified 5259 young individuals aged less than 18 years who were diagnosed as having ADHD between 1996 and 2013 from the National Health Insurance Research Database of Taiwan, and we tested whether methylphenidate use affects influenza risk using Cox proportional hazard models. RESULTS After controlling for confounding factors, the results indicated that influenza risk significantly reduced in the group of ADHD patients who were prescribed methylphenidate for 90 days and more (hazard ratio [HR]: 0.62, 95% confidence interval [CI]: 0.52-0.75, p<0.001), demonstrating a 38% reduction in the risk of influenza in this group. However, this was not observed in the group of ADHD patients who used methylphenidate for 1-90 days (HR: 0.69, 95% CI: 0.89-1.05, p=0.12). CONCLUSION The lower incidence of influenza observed in the group prescribed with methylphenidate for a longer period highlights the importance of compliance to medication and psychoeducation with regard to ADHD management.
Collapse
Affiliation(s)
- Hsuan Lee
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Vincent Chin-Hung Chen
- Department of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Psychiatry, Chang Gung Memorial Hospital at Chiayi,Chiayi,Taiwan
| | - Yao-Hsu Yang
- Health Information and Epidemiology Laboratory of Chang Gung Memorial Hospital, Chiayi Branch, Chiayi,Taiwan.,Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital,Taiwan.,School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ting-Yu Kuo
- Health Information and Epidemiology Laboratory of Chang Gung Memorial Hospital, Chiayi Branch, Chiayi,Taiwan
| | - Tzu-Chin Lin
- Bethel Psychiatric Clinic, Taipei, Taiwan.,Institute of Science, Technology and Society, National Yang-Ming University Taipei, Taiwan
| | - Shu-I Wu
- Mackay Memorial Hospital, Section of Psychiatry, Taipei, Taiwan.,Department of Medicine, Mackay Medical College, Taipei, Taiwan
| | - Kai-Liang Kao
- Department of Pediatrics, Far Eastern Memorial Hospital, Taipei, Taiwan.,Department of Industrial Management, Oriental Institute of Technology, Taipei, Taiwan
| | - Jun-Cheng Weng
- Department of Psychiatry, Chang Gung Memorial Hospital at Chiayi,Chiayi,Taiwan.,Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Brent Allan Kelsen
- Language Center, National Taipei University, New Taipei City, Taiwan.,Department of Psychology, Auckland University of Technology, Auckland, New Zealand
| | - Sophie Hsin-Yi Liang
- Department of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Child Psychiatry, Chang Gung Memorial Hospital at Taoyuan, Taoyuan, Taiwan
| |
Collapse
|
46
|
Donovan C, Hansbro PM. IL-33 in Chronic Respiratory Disease: From Preclinical to Clinical Studies. ACS Pharmacol Transl Sci 2019; 3:56-62. [PMID: 32259088 DOI: 10.1021/acsptsci.9b00099] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Indexed: 01/06/2023]
Abstract
IL-33 has been deorphanized as a member of the IL-1 family and has key roles as an alarmin and cytokine with potent capacity to drive type 2 inflammation. This has led to a plethora of studies surrounding its role in chronic diseases with a type 2 inflammatory component. Here, we review the roles of IL-33 in two chronic respiratory diseases, asthma and chronic obstructive pulmonary disease (COPD). We discuss the hallmark and paradigm-shifting studies that have contributed to our understanding of IL-33 biology. We cover animal studies that have elucidated the mechanisms of IL-33 and assessed the role of anti-IL-33 treatment and immunization against IL-33. We highlight key clinical evidence for the potential of targeting increased IL-33 in respiratory diseases including exacerbations, and we outline current clinical trials using an anti-IL-33 monoclonal antibody in asthma patients. Finally, we discuss some of the challenges that have arisen in IL-33 biology and highlight potential future directions in targeting this cytokine in chronic respiratory diseases.
