51
|
Williams TC, Loo SL, Nichol KS, Reid AT, Veerati PC, Esneau C, Wark PAB, Grainge CL, Knight DA, Vincent T, Jackson CL, Alton K, Shimkets RA, Girkin JL, Bartlett NW. IL-25 blockade augments antiviral immunity during respiratory virus infection. Commun Biol 2022; 5:415. [PMID: 35508632 PMCID: PMC9068710 DOI: 10.1038/s42003-022-03367-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 04/13/2022] [Indexed: 12/12/2022] Open
Abstract
IL-25 is implicated in the pathogenesis of viral asthma exacerbations. However, the effect of IL-25 on antiviral immunity has yet to be elucidated. We observed abundant expression and colocalization of IL-25 and IL-25 receptor at the apical surface of uninfected airway epithelial cells and rhinovirus infection increased IL-25 expression. Analysis of immune transcriptome of rhinovirus-infected differentiated asthmatic bronchial epithelial cells (BECs) treated with an anti-IL-25 monoclonal antibody (LNR125) revealed a re-calibrated response defined by increased type I/III IFN and reduced expression of type-2 immune genes CCL26, IL1RL1 and IL-25 receptor. LNR125 treatment also increased type I/III IFN expression by coronavirus infected BECs. Exogenous IL-25 treatment increased viral load with suppressed innate immunity. In vivo LNR125 treatment reduced IL-25/type 2 cytokine expression and increased IFN-β expression and reduced lung viral load. We define a new immune-regulatory role for IL-25 that directly inhibits virus induced airway epithelial cell innate anti-viral immunity.
Collapse
Affiliation(s)
- Teresa C Williams
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Su-Ling Loo
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Kristy S Nichol
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Andrew T Reid
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Punnam C Veerati
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Camille Esneau
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Peter A B Wark
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Christopher L Grainge
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Darryl A Knight
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
- UBC Providence Health Care Research Institute, Vancouver, BC, Canada
- Department of Anaesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Thomas Vincent
- Abeome Corporation/Lanier Biotherapeutics, Athens, GA, USA
| | | | - Kirby Alton
- Abeome Corporation/Lanier Biotherapeutics, Athens, GA, USA
| | | | - Jason L Girkin
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Nathan W Bartlett
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia.
| |
Collapse
|
52
|
Liew KY, Koh SK, Hooi SL, Ng MKL, Chee HY, Harith HH, Israf DA, Tham CL. Rhinovirus-Induced Cytokine Alterations With Potential Implications in Asthma Exacerbations: A Systematic Review and Meta-Analysis. Front Immunol 2022; 13:782936. [PMID: 35242128 PMCID: PMC8886024 DOI: 10.3389/fimmu.2022.782936] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 01/13/2022] [Indexed: 12/01/2022] Open
Abstract
Background Rhinovirus (RV) infections are a major cause of asthma exacerbations. Unlike other respiratory viruses, RV causes minimal cytotoxic effects on airway epithelial cells and cytokines play a critical role in its pathogenesis. However, previous findings on RV-induced cytokine responses were largely inconsistent. Thus, this study sought to identify the cytokine/chemokine profiles induced by RV infection and their correlations with airway inflammatory responses and/or respiratory symptoms using systematic review, and to determine whether a quantitative difference exists in cytokine levels between asthmatic and healthy individuals via meta-analysis. Methods Relevant articles were obtained from PubMed, Scopus, and ScienceDirect databases. Studies that compared RV-induced cytokine responses between asthmatic and healthy individuals were included in the systematic review, and their findings were categorized based on the study designs, which were ex vivo primary bronchial epithelial cells (PBECs), ex vivo peripheral blood mononuclear cells (PBMCs), and human experimental studies. Data on cytokine levels were also extracted and analyzed using Review Manager 5.4. Results Thirty-four articles were included in the systematic review, with 18 of these further subjected to meta-analysis. Several studies reported the correlations between the levels of cytokines, such as IL-8, IL-4, IL-5, and IL-13, and respiratory symptoms. Evidence suggests that IL-25 and IL-33 may be the cytokines that promote type 2 inflammation in asthmatics after RV infection. Besides that, a meta-analysis revealed that PBECs from children with atopic asthma produced significantly lower levels of IFN-β [Effect size (ES): -0.84, p = 0.030] and IFN-λ (ES: -1.00, p = 0.002), and PBECs from adult atopic asthmatics produced significantly lower levels of IFN-β (ES: -0.68, p = 0.009), compared to healthy subjects after RV infection. A trend towards a deficient production of IFN-γ (ES: -0.56, p = 0.060) in PBMCs from adult atopic asthmatics was observed. In lower airways, asthmatics also had significantly lower baseline IL-15 (ES: -0.69, p = 0.020) levels. Conclusion Overall, RV-induced asthma exacerbations are potentially caused by an imbalance between Th1 and Th2 cytokines, which may be contributed by defective innate immune responses at cellular levels. Exogenous IFNs delivery may be beneficial as a prophylactic approach for RV-induced asthma exacerbations. Systematic Review Registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=184119, identifier CRD42020184119.
Collapse
Affiliation(s)
- Kong Yen Liew
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Sue Kie Koh
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Suet Li Hooi
- School of Science, Monash University Malaysia, Subang Jaya, Malaysia
| | | | - Hui-Yee Chee
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Hanis Hazeera Harith
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Daud Ahmad Israf
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Chau Ling Tham
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
53
|
Robert M, Miossec P, Hot A. The Th17 Pathway in Vascular Inflammation: Culprit or Consort? Front Immunol 2022; 13:888763. [PMID: 35479069 PMCID: PMC9035791 DOI: 10.3389/fimmu.2022.888763] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
The involvement of IL-17A in autoimmune and inflammatory diseases has prompted the development of therapeutic strategies to block the Th17 pathway. Promising results came from their use in psoriasis and in ankylosing spondylitis. IL-17A acts on various cell types and has both local and systemic effects. Considering the premature mortality observed during chronic inflammatory diseases, IL-17A action on vascular cells was studied. Both in vitro and in vivo results suggest that this cytokine favors inflammation, coagulation and thrombosis and promotes the occurrence of cardiovascular events. These observations led to study the role of IL-17A in diseases characterized by vascular inflammation, namely allograft rejection and vasculitis. Increased circulating levels of IL-17A and histological staining reveal that the Th17 pathway is involved in the pathogenesis of these diseases. Vasculitis treatment faces challenges while the use of steroids has many side effects. Regarding results obtained in giant cell arteritis with IL-6 inhibitors, a cytokine involved in Th17 differentiation, the use of anti-IL-17 is a promising strategy. However, lessons from rheumatoid arthritis and multiple sclerosis must be learnt before targeting IL-17 in vasculitis, which may be culprit, consort or both of them.
Collapse
Affiliation(s)
- Marie Robert
- Department of Clinical Immunology and Rheumatology, and Immunogenomics and Inflammation Research Unit, University of Lyon, Hôpital Edouard Herriot, Lyon, France
- Department of Internal Medicine, University of Lyon, Hôpital Edouard Herriot, Lyon, France
- *Correspondence: Marie Robert,
| | - Pierre Miossec
- Department of Clinical Immunology and Rheumatology, and Immunogenomics and Inflammation Research Unit, University of Lyon, Hôpital Edouard Herriot, Lyon, France
| | - Arnaud Hot
- Department of Internal Medicine, University of Lyon, Hôpital Edouard Herriot, Lyon, France
| |
Collapse
|
54
|
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, the MRC‐GSK Strategic Alliance Consortium. 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
|
55
|
Feng X, Lawrence MG, Payne SC, Mattos J, Etter E, Negri JA, Murphy D, Kennedy JL, Steinke JW, Borish L. Lower viral loads in subjects with rhinovirus-challenged allergy despite reduced innate immunity. Ann Allergy Asthma Immunol 2022; 128:414-422.e2. [PMID: 35031416 PMCID: PMC10666001 DOI: 10.1016/j.anai.2022.01.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/04/2021] [Accepted: 01/05/2022] [Indexed: 02/02/2023]
Abstract
BACKGROUND Viral infections, especially those caused by rhinovirus, are the most common cause of asthma exacerbations. Previous studies have argued that impaired innate antiviral immunity and, as a consequence, more severe infections contribute to these exacerbations. OBJECTIVE These studies explored the innate immune response in the upper airway of volunteers with allergic rhinitis and asthma in comparison to healthy controls and interrogated how these differences corresponded to severity of infection. METHODS Volunteers with allergic rhinitis, those with asthma, and those who are healthy were inoculated with rhinovirus A16 and monitored for clinical symptoms. Tissue and nasal wash samples were evaluated for antiviral signature and viral load. RESULTS Both subjects with allergic rhinitis and asthma were found to have more severe cold symptoms. Subjects with asthma had worsened asthma control and increased bronchial hyperreactivity in the setting of higher fractional exhaled breath nitric oxide and blood eosinophils. These studies confirmed reduced expression of interferons and virus-specific pattern recognition receptors in both cohorts with atopy. Nevertheless, despite this defect in innate immunity, volunteers with allergic rhinitis/asthma had reduced rhinovirus concentrations in comparison to the controls. CONCLUSION These results confirm that the presence of an allergic inflammatory disorder of the airway is associated with reduced innate immune responsive to rhinovirus infection. Despite this, these volunteers with allergy have reduced viral loads, arguing for the presence of a compensatory mechanism to clear the infection. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02910401.
Collapse
Affiliation(s)
- Xin Feng
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, NHC Key Laboratory of Otorhinolaryngology (Shandong University), Jinan, Shandong, People's Republic of China
| | - Monica G Lawrence
- Department of Medicine, University of Virginia Health System, Charlottesville, Virginia; Department of Pediatrics, University of Virginia Health System, Charlottesville, Virginia
| | - Spencer C Payne
- Department of Otolaryngology, University of Virginia Health System, Charlottesville, Virginia
| | - Jose Mattos
- Department of Otolaryngology, University of Virginia Health System, Charlottesville, Virginia
| | - Elaine Etter
- Department of Medicine, University of Virginia Health System, Charlottesville, Virginia
| | - Julie A Negri
- Department of Medicine, University of Virginia Health System, Charlottesville, Virginia
| | - Deborah Murphy
- Department of Pediatrics, University of Virginia Health System, Charlottesville, Virginia
| | - Joshua L Kennedy
- Department of Pediatrics and Medicine, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, Arkansas
| | - John W Steinke
- Department of Medicine, University of Virginia Health System, Charlottesville, Virginia
| | - Larry Borish
- Department of Medicine, University of Virginia Health System, Charlottesville, Virginia; Department of Microbiology, University of Virginia Health System, Charlottesville, Virginia.
| |
Collapse
|
56
|
Aydin Guclu O, Goktas SS, Gorek Dilektasli A, Acet Ozturk NA, Demirdogen E, Coskun F, Ediger D, Ursavas A, Uzaslan E, Erol HA, Karacay ND, Kaya Sel U, Karadag M. A Pilot Study for IgE as a Prognostic Biomarker in COVID-19. Intern Med J 2022; 52:1495-1504. [PMID: 35257465 PMCID: PMC9111421 DOI: 10.1111/imj.15728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 02/14/2022] [Accepted: 02/27/2022] [Indexed: 01/08/2023]
Abstract
Background Laboratory biomarkers to estimate the severity of COVID‐19 are crucial during the pandemic, since resource allocation must be carefully planned. Aim In the present study, we aim to evaluate the effects of basal serum total immunoglobulin E (IgE) levels and changes in inflammatory parameters on the clinical progression of patients hospitalized with COVID‐19. Methods Patients hospitalized with confirmed COVID‐19 were included in the study. Laboratory data and total IgE levels were measured upon admission. Lymphocyte, eosinophil, ferritin, d‐dimer and CRP parameters were recorded on the baseline and on the 3rd and 14th days of hospitalization. Results The study enrolled 202 patients, of which 102(50.5%) were males. The average age was 50.17 ± 19.68. Of the COVID‐19 patients, 41 (20.3%) showed clinical progression. Serum total IgE concentrations were markedly higher (172.90 [0‐2124] vs 38.70 [0‐912], p<0.001) and serum eosinophil levels were significantly lower (0.015 [0‐1.200] vs 0.040 [0‐1.360], p=0.002) in clinically worsened COVID‐19 patients when compared to stable patients. The optimal cut‐off for predicting clinical worsening was 105.2 ng/L; with 61% sensitivity, 82% specificity, 46.3% positive predictive value and 89.2% negative predictive value (area under the curve=0.729). Multivariable analysis to define risk factors for disease progression identified higher total IgE and CRP levels as independent predictors. Conclusions Our single‐center pilot study determined that total IgE levels may be a negative prognostic factor for clinical progression in patients hospitalized due to COVID‐19 infection. Future studies are required to determine the impact of individuals' underlying immune predispositions on outcomes of COVID‐19 infections. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ozge Aydin Guclu
- Uludag University Faculty of Medicine, Department of Pulmonary Diseases, Bursa, Turkey.,Boyabat 75th Year State Hospital, Department of Pulmonary Disease, Sinop, Turkey
| | - Seda Sezen Goktas
- Boyabat 75th Year State Hospital, Department of Otolaryngology, Sinop, Turkey
| | - Asli Gorek Dilektasli
- Uludag University Faculty of Medicine, Department of Pulmonary Diseases, Bursa, Turkey
| | | | - Ezgi Demirdogen
- Uludag University Faculty of Medicine, Department of Pulmonary Diseases, Bursa, Turkey
| | - Funda Coskun
- Uludag University Faculty of Medicine, Department of Pulmonary Diseases, Bursa, Turkey
| | - Dane Ediger
- Uludag University Faculty of Medicine, Department of Pulmonary Diseases, Section of Immunology and Allergy Diseases, Bursa, Turkey
| | - Ahmet Ursavas
- Uludag University Faculty of Medicine, Department of Pulmonary Diseases, Bursa, Turkey
| | - Esra Uzaslan
- Uludag University Faculty of Medicine, Department of Pulmonary Diseases, Bursa, Turkey
| | - Hasim Atakan Erol
- Kocaeli University Faculty of Medicine, Department of Hematology, Kocaeli, Turkey
| | - Nurdan Derya Karacay
- Kocaeli University Faculty of Medicine, Department of Hematology, Kocaeli, Turkey
| | - Umut Kaya Sel
- Boyabat 75th Year State Hospital, Department of Anesthesiology and Intensive Care, Sinop, Turkey
| | - Mehmet Karadag
- Uludag University Faculty of Medicine, Department of Pulmonary Diseases, Bursa, Turkey
| |
Collapse
|
57
|
Albano GD, Montalbano AM, Gagliardo R, Anzalone G, Profita M. Impact of Air Pollution in Airway Diseases: Role of the Epithelial Cells (Cell Models and Biomarkers). Int J Mol Sci 2022; 23:2799. [PMID: 35269941 PMCID: PMC8911203 DOI: 10.3390/ijms23052799] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/18/2022] [Accepted: 02/26/2022] [Indexed: 02/05/2023] Open
Abstract
Biomedical research is multidisciplinary and often uses integrated approaches performing different experimental models with complementary functions. This approach is important to understand the pathogenetic mechanisms concerning the effects of environmental pollution on human health. The biological activity of the substances is investigated at least to three levels using molecular, cellular, and human tissue models. Each of these is able to give specific answers to experimental problems. A scientific approach, using biological methods (wet lab), cell cultures (cell lines or primary), isolated organs (three-dimensional cell cultures of primary epithelial cells), and animal organisms, including the human body, aimed to understand the effects of air pollution on the onset of diseases of the respiratory system. Biological methods are divided into three complementary models: in vitro, ex vivo, and in vivo. In vitro experiments do not require the use of whole organisms (in vivo study), while ex vivo experiments use isolated organs or parts of organs. The concept of complementarity and the informatic support are useful tools to organize, analyze, and interpret experimental data, with the aim of discussing scientific notions with objectivity and rationality in biology and medicine. In this scenario, the integrated and complementary use of different experimental models is important to obtain useful and global information that allows us to identify the effect of inhaled pollutants on the incidence of respiratory diseases in the exposed population. In this review, we focused our attention on the impact of air pollution in airway diseases with a rapid and descriptive analysis on the role of epithelium and on the experimental cell models useful to study the effect of toxicants on epithelial cells.
Collapse
Affiliation(s)
- Giusy Daniela Albano
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), 00133 Rome, Italy; (G.D.A.); (A.M.M.); (R.G.)
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90100 Palermo, Italy;
| | - Angela Marina Montalbano
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), 00133 Rome, Italy; (G.D.A.); (A.M.M.); (R.G.)
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90100 Palermo, Italy;
| | - Rosalia Gagliardo
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), 00133 Rome, Italy; (G.D.A.); (A.M.M.); (R.G.)
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90100 Palermo, Italy;
| | - Giulia Anzalone
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90100 Palermo, Italy;
| | - Mirella Profita
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), 00133 Rome, Italy; (G.D.A.); (A.M.M.); (R.G.)