Collapse
Affiliation(s)
- Chantal Donovan
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, New South Wales 2050, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales 2308, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, Sydney, New South Wales 2050, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales 2308, Australia
| |
Collapse
|
47
|
Werder RB, Lynch JP, Simpson JC, Zhang V, Hodge NH, Poh M, Forbes-Blom E, Kulis C, Smythe ML, Upham JW, Spann K, Everard ML, Phipps S. PGD2/DP2 receptor activation promotes severe viral bronchiolitis by suppressing IFN- λ production. Sci Transl Med 2019; 10:10/440/eaao0052. [PMID: 29743346 DOI: 10.1126/scitranslmed.aao0052] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 11/17/2017] [Accepted: 03/12/2018] [Indexed: 12/27/2022]
Abstract
Prostaglandin D2 (PGD2) signals through PGD2 receptor 2 (DP2, also known as CRTH2) on type 2 effector cells to promote asthma pathogenesis; however, little is known about its role during respiratory syncytial virus (RSV) bronchiolitis, a major risk factor for asthma development. We show that RSV infection up-regulated hematopoietic prostaglandin D synthase expression and increased PGD2 release by cultured human primary airway epithelial cells (AECs). Moreover, PGD2 production was elevated in nasopharyngeal samples from young infants hospitalized with RSV bronchiolitis compared to healthy controls. In a neonatal mouse model of severe viral bronchiolitis, DP2 antagonism decreased viral load, immunopathology, and morbidity and ablated the predisposition for subsequent asthma onset in later life. This protective response was abolished upon dual DP1/DP2 antagonism and replicated with a specific DP1 agonist. Rather than mediating an effect via type 2 inflammation, the beneficial effects of DP2 blockade or DP1 agonism were associated with increased interferon-λ (IFN-λ) [interleukin-28A/B (IL-28A/B)] expression and were lost upon IL-28A neutralization. In RSV-infected AEC cultures, DP1 activation up-regulated IFN-λ production, which, in turn, increased IFN-stimulated gene expression, accelerating viral clearance. Our findings suggest that DP2 antagonists or DP1 agonists may be useful antivirals for the treatment of viral bronchiolitis and possibly as primary preventatives for asthma.
Collapse
Affiliation(s)
- Rhiannon B Werder
- School of Biomedical Sciences, University of Queensland, Queensland 4072, Australia
| | - Jason P Lynch
- School of Biomedical Sciences, University of Queensland, Queensland 4072, Australia
| | - Jennifer C Simpson
- School of Biomedical Sciences, University of Queensland, Queensland 4072, Australia.,Queensland Institute of Medical Research Berghofer Medical Research Institute, Herston 4006, Australia
| | - Vivian Zhang
- Queensland Institute of Medical Research Berghofer Medical Research Institute, Herston 4006, Australia
| | - Nick H Hodge
- School of Biomedical Sciences, University of Queensland, Queensland 4072, Australia
| | - Matthew Poh
- School of Paediatrics and Child Health, University of Western Australia, Western Australia 6840, Australia
| | | | - Christina Kulis
- Institute for Molecular Bioscience, University of Queensland, Queensland 4072, Australia
| | - Mark L Smythe
- Institute for Molecular Bioscience, University of Queensland, Queensland 4072, Australia
| | - John W Upham
- Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Queensland 4102, Australia
| | - Kirsten Spann
- Australian Infectious Diseases Research Centre, University of Queensland, Queensland 4067, Australia.,School of Biomedical Sciences, Queensland University of Technology, Queensland 4001, Australia
| | - Mark L Everard
- School of Paediatrics and Child Health, University of Western Australia, Western Australia 6840, Australia
| | - Simon Phipps
- Queensland Institute of Medical Research Berghofer Medical Research Institute, Herston 4006, Australia. .,Australian Infectious Diseases Research Centre, University of Queensland, Queensland 4067, Australia
| |
Collapse
|
48
|
Johansson K, McSorley HJ. Interleukin-33 in the developing lung-Roles in asthma and infection. Pediatr Allergy Immunol 2019; 30:503-510. [PMID: 30734382 DOI: 10.1111/pai.13040] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/06/2019] [Accepted: 01/08/2019] [Indexed: 02/07/2023]
Abstract
It has become increasingly clear that interleukin-33 (IL-33) plays a crucial role in initiation of type 2 immunity. The last decade of intense research has uncovered multiple mechanisms through which IL-33 targets key effector cells of the allergic immune response. Recently, IL-33 has been implicated in shaping the immune system of the lungs early in life, at a time which is crucial in the subsequent development of allergic asthma. In this review, we will address the current literature describing the role of IL-33 in the healthy and diseased lung. In particular, we will focus on the evidence for IL-33 in the development of immune responses in the lung, including the role of IL-33-responsive immune cells that may explain susceptibility to allergic sensitization at a young age and the association between genetic variants of IL-33 and asthma in humans. Finally, we will indicate areas for potential therapeutic modulation of the IL-33 pathway.