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), 90100 Palermo, Italy;
| |
Collapse
|
58
|
Jackson DJ, Gern JE. Rhinovirus Infections and Their Roles in Asthma: Etiology and Exacerbations. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:673-681. [PMID: 35074599 DOI: 10.1016/j.jaip.2022.01.006] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/06/2022] [Accepted: 01/06/2022] [Indexed: 12/17/2022]
Abstract
Rhinovirus infections can cause wheezing illnesses in all age groups. In preschool children, rhinovirus infections frequently initiate acute wheezing illnesses. Children who wheeze with rhinoviruses are at increased risk to go on to develop asthma. Once asthma is established, rhinovirus infections are potent triggers for acute airway obstruction and exacerbations in children and adults. Paradoxically, for most individuals, rhinovirus infections commonly cause cold symptoms with little or no involvement of the lower airways. This paradox has led investigators to identify specific risk factors and mechanisms for rhinovirus wheezing, and this review will outline progress in 3 main areas. First, the 3 species of rhinoviruses have different patterns of infection and virulence. Second, personal factors such as lung function and immunity influence lower respiratory outcomes of rhinovirus infection. The mucosal immune response is critical, and the quality of the interferon response and allergic inflammation interacts to determine the risk for rhinovirus wheezing. Finally, rhinovirus infections can promote pathogen-dominated airway microbiota that increase the risk for wheezing. Although specific antivirals for rhinovirus are still not available, identifying risk factors for wheezing illnesses has provided several other potential targets and strategies for reducing the risk of rhinovirus-induced wheezing and exacerbations of asthma.
Collapse
Affiliation(s)
- David J Jackson
- Guy's Severe Asthma Centre, Guy's & St Thomas' NHS Trust, London, United Kingdom; School of Immunology & Microbial Sciences, King's College London, London, United Kingdom.
| | - James E Gern
- Departments of Pediatrics and Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| |
Collapse
|
59
|
Kawakami T, Kasakura K, Kawakami Y, Ando T. Immunoglobulin E-Dependent Activation of Immune Cells in Rhinovirus-Induced Asthma Exacerbation. FRONTIERS IN ALLERGY 2022; 3:835748. [PMID: 35386658 PMCID: PMC8974681 DOI: 10.3389/falgy.2022.835748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/24/2022] [Indexed: 11/26/2022] Open
Abstract
Acute exacerbation is the major cause of asthma morbidity, mortality, and health-care costs. Respiratory viral infections, particularly rhinovirus (RV) infections, are associated with the majority of asthma exacerbations. The risk for bronchoconstriction with RV is associated with allergic sensitization and type 2 airway inflammation. The efficacy of the humanized anti-IgE monoclonal antibody omalizumab in treating asthma and reducing the frequency and severity of RV-induced asthma exacerbation is well-known. Despite these clinical data, mechanistic details of omalizumab's effects on RV-induced asthma exacerbation have not been well-defined for years due to the lack of appropriate animal models. In this Perspective, we discuss potential IgE-dependent roles of mast cells and dendritic cells in asthma exacerbations.
Collapse
Affiliation(s)
- Toshiaki Kawakami
- Laboratory of Allergic Diseases, Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, CA, United States
- *Correspondence: Toshiaki Kawakami
| | - Kazumi Kasakura
- Laboratory of Allergic Diseases, Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Yu Kawakami
- Laboratory of Allergic Diseases, Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Tomoaki Ando
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
60
|
Lin TL, Fan YH, Chang YL, Ho HJ, Wu CY, Chen YJ. Early-life infections in association with development of atopic dermatitis in infancy and early childhood: A nationwide nested case-control study. J Eur Acad Dermatol Venereol 2022; 36:615-622. [PMID: 35000246 DOI: 10.1111/jdv.17908] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/07/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Microbial dysbiosis has been implicated in the development of atopic dermatitis (AD). The risk of development of AD following early-life infections remains unclear. OBJECTIVE To investigate the impact of early-life infections on AD development. METHODS This population-based nested case-control study was conducted using the Taiwan's National Health Insurance Research Database. A total of 5,454 AD patients and 16,362 control subjects without AD were identified, for the period 1997 to 2013. Demographic characteristics, comorbidities, and maternal factors were compared. Adjusted odds ratio (aOR) was calculated to examine the associations between early-life infections and subsequent AD by conditional stepwise logistic regression analysis. RESULTS Mean age was 2.6±2.9 years in both groups. Overall infections (41.8% vs 28.9%) before the diagnosis of AD were more common in AD patients than in control subjects (p<.001). Infectious diseases (aOR, 1.40; 95% confidence interval [CI], 1.29-1.51), skin infections (aOR, 1.55; 95% CI, 1.40-1.71) and systemic antibiotic exposure (aOR 1.67, 95% CI 1.55-1.79) before AD diagnosis were independently associated with AD development on multivariate analyses. These results were consistent across observation periods (0-1, 1-2, and >2 years after birth) and sensitivity analyses after redefining the index date as 3 or 6 months before the date of AD diagnosis. Other independent risk factors included asthma, allergic rhinitis, intussusception, and neonatal hyperbilirubinemia. No association with subsequent AD was found for maternal age at delivery, Cesarean delivery, or prenatal antibiotic exposure. CONCLUSION Infections in early life are associated with AD development in infancy and early childhood.
Collapse
Affiliation(s)
- Teng-Li Lin
- Department of Dermatology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yi-Hsuan Fan
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Ling Chang
- Department of Dermatology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hsiu J Ho
- Institute of Biomedical Informatics, Institute of Public Health, National Yang-Ming University, Taipei, Taiwan
| | - Chun-Ying Wu
- Institute of Biomedical Informatics, Institute of Public Health, National Yang-Ming University, Taipei, Taiwan.,Faculty of Medicine and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Translational Research and Center of Excellence for Cancer Research, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Public Health, China Medical University, Taichung, Taiwan.,National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Yi-Ju Chen
- Department of Dermatology, Taichung Veterans General Hospital, Taichung, Taiwan.,Faculty of Medicine and Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Translational Medicine, National Chung-Hsing University, Taichung, Taiwan
| |
Collapse
|
61
|
Mikkola H, Honkila M, Tapiainen T, Jartti T. Susceptibility to rhinovirus-induced early wheezing as a risk factor for subsequent asthma development. CURRENT RESPIRATORY MEDICINE REVIEWS 2022. [DOI: 10.2174/1573398x18666220103113813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Abstract:
Rhinovirus is one of the two most common viral agents that cause bronchiolitis in young children. During the first 12 months, it is second to the respiratory syncytial virus, but after 12 months, it begins dominating the statistics. Wheezing and dry cough are typical clinical symptoms indicative of rhinovirus-induced bronchiolitis, although overlap of symptoms with other virus infections is common. Several studies have shown that atopic predisposition and reduced interferon responses increase susceptibility to rhinovirus-induced wheezing. More recent studies have found that certain genetic variations at strong asthma loci also increase susceptibility. Rhinovirus-induced wheezing in the early years of life is known to increase the risk of subsequent asthma development and may be associated with airway remodeling. This risk is increased by aeroallergen sensitization. Currently, there are no clinically approved preventive treatments for asthma. However, studies show promising results indicating that children with rhinovirus-affected first-time wheezing respond to bronchodilators in terms of less short-term symptoms and that controlling airway inflammatory responses with anti-inflammatory medication may markedly decrease asthma development. Also, enhancing resistance to respiratory viruses has been a topic of discussion. Primary and secondary prevention strategies are being developed with the aim of decreasing the incidence of asthma. Here, we review the current knowledge on rhinovirus-induced early wheezing as a risk factor for subsequent asthma development and related asthma-prevention strategies.
Collapse
Affiliation(s)
- Hannele Mikkola
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu, Oulu, Finland
| | - Minna Honkila
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, Oulu, Finland
| | - Terhi Tapiainen
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, Oulu, Finland
| | - Tuomas Jartti
- PEDEGO Research Unit and Medical Research Center Oulu, University of Oulu, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, Oulu, Finland
- Department of Pediatrics and Adolescent Medicine, University of Turku and Turku University Hospital, Turku, Finland
| |
Collapse
|
62
|
Mahdi B. Asthma as a risk factor for The progression of COVID-19. ACTA FACULTATIS MEDICAE NAISSENSIS 2022. [DOI: 10.5937/afmnai39-33234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Background: Asthma is one of the most common chronic respiratory diseases in the world, standing for the most frequent cause for hospitalization and emergency cases. Respiratory viruses are the most triggering cause. Aim: To assess the role of viral infections, especially COVID-19, in the pathogenesis of asthma initiation and exacerbations. Method: Electronic search was done for the manuscripts focusing on asthma as a risk factor for complications after COVID-19 infection. The outcomes were titles, materials, methods and classified studies related or not related to the review study. Three hundred publications were identified and only ten studies were selected for analysis. Seven studies were review, one retrospective, one longitudinal cohort study and one letter to the editor. Results: The included literature sources have highlighted different effect of asthma on COVID-19 progression. Asthma can be triggered by this virus and asthmatic patients with COVID-19 should not stop their treatment. Others suggest that asthma does not appear to be a significant risk factor for developing severe COVID-19 requiring hospitalization or intubation. Conclusions: Asthma is considered as comorbidity factor leading to complications and mortality in subjects infected with COVID-19.
Collapse
|
63
|
Feng KN, Meng P, Zhang M, Zou XL, Li S, Huang CQ, Lai KF, Li HT, Zhang TT. IL-24 Contributes to Neutrophilic Asthma in an IL-17A-Dependent Manner and Is Suppressed by IL-37. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2022; 14:505-527. [PMID: 36174993 PMCID: PMC9523421 DOI: 10.4168/aair.2022.14.5.505] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 07/04/2022] [Accepted: 07/08/2022] [Indexed: 11/25/2022]
Abstract
Purpose Neutrophilic asthma is associated with asthma exacerbation, steroid insensitivity, and severe asthma. Interleukin (IL)-24 is overexpressed in asthma and is involved in the pathogenesis of several allergic inflammatory diseases. However, the role and specific mechanism of IL-24 in neutrophilic asthma are unclear. We aimed to elucidate the roles of IL-24 and IL-37 in neutrophilic asthma, the relationships with IL-17A and the mechanisms regulating neutrophilic asthma progression. Methods Purified human neutrophils were isolated from healthy volunteers, and a cell coculture system was used to evaluate the function of IL-24 in epithelium-derived IL-17A-dependent neutrophil migration. IL-37 or a small interfering RNA (siRNA) targeting IL-24 was delivered intranasally to verify the effect in a murine model of house dust mite (HDM)/lipopolysaccharide (LPS)-induced neutrophilic asthma. Results IL-24 enhanced IL-17A production in bronchial epithelial cells via the STAT3 and ERK1/2 signaling pathways; this effect was reversed by exogenous IL-37. Anti-IL-17A monoclonal antibodies reduced neutrophil chemotaxis induced by IL-24-treated epithelial cells in vitro. Increased IL-24 and IL-17A expression in the airway epithelium was observed in HDM/LPS-induced neutrophilic asthma. IL-37 administration or IL-24 silencing attenuated neutrophilic asthma, reducing IL-17A levels and decreasing neutrophil airway infiltration, airway hyperresponsiveness, and goblet cell metaplasia. Silencing IL-24 inhibited T-helper 17 (Th17) immune responses, but not Th1 or Th2 immune responses, in the lungs of a neutrophilic asthma model. Conclusions IL-24 aggravated neutrophilic airway inflammation by increasing epithelium-derived IL-17A production, which could be suppressed by IL-37. Targeting the IL-24/IL-17A signaling axis is a potential strategy, and IL-37 is a potential candidate agent for alleviating neutrophilic airway inflammation in asthma.
Collapse
Affiliation(s)
- Kang-ni Feng
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Disease of Sun Yat-sen University, Guangzhou, China
| | - Ping Meng
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Disease of Sun Yat-sen University, Guangzhou, China
| | - Min Zhang
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Disease of Sun Yat-sen University, Guangzhou, China
| | - Xiao-ling Zou
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Disease of Sun Yat-sen University, Guangzhou, China
| | - Shuang Li
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Disease of Sun Yat-sen University, Guangzhou, China
| | - Chu-qin Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ke-fang Lai
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hong-tao Li
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Disease of Sun Yat-sen University, Guangzhou, China
| | - Tian-tuo Zhang
- Department of Pulmonary and Critical Care Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Institute of Respiratory Disease of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
64
|
The Role of Interleukins in the Pathogenesis of Dermatological Immune-Mediated Diseases. Adv Ther 2022; 39:4474-4508. [PMID: 35997892 PMCID: PMC9395905 DOI: 10.1007/s12325-022-02241-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/22/2022] [Indexed: 01/30/2023]
Abstract
Autoimmune inflammatory diseases are primarily characterized by deregulated expression of cytokines, which drive pathogenesis of these diseases. A number of approved and experimental therapies utilize monoclonal antibodies against cytokine proteins. Cytokines can be classified into different families including the interleukins, which are secreted and act on leukocytes, the tumor necrosis factor (TNF) family, as well as chemokine proteins. In this review article, we focus on the interleukin family of cytokines, of which 39 members have been identified to this date. We outline the role of each of these interleukins in the immune system, and various dermatological inflammatory diseases with a focused discussion on the pathogenesis of psoriasis and atopic dermatitis. In addition, we describe the roles of various interleukins in psychiatric, cardiovascular, and gastrointestinal comorbidities. Finally, we review clinical efficacy and safety data from emerging late-phase anti-interleukin therapies under development for psoriasis and atopic dermatitis. Collectively, additional fundamental and clinical research remains necessary to fully elucidate the roles of various interleukin proteins in the pathogenesis of inflammatory dermatologic diseases, and treatment outcomes in patients.
Collapse
|
65
|
Carsuzaa F, Béquignon É, Dufour X, de Bonnecaze G, Lecron JC, Favot L. Cytokine Signature and Involvement in Chronic Rhinosinusitis with Nasal Polyps. Int J Mol Sci 2021; 23:ijms23010417. [PMID: 35008843 PMCID: PMC8745309 DOI: 10.3390/ijms23010417] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 12/28/2021] [Accepted: 12/29/2021] [Indexed: 02/07/2023] Open
Abstract
Cytokines are well known to play a central role in chronic rhinosinusitis with nasal polyps (CRSwNP), particularly in maintenance of the inflammatory response and the recruitment of eosinophils. The pathophysiological concepts concerning the involvement of inflammatory cytokines in CRSwNP have gradually evolved. Although the Th2 cytokines environment associated with an eosinophilic infiltration has retained a central role in the genesis of polyps, the role of other cytokine subpopulations has also and more recently been detailed, leading to a specific and complex signature in CRSwNP. The purpose of this review is to summarize the current state of knowledge about the cytokine signature in CRSwNP, the role of cytokines in the pathogenesis of this disease and in the intercellular dialog between epithelial cells, fibroblasts and inflammatory cells. Knowledge of this precise cytokine signature in CRSwNP is fundamental in the perspective of potential targeting biotherapies.
Collapse
Affiliation(s)
- Florent Carsuzaa
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines (LITEC), Université de Poitiers, 86000 Poitiers, France; (X.D.); (J.-C.L.); (L.F.)
- Oto-Rhino-Laryngologie et Chirurgie Cervico-Maxillo-Faciale, Centre Hospitalier Universitaire de Poitiers, 86021 Poitiers, France
- Correspondence: ; Tel.: +33-(0)5-49-44-43-28
| | - Émilie Béquignon
- Oto-Rhino-Laryngologie et Chirurgie Cervico-Faciale, Hôpital Henri Mondor et Centre Hospitalier Intercommunal de Créteil, 94010 Créteil, France;
- INSERM U955, Équipe 13, Centre Henri Mondor de Recherche Biomédicale, 94000 Créteil, France
| | - Xavier Dufour
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines (LITEC), Université de Poitiers, 86000 Poitiers, France; (X.D.); (J.-C.L.); (L.F.)
- Oto-Rhino-Laryngologie et Chirurgie Cervico-Maxillo-Faciale, Centre Hospitalier Universitaire de Poitiers, 86021 Poitiers, France
| | - Guillaume de Bonnecaze
- Oto-Rhino-Laryngologie et Chirurgie Cervico-Faciale, Centre Hospitalier Universitaire de Toulouse, 31400 Toulouse, France;
| | - Jean-Claude Lecron
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines (LITEC), Université de Poitiers, 86000 Poitiers, France; (X.D.); (J.-C.L.); (L.F.)