Collapse
Affiliation(s)
- Kristina Johansson
- Department of Microbiology and Immunology, Sandler Asthma Basic Research Center, University of California, San Francisco, California.,Department of Medicine, University of California, San Francisco, California
| | - Henry J McSorley
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
49
|
Fardisi M, Gondhalekar AD, Ashbrook AR, Scharf ME. Rapid evolutionary responses to insecticide resistance management interventions by the German cockroach (Blattella germanica L.). Sci Rep 2019; 9:8292. [PMID: 31165746 PMCID: PMC6549143 DOI: 10.1038/s41598-019-44296-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 05/14/2019] [Indexed: 11/12/2022] Open
Abstract
The German cockroach (Blattella germanica L.) is a worldwide pest that lives exclusively in human environments. B. germanica threatens human health by producing asthma-triggering allergens, vectoring pathogenic/antibiotic-resistant microbes, and by contributing to unhealthy indoor environments. While insecticides are essential for reducing cockroach populations and improving health outcomes, insecticide resistance has been a consistent barrier to cockroach control since the 1950s. We conducted seminal field studies to compare three insecticide resistance intervention strategies for cockroaches and evaluated resistance evolution across multiple generations. Using pre-treatment resistance assessment to drive decisions, we found that single active ingredient (AI) treatments can successfully eliminate cockroaches if starting resistance levels are low. We further established that rotation treatments intuitively reduce selection pressure, and are effective when insecticides with no/low resistance are used. We also found that mixture products containing thiamethoxam + λ-cyhalothrin AIs were universally ineffective and highly repellent; and finally, evolution of cross-resistance among AIs is a significant, previously unrealized challenge.
Collapse
Affiliation(s)
- Mahsa Fardisi
- Purdue University, Department of Entomology, West Lafayette, IN, 47907, USA
| | | | - Aaron R Ashbrook
- Purdue University, Department of Entomology, West Lafayette, IN, 47907, USA
| | - Michael E Scharf
- Purdue University, Department of Entomology, West Lafayette, IN, 47907, USA.
| |
Collapse
|
50
|
Hijano DR, Vu LD, Kauvar LM, Tripp RA, Polack FP, Cormier SA. Role of Type I Interferon (IFN) in the Respiratory Syncytial Virus (RSV) Immune Response and Disease Severity. Front Immunol 2019; 10:566. [PMID: 30972063 PMCID: PMC6443902 DOI: 10.3389/fimmu.2019.00566] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/04/2019] [Indexed: 12/22/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the most common cause of lower respiratory tract disease in children <2 years of age. Increased morbidity and mortality have been reported in high-risk patients, such as premature infants, patients with cardiac disease, and severely immune compromised patients. Severe disease is associated with the virulence of the virus as well as host factors specifically including the innate immune response. The role of type I interferons (IFNs) in the response to RSV infection is important in regulating the rate of virus clearance and in directing the character of the immune response, which is normally associated with protection and less severe disease. Two RSV non-structural proteins, NS1 and NS2, as well as the envelope G glycoprotein are known to suppress type I IFN production and a robust type I IFN response to RSV does not occur in human infants or neonatal mouse models of RSV infection. Additionally, presence of type I IFNs are associated with mild symptoms in infants and administration of IFN-α prior to infection of neonatal mice with RSV reduces immunopathology. This evidence has driven RSV prophylaxis and therapeutic efforts to consider strategies for enhancing type I IFN production.
Collapse
Affiliation(s)
- Diego R Hijano
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, United States
| | - Luan D Vu
- Department of Biological Sciences, Louisiana State University and School of Veterinary Medicine, Baton Rouge, LA, United States.,Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | | | - Ralph A Tripp
- Department of Infectious Disease, University of Georgia, Athens, GA, United States
| | | | - Stephania A Cormier
- Department of Biological Sciences, Louisiana State University and School of Veterinary Medicine, Baton Rouge, LA, United States.,Department of Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| |
Collapse
|