- Service Immunologie et Inflammation, Centre Hospitalier Universitaire de Poitiers, 86021 Poitiers, France
| | - Laure Favot
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines (LITEC), Université de Poitiers, 86000 Poitiers, France; (X.D.); (J.-C.L.); (L.F.)
| |
Collapse
|
66
|
Ualiyeva S, Lemire E, Aviles EC, Wong C, Boyd AA, Lai J, Liu T, Matsumoto I, Barrett NA, Boyce JA, Haber AL, Bankova LG. Tuft cell-produced cysteinyl leukotrienes and IL-25 synergistically initiate lung type 2 inflammation. Sci Immunol 2021; 6:eabj0474. [PMID: 34932383 DOI: 10.1126/sciimmunol.abj0474] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Saltanat Ualiyeva
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women's Hospital and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Evan Lemire
- Department of Environmental Health, Harvard School of Public Health, Boston, MA, USA
| | - Evelyn C Aviles
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Caitlin Wong
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women's Hospital and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Amelia A Boyd
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women's Hospital and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Juying Lai
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women's Hospital and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Tao Liu
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women's Hospital and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | | | - Nora A Barrett
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women's Hospital and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Joshua A Boyce
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women's Hospital and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Adam L Haber
- Department of Environmental Health, Harvard School of Public Health, Boston, MA, USA
| | - Lora G Bankova
- Division of Allergy and Clinical Immunology, Jeff and Penny Vinik Center for Allergic Disease Research, Brigham and Women's Hospital and Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
67
|
IL-25 Induced ROS-Mediated M2 Macrophage Polarization via AMPK-Associated Mitophagy. Int J Mol Sci 2021; 23:ijms23010003. [PMID: 35008429 PMCID: PMC8744791 DOI: 10.3390/ijms23010003] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/17/2021] [Accepted: 12/18/2021] [Indexed: 12/29/2022] Open
Abstract
Interleukin (IL)-25 is a cytokine released by airway epithelial cells responding to pathogens. Excessive production of reactive oxygen species (ROS) leads to airway inflammation and remodeling in asthma. Mitochondria are the major source of ROS. After stress, defective mitochondria often undergo selective degradation, known as mitophagy. In this study, we examined the effects of IL-25 on ROS production and mitophagy and investigated the underlying mechanisms. The human monocyte cell line was pretreated with IL-25 at different time points. ROS production was measured by flow cytometry. The involvement of mitochondrial activity in the effects of IL-25 on ROS production and subsequent mitophagy was evaluated by enzyme-linked immunosorbent assay, Western blotting, and confocal microscopy. IL-25 stimulation alone induced ROS production and was suppressed by N-acetylcysteine, vitamin C, antimycin A, and MitoTEMPO. The activity of mitochondrial complex I and complex II/III and the levels of p-AMPK and the mitophagy-related proteins were increased by IL-25 stimulation. The CCL-22 secretion was increased by IL-25 stimulation and suppressed by mitophagy inhibitor treatment and PINK1 knockdown. The Th2-like cytokine IL-25 can induce ROS production, increase mitochondrial respiratory chain complex activity, subsequently activate AMPK, and induce mitophagy to stimulate M2 macrophage polarization in monocytes.
Collapse
|
68
|
Rozario C, Martínez-Sobrido L, McSorley HJ, Chauché C. Could Interleukin-33 (IL-33) Govern the Outcome of an Equine Influenza Virus Infection? Learning from Other Species. Viruses 2021; 13:2519. [PMID: 34960788 PMCID: PMC8704309 DOI: 10.3390/v13122519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/04/2021] [Accepted: 12/13/2021] [Indexed: 11/16/2022] Open
Abstract
Influenza A viruses (IAVs) are important respiratory pathogens of horses and humans. Infected individuals develop typical respiratory disorders associated with the death of airway epithelial cells (AECs) in infected areas. Virulence and risk of secondary bacterial infections vary among IAV strains. The IAV non-structural proteins, NS1, PB1-F2, and PA-X are important virulence factors controlling AEC death and host immune responses to viral and bacterial infection. Polymorphism in these proteins impacts their function. Evidence from human and mouse studies indicates that upon IAV infection, the manner of AEC death impacts disease severity. Indeed, while apoptosis is considered anti-inflammatory, necrosis is thought to cause pulmonary damage with the release of damage-associated molecular patterns (DAMPs), such as interleukin-33 (IL-33). IL-33 is a potent inflammatory mediator released by necrotic cells, playing a crucial role in anti-viral and anti-bacterial immunity. Here, we discuss studies in human and murine models which investigate how viral determinants and host immune responses control AEC death and subsequent lung IL-33 release, impacting IAV disease severity. Confirming such data in horses and improving our understanding of early immunologic responses initiated by AEC death during IAV infection will better inform the development of novel therapeutic or vaccine strategies designed to protect life-long lung health in horses and humans, following a One Health approach.
Collapse
Affiliation(s)
- Christoforos Rozario
- Centre for Inflammation Research, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4TJ, UK;
| | | | - Henry J. McSorley
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Wellcome Trust Building, Dow Street, Dundee DD1 5EH, UK;
| | - Caroline Chauché
- Centre for Inflammation Research, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4TJ, UK;
| |
Collapse
|
69
|
Ghezzi M, Pozzi E, Abbattista L, Lonoce L, Zuccotti GV, D’Auria E. Barrier Impairment and Type 2 Inflammation in Allergic Diseases: The Pediatric Perspective. CHILDREN (BASEL, SWITZERLAND) 2021; 8:1165. [PMID: 34943362 PMCID: PMC8700706 DOI: 10.3390/children8121165] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/30/2021] [Accepted: 11/30/2021] [Indexed: 01/02/2023]
Abstract
Allergic diseases represent a global burden. Although the patho-physiological mechanisms are still poorly understood, epithelial barrier dysfunction and Th2 inflammatory response play a pivotal role. Barrier dysfunction, characterized by a loss of differentiation, reduced junctional integrity, and altered innate defence, underpins the pathogenesis of allergic diseases. Epithelial barrier impairment may be a potential therapeutic target for new treatment strategies Up now, monoclonal antibodies and new molecules targeting specific pathways of the immune response have been developed, and others are under investigation, both for adult and paediatric populations, which are affected by atopic dermatitis (AD), asthma, allergic rhinitis (AR), chronic rhinosinusitis with nasal polyps (CRSwNP), or eosinophilic esophagitis (EoE). In children affected by severe asthma biologics targeting IgE, IL-5 and against IL-4 and IL-13 receptors are already available, and they have also been applied in CRSwNP. In severe AD Dupilumab, a biologic which inhibits both IL-4 and IL-13, the most important cytokines involved in inflammation response, has been approved for treatment of patients over 12 years. While a biological approach has already shown great efficacy on the treatment of severe atopic conditions, early intervention to restore epithelial barrier integrity, and function may prevent the inflammatory response and the development of the atopic march.
Collapse
Affiliation(s)
- Michele Ghezzi
- Allergology and Pneumology Unit, V. Buzzi Children’s Hospital, 20154 Milan, Italy;
| | - Elena Pozzi
- Department of Pediatrics, V. Buzzi Children’s Hospital, 20154 Milan, Italy; (E.P.); (L.A.); (L.L.); (G.V.Z.)
| | - Luisa Abbattista
- Department of Pediatrics, V. Buzzi Children’s Hospital, 20154 Milan, Italy; (E.P.); (L.A.); (L.L.); (G.V.Z.)
| | - Luisa Lonoce
- Department of Pediatrics, V. Buzzi Children’s Hospital, 20154 Milan, Italy; (E.P.); (L.A.); (L.L.); (G.V.Z.)
| | - Gian Vincenzo Zuccotti
- Department of Pediatrics, V. Buzzi Children’s Hospital, 20154 Milan, Italy; (E.P.); (L.A.); (L.L.); (G.V.Z.)
- Department of Biomedical and Clinical Science “L. Sacco”, University of Milan, 20157 Milan, Italy
| | - Enza D’Auria
- Allergology and Pneumology Unit, V. Buzzi Children’s Hospital, 20154 Milan, Italy;
| |
Collapse
|
70
|
Bhaker S, Portelli MA, Rakkar K, Shaw D, Johnson S, Brightling C, Sayers I. Human bronchial epithelial cells from patients with asthma have an altered gene expression profile. ERJ Open Res 2021; 8:00625-2021. [PMID: 35198626 PMCID: PMC8859501 DOI: 10.1183/23120541.00625-2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 11/10/2021] [Indexed: 11/22/2022] Open
Abstract
Asthma is a multifactorial disease presenting with wheeze and shortness of breath, and is known to be exacerbated by triggers such as pollen, house dust mites and viral infection. In the lung, the bronchial epithelium is recognised as a central driver of airway structural changes, including epithelial goblet cell hyperplasia and metaplasia, which are features of asthma. Bronchial epithelial cells (BECs) isolated from patients with asthma and cultured in vitro have altered barrier properties [1], elevated expression of remodelling factors [2] and defective repair [3]. Interestingly, genome-wide association studies (GWAS) of asthma have implicated a number of genes that are known to be expressed and functional in the airway epithelium, including IL33, IL1RL1, TSLP and MUC5AC [4]. To identify the molecular mechanisms underlying altered BECs phenotype in asthma patients, several studies have completed transcriptomic analyses using bronchial brush samples. Two recent meta-analyses [5, 6] suggested that alterations in chemical stimulus, extracellular region, pathways in cancer and arachidonic acid metabolism were features of the bronchial epithelium in the lungs of patients with asthma, and included 78 up- and 75 down-regulated genes [5]. While useful, a key question is how much the airway environment of a patient is driving this differential gene expression profile (GEP) and how much is intrinsic to the BECs themselves? To answer this question, we completed transcriptomic analyses of BECs cultured two-dimensionally through multiple passages in the laboratory that had originally been isolated from control subjects without disease or patients with asthma. An attrition rate (for successful culture) of 54% and 42% was observed in the asthma and control populations respectively. Gene changes observed in asthma bronchial epithelial cells are maintained following repeated culture, presenting with an exaggerated response to viral infection and immune responses as well as having differences in the rate of cell division and replicationhttps://bit.ly/3Cq2xKf
Collapse
|
71
|
Huang M, Wu J, Dong J. Modified BuShenYiQi formula alleviates experimental allergic asthma in mice by negative regulation of type 2 innate lymphoid cells and CD4 + type 9 helper T cells and the VIP-VPAC2 signalling pathway. PHARMACEUTICAL BIOLOGY 2021; 59:1216-1232. [PMID: 34493162 PMCID: PMC8425750 DOI: 10.1080/13880209.2021.1970198] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 06/23/2021] [Accepted: 08/14/2021] [Indexed: 06/01/2023]
Abstract
CONTEXT Modified BuShenYiQi formula (M-BYF) is derived from BuShenYiQi formula, used for the treatment of allergic asthma. The exact effect and mechanism of M-BYF on the improvement of asthma remain unclear. OBJECTIVE We investigated the mechanism underlying the therapeutic effect of M-BYF on allergic asthma. MATERIALS AND METHODS The asthma model was established in female BALB/c mice that were sensitized and challenged with ovalbumin (OVA). Mice in the treated groups were orally treated once a day with M-BYF (7, 14 and 28 g/kg/d) or dexamethasone before OVA challenge. Control and Model group received saline. Pathophysiological abnormalities and percentages of lung type 2 innate lymphoid cells (ILC2s) and Th9 cells were measured. Expression levels of type 2 cytokines and transcription factors required for these cells function and differentiation were analysed. Expression of vasoactive intestinal polypeptide (VIP)-VPAC2 signalling pathway-related proteins, and percentages of VIP expressing (VIP+) cells and VPAC2, CD90 co-expressing (VPAC2+CD90+) cells were detected. RESULTS M-BYF alleviated airway hyperresponsiveness, inflammation, mucus hypersecretion and collagen deposition in asthmatic mice. M-BYF down-regulated percentages of ILC2s and Th9 cells with lower expression of GATA3, PU.1 and IRF4, reduced IL-5, IL-13, IL-9 and VIP production. The decrease in the expression of VIP-VPAC2 signalling pathway and percentages of VIP+ cells, VPAC2+CD90+ cells were observed after M-BYF treatment. The LD50 value of M-BYF was higher than 90 g/kg. DISCUSSION AND CONCLUSIONS M-BYF alleviated experimental asthma by negatively regulating ILC2s and Th9 cells and the VIP-VPAC2 signalling pathway. These findings provide the theoretical basis for future research of M-BYF in asthma patient population.
Collapse
Affiliation(s)
- Muhua Huang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinfeng Wu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
72
|
Thymic stromal lymphopoietin and alarmins as possible therapeutical targets for asthma. Curr Opin Allergy Clin Immunol 2021; 21:590-596. [PMID: 34608100 PMCID: PMC9722372 DOI: 10.1097/aci.0000000000000793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE OF REVIEW Overview of epithelial cytokines, particularly thymic stromal lymphopoietin (TSLP), released by the airway epithelium and the effects of their inhibition on the outcomes of patients with asthma. RECENT FINDINGS The epithelial cytokines are early mediators at the top of the inflammatory cascade and are attractive therapeutic targets to prevent exacerbations and improve lung function in patients with type 2 and nontype 2 asthma. SUMMARY Clinical trials demonstrated that tezepelumab, an anti-TSLP monoclonal antibody, is a promising alternative treatment for asthma that is effective also in nontype 2 asthma. The PATHWAY and NAVIGATOR trials have assessed its effects in improving outcomes on broad clinically diverse populations. The identification of biomarkers will help to predict potential responders and help in asthma treatment personalization.
Collapse
|
73
|
Allergic Diseases: A Comprehensive Review on Risk Factors, Immunological Mechanisms, Link with COVID-19, Potential Treatments, and Role of Allergen Bioinformatics. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182212105. [PMID: 34831860 PMCID: PMC8622387 DOI: 10.3390/ijerph182212105] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/02/2021] [Accepted: 11/16/2021] [Indexed: 12/24/2022]
Abstract
The prevalence of allergic diseases is regarded as one of the key challenges in health worldwide. Although the precise mechanisms underlying this rapid increase in prevalence are unknown, emerging evidence suggests that genetic and environmental factors play a significant role. The immune system, microbiota, viruses, and bacteria have all been linked to the onset of allergy disorders in recent years. Avoiding allergen exposure is the best treatment option; however, steroids, antihistamines, and other symptom-relieving drugs are also used. Allergen bioinformatics encompasses both computational tools/methods and allergen-related data resources for managing, archiving, and analyzing allergological data. This study highlights allergy-promoting mechanisms, algorithms, and concepts in allergen bioinformatics, as well as major areas for future research in the field of allergology.
Collapse
|
74
|
Ghiciuc CM, Vicovan AG, Stafie CS, Antoniu SA, Postolache P. Marine-Derived Compounds for the Potential Treatment of Glucocorticoid Resistance in Severe Asthma. Mar Drugs 2021; 19:586. [PMID: 34822457 PMCID: PMC8620935 DOI: 10.3390/md19110586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 02/07/2023] Open
Abstract
One of the challenges to the management of severe asthma is the poor therapeutic response to treatment with glucocorticosteroids. Compounds derived from marine sources have received increasing interest in recent years due to their prominent biologically active properties for biomedical applications, as well as their sustainability and safety for drug development. Based on the pathobiological features associated with glucocorticoid resistance in severe asthma, many studies have already described many glucocorticoid resistance mechanisms as potential therapeutic targets. On the other hand, in the last decade, many studies described the potentially anti-inflammatory effects of marine-derived biologically active compounds. Analyzing the underlying anti-inflammatory mechanisms of action for these marine-derived biologically active compounds, we observed some of the targeted pathogenic molecular mechanisms similar to those described in glucocorticoid (GC) resistant asthma. This article gathers the marine-derived compounds targeting pathogenic molecular mechanism involved in GC resistant asthma and provides a basis for the development of effective marine-derived drugs.
Collapse
Affiliation(s)
- Cristina Mihaela Ghiciuc
- Department of Morpho-Functional Sciences II—Pharmacology and Clinical Pharmacology, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iași, 16 Universitatii Street, 700115 Iasi, Romania
| | - Andrei Gheorghe Vicovan
- Department of Morpho-Functional Sciences II—Pharmacology and Clinical Pharmacology, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iași, 16 Universitatii Street, 700115 Iasi, Romania
| | - Celina Silvia Stafie
- Department of Preventive Medicine and Interdisciplinarity—Family Medicine Discipline, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 16 Universitatii Street, 700115 Iasi, Romania;
| | - Sabina Antonela Antoniu
- Department of Medicine II—Palliative Care Nursing, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 16 Universitatii Street, 700115 Iasi, Romania;
| | - Paraschiva Postolache
- Department of Medicine I—Pulmonary Rehabilitation Clinic, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 16 Universitatii Street, 700115 Iasi, Romania;
| |
Collapse
|
75
|
Dhariwal J, Cameron A, Wong E, Paulsen M, Trujillo-Torralbo B, Del Rosario A, Bakhsoliani E, Kebadze T, Almond M, Farne H, Gogsadze L, Aniscenko J, Rana B, Hansel TT, Jackson DJ, Kon OM, Edwards MR, Solari R, Cousins D, Walton RP, Johnston SL. Pulmonary Innate Lymphoid Cell Responses during Rhinovirus-induced Asthma Exacerbations In Vivo: A Clinical Trial. Am J Respir Crit Care Med 2021; 204:1259-1273. [PMID: 34469272 DOI: 10.1164/rccm.202010-3754oc] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rationale Type 2 innate lymphoid cells (ILC2s) are significant sources of type 2 cytokines, which are implicated in the pathogenesis of asthma and asthma exacerbations. The role of ILC2s in virus-induced asthma exacerbations is not well-characterized. Objectives To characterize pulmonary ILC responses following experimental rhinovirus challenge in patients with moderate asthma and healthy subjects. Methods Patients with moderate asthma and healthy subjects were inoculated with rhinovirus-16, and underwent bronchoscopy at baseline, day 3 and day 8 post-inoculation. Pulmonary ILC1s and ILC2s were quantified in bronchoalveolar lavage (BAL) using flow cytometry. The ratio of BAL ILC2:ILC1 was assessed to determine their relative contributions to the clinical and immune response to rhinovirus challenge. Measurements and Main Results At baseline, ILC2s were significantly higher in patients with asthma than healthy subjects. At day 8, ILC2s significantly increased from baseline in both groups, which was significantly higher in asthma than in healthy subjects (all comparisons P<0.05). In healthy subjects, ILC1s increased from baseline at day 3 (P=0.001), while in patients with asthma, ILC1s increased from baseline at day 8 (P=0.042). Patients with asthma had significantly higher ILC2:ILC1 ratios at baseline (P=0.024) and day 8 (P=0.005). Increased ILC2:ILC1 ratio in asthma correlated with clinical exacerbation severity and type 2 cytokines in nasal mucosal lining fluid. Conclusions An ILC2-predominant inflammatory profile in asthma was associated with increased severity and duration of rhinovirus infection compared with healthy subjects, supporting the potential role of ILC2s in the pathogenesis of virus-induced asthma exacerbations. Clinical trial registration available at www.clinicaltrials.gov, ID: NCT01773590.
Collapse
Affiliation(s)
- Jaideep Dhariwal
- Guy's and St Thomas' Hospitals NHS Trust, 8945, London, United Kingdom of Great Britain and Northern Ireland;
| | - Aoife Cameron
- Imperial College London, NHLI, London, United Kingdom of Great Britain and Northern Ireland
| | - Ernie Wong
- Imperial College London, NHLI, London, United Kingdom of Great Britain and Northern Ireland
| | - Malte Paulsen
- St Mary's Flow Cytometry Core Facility, London, United Kingdom of Great Britain and Northern Ireland
| | - Belen Trujillo-Torralbo
- National Heart and Lung Institute, Respiratory Science, London, United Kingdom of Great Britain and Northern Ireland
| | - Ajerico Del Rosario
- Imperial College London, 4615, NHLI, London, United Kingdom of Great Britain and Northern Ireland
| | - Eteri Bakhsoliani
- National Heart and Lung Institute, Respiratory Science, London, United Kingdom of Great Britain and Northern Ireland
| | - Tatiana Kebadze
- Imperial College London, 4615, NHLI, London, United Kingdom of Great Britain and Northern Ireland
| | - Mark Almond
- Imperial College London, 4615, NHLI, London, United Kingdom of Great Britain and Northern Ireland
| | - Hugo Farne
- Imperial College, London, Airway Disease Infection Section, National Heart and Lung Institute, London, United Kingdom of Great Britain and Northern Ireland.,MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom of Great Britain and Northern Ireland
| | - Leila Gogsadze
- National Heart and Lung Institute, Respiratory Science, London, United Kingdom of Great Britain and Northern Ireland
| | - Julia Aniscenko
- Imperial College London, 4615, Airway Disease Infection, National Heart and Lung Institute, London, United Kingdom of Great Britain and Northern Ireland
| | - Batika Rana
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom of Great Britain and Northern Ireland
| | - Trevor T Hansel
- Imperial College London, 4615, ICRRU(Research Unit), London, United Kingdom of Great Britain and Northern Ireland
| | - David J Jackson
- Guy's and St Thomas' NHS Foundation Trust, 8945, National Heart and Lung Institute, London, United Kingdom of Great Britain and Northern Ireland
| | - Onn Min Kon
- Imperial College Healthcare NHS Trust, 8946, London, United Kingdom of Great Britain and Northern Ireland
| | - Michael R Edwards
- Imperial College London, Airway Disease Infection, London, United Kingdom of Great Britain and Northern Ireland
| | - Roberto Solari
- National Heart and Lung Institute, Respiratory Science, London, United Kingdom of Great Britain and Northern Ireland
| | - David Cousins
- University of Leicester, Department of Infection, Immunity and Inflammation, Leicester, United Kingdom of Great Britain and Northern Ireland
| | - Ross P Walton
- Imperial College, National Heart and Lung Institute, London, United Kingdom of Great Britain and Northern Ireland
| | - Sebastian L Johnston
- National Heart & Lung and Wright Felming Institute of Infection & Immunity, Respiratory Medicine, London, United Kingdom of Great Britain and Northern Ireland
| | | |
Collapse
|
76
|
Pelaia C, Pelaia G, Longhini F, Crimi C, Calabrese C, Gallelli L, Sciacqua A, Vatrella A. Monoclonal Antibodies Targeting Alarmins: A New Perspective for Biological Therapies of Severe Asthma. Biomedicines 2021; 9:biomedicines9091108. [PMID: 34572294 PMCID: PMC8465735 DOI: 10.3390/biomedicines9091108] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/10/2021] [Accepted: 08/26/2021] [Indexed: 12/16/2022] Open
Abstract
Alarmins are innate cytokines, including thymic stromal lymphopoietin (TSLP), interleukin-33 (IL-33), and interleukin-25 (IL-25), which are mainly produced by airway epithelium and exert a prominent role in asthma pathobiology. In particular, several environmental factors such as allergens, cigarette smoking, airborne pollutants, and infectious agents trigger the release of alarmins, which in turn act as upstream activators of pro-inflammatory pathways underlying type 2 (T2-high) asthma. Indeed, alarmins directly activate group 2 innate lymphoid cells (ILC2), eosinophils, basophils, and mast cells and also stimulate dendritic cells to drive the commitment of naïve T helper (Th) cells towards the Th2 immunophenotype. Therefore, TSLP, IL-33, and IL-25 represent suitable targets for add-on therapies of severe asthma. Within this context, the fully human anti-TSLP monoclonal antibody tezepelumab has been evaluated in very promising randomized clinical trials. Tezepelumab and other anti-alarmins are thus likely to become, in the near future, valuable therapeutic options for the biological treatment of uncontrolled severe asthma.
Collapse
Affiliation(s)
- Corrado Pelaia
- Department of Health Sciences, University “Magna Graecia” of Catanzaro, Viale Europa-Località Germaneto, 88100 Catanzaro, Italy;
- Correspondence: ; Tel.: +39-0961-3647007; Fax: +39-0961-3647193
| | - Giulia Pelaia
- Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (G.P.); (F.L.); (A.S.)
| | - Federico Longhini
- Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (G.P.); (F.L.); (A.S.)
| | - Claudia Crimi
- Department of Clinical and Experimental Medicine, University of Catania, 95131 Catania, Italy;
| | - Cecilia Calabrese
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy;
| | - Luca Gallelli
- Department of Health Sciences, University “Magna Graecia” of Catanzaro, Viale Europa-Località Germaneto, 88100 Catanzaro, Italy;
| | - Angela Sciacqua
- Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (G.P.); (F.L.); (A.S.)
| | - Alessandro Vatrella
- Department of Medicine, Surgery, and Dentistry, University of Salerno, 84084 Salerno, Italy;
| |
Collapse
|
77
|
Komlósi ZI, van de Veen W, Kovács N, Szűcs G, Sokolowska M, O'Mahony L, Akdis M, Akdis CA. Cellular and molecular mechanisms of allergic asthma. Mol Aspects Med 2021; 85:100995. [PMID: 34364680 DOI: 10.1016/j.mam.2021.100995] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 12/21/2022]
Abstract
Asthma is a chronic disease of the airways, which affects more than 350 million people worldwide. It is the most common chronic disease in children, affecting at least 30 million children and young adults in Europe. Asthma is a complex, partially heritable disease with a marked heterogeneity. Its development is influenced both by genetic and environmental factors. The most common, as well as the most well characterized subtype of asthma is allergic eosinophilic asthma, which is characterized by a type 2 airway inflammation. The prevalence of asthma has substantially increased in industrialized countries during the last 60 years. The mechanisms underpinning this phenomenon are incompletely understood, however increased exposure to various environmental pollutants probably plays a role. Disease inception is thought to be enabled by a disadvantageous shift in the balance between protective and harmful lifestyle and environmental factors, including exposure to protective commensal microbes versus infection with pathogens, collectively leading to airway epithelial cell damage and disrupted barrier integrity. Epithelial cell-derived cytokines are one of the main drivers of the type 2 immune response against innocuous allergens, ultimately leading to infiltration of lung tissue with type 2 T helper (TH2) cells, type 2 innate lymphoid cells (ILC2s), M2 macrophages and eosinophils. This review outlines the mechanisms responsible for the orchestration of type 2 inflammation and summarizes the novel findings, including but not limited to dysregulated epithelial barrier integrity, alarmin release and innate lymphoid cell stimulation.
Collapse
Affiliation(s)
- Zsolt I Komlósi
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad Sqr. 4, 1089, Budapest, Hungary.
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), Hermann-Burchard Strasse 9, CH7265, Davos Wolfgand, Switzerland; Christine Kühne - Center for Allergy Research and Education, Davos, Switzerland
| | - Nóra Kovács
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad Sqr. 4, 1089, Budapest, Hungary; Lung Health Hospital, Munkácsy Mihály Str. 70, 2045, Törökbálint, Hungary
| | - Gergő Szűcs
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Nagyvárad Sqr. 4, 1089, Budapest, Hungary; Department of Pulmonology, Semmelweis University, Tömő Str. 25-29, 1083, Budapest, Hungary
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), Hermann-Burchard Strasse 9, CH7265, Davos Wolfgand, Switzerland; Christine Kühne - Center for Allergy Research and Education, Davos, Switzerland
| | - Liam O'Mahony
- Department of Medicine and School of Microbiology, APC Microbiome Ireland, University College Cork, Ireland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), Hermann-Burchard Strasse 9, CH7265, Davos Wolfgand, Switzerland; Christine Kühne - Center for Allergy Research and Education, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), Hermann-Burchard Strasse 9, CH7265, Davos Wolfgand, Switzerland; Christine Kühne - Center for Allergy Research and Education, Davos, Switzerland
| |
Collapse
|
78
|
Prevention and Treatment of Asthma Exacerbations in Adults. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 9:2578-2586. [PMID: 34246434 DOI: 10.1016/j.jaip.2021.05.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 02/08/2023]
Abstract
Asthma exacerbations are major contributors to disease morbidity in patients of all ages. To develop strategies that reduce the disease burden from exacerbations, it is helpful to review current concepts about the risk factors for asthma attacks and current approaches for prevention and treatment. Multiple factors contribute as risks and to the development of asthma exacerbations, including allergic and infectious processes. Viral respiratory infections, primarily from rhinoviruses, are the dominant exacerbating cause for most asthma patients. Allergic sensitization and allergen exposure contribute directly and enhance susceptibility for respiratory viral infections. Respiratory viruses infect airway epithelium to promote underlying type 2 inflammation with eosinophils, the predominant cellular component of increased inflammation. Deficiencies of antiviral interferon responses and generation have been identified that increase susceptibility to viral infections in asthma. Exacerbation treatment focuses on reducing airflow obstruction and suppressing inflammation, followed by improving long-term asthma control. Increasing concern exists regarding the side effects associated with frequent systemic corticosteroid use. A major advance has been the selective use of biologics to prevent exacerbations, primarily in patients with existing type 2 inflammation. Future research to prevent exacerbations is being directed toward antiviral activity and a more encompassing regulation of underlying airway inflammation.
Collapse
|
79
|
Murphy RC, Pavord ID, Alam R, Altman MC. Management Strategies to Reduce Exacerbations in non-T2 Asthma. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 9:2588-2597. [PMID: 34246435 DOI: 10.1016/j.jaip.2021.04.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 11/25/2022]
Abstract
There have been considerable advances in our understanding of asthmatic airway inflammation, resulting in a paradigm shift of classifying individuals on the basis of either the presence or the absence of type 2 (T2) inflammatory markers. Several novel monoclonal antibody therapies targeting T2 cytokines have demonstrated significant clinical effects including reductions in acute exacerbations and improvements in asthma-related quality of life and lung function for individuals with T2-high asthma. However, there have been fewer advancements in developing therapies for those without evidence of T2 airway inflammation (so-called non-T2 asthma). Here, we review the heterogeneity of molecular mechanisms responsible for initiation and regulation of non-T2 inflammation and discuss both current and potential future therapeutic options for individuals with non-T2 asthma.
Collapse
Affiliation(s)
- Ryan C Murphy
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Washington, Seattle, Wash; Center for Lung Biology, Department of Medicine, University of Washington, Seattle, Wash.
| | - Ian D Pavord
- Respiratory Medicine Unit and Oxford Respiratory NIHR Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Rafeul Alam
- Division of Allergy and Immunology, Department of Medicine, National Jewish Health and University of Colorado, Denver, Colo
| | - Matthew C Altman
- Center for Lung Biology, Department of Medicine, University of Washington, Seattle, Wash; Division of Allergy and Immunology, University of Washington, Seattle, Wash
| |
Collapse
|
80
|
Hong H, Tan KS, Yan Y, Chen F, Ong HH, Oo Y, Liu J, Ong YK, Thong M, Sugrue R, Chow VT, Wang DY. Induction of IL-25 Expression in Human Nasal Polyp Epithelium by Influenza Virus Infection is Abated by Interferon-Alpha Pretreatment. J Inflamm Res 2021; 14:2769-2780. [PMID: 34234504 PMCID: PMC8254189 DOI: 10.2147/jir.s304320] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 06/08/2021] [Indexed: 01/23/2023] Open
Abstract
Background Epithelial cytokines including IL-25, IL-33 and thymic stromal lymphopoietin (TLSP) are recently established as drivers of type 2 chronic inflammatory diseases such as chronic rhinosinusitis with nasal polyps (CRSwNP). Here, we further confirmed the increased expression of IL-25 in CRSwNP and investigated potential contributors of IL-25 in CRSwNP epithelium. Methods Sixty CRSwNP, 25 CRSsNP and 15 healthy control tissues were examined for IL-25 expression and for the accompanying type 2 inflammatory cytokines. We then tested different respiratory virus infections on human nasal epithelial cells (hNECs) for their ability to trigger IL-25 expression. In addition, we subjected hNECs generated from CRSwNP tissues to pretreatment with recombinant interferon-alpha (IFN-α) prior to viral infection to evaluate IFN effects on IL-25 induction. Results We confirmed that significantly enhanced levels of IL-25 were observed in CRSwNP tissues, and that IL-25 expression correlated with type 2 inflammatory cytokine expression. In vitro, we observed significantly elevated IL-25 in hNECs infected with influenza A virus as early as 24 hours post-infection (hpi), regardless of tissue origin, and IL-25 correlated positively with viral load. While other respiratory viruses exhibited increasing trends of IL-25, these were not significant at the time-points tested. IFN-α treatment of CRSwNP epithelium was found to exert bimodal effects, ie IFN-α treatment alone induced moderate IL-25 expression, whereas IFN-α pretreatment of hNECs before influenza infection significantly diminished IL-25 induction by active influenza virus infection. Conclusion We have authenticated the observation of elevated IL-25 in CRSwNP, which is correlated with type 2 inflammatory cytokines. Notably, we identified influenza virus infection as a potential contributor of IL-25 in both control and CRSwNP epithelium during active infection. This IL-25 induction can be abated by IFN-α pretreatment which ameliorated active influenza infection. Trial Registration Chictr.org.cn ChiCTR-BON-16010179, Registered 18 December 2016, http://www.chictr.org.cn/showproj.aspx?proj=17331. The authors agree on the sharing of deidentified participant data where it pertains to request directly related to the data in this article when contacted (Haiyu Hong; honghy@mail.sysu.edu.cn).
Collapse
Affiliation(s)
- Haiyu Hong
- Allergy Center, Department of Otolaryngology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, People's Republic of China.,Otorhinolaryngology Hospital, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China.,Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore
| | - Kai Sen Tan
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore.,NUHS Infectious Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Biosafety level 3 Core Facility, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore
| | - Yan Yan
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore.,Guangdong Provincial Key Laboratory of Biomedical and Guangdong Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, People's Republic of China.,Central Laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, People's Republic of China
| | - Fenghong Chen
- Otorhinolaryngology Hospital, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hsiao Hui Ong
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore.,NUHS Infectious Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yukei Oo
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore
| | - Jing Liu
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore.,NUHS Infectious Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yew Kwang Ong
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore.,Department of Otolaryngology Head & Neck Surgery, National University Hospital, National University Health System, Singapore
| | - Mark Thong
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore.,Department of Otolaryngology Head & Neck Surgery, National University Hospital, National University Health System, Singapore
| | - Richard Sugrue
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Vincent T Chow
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore.,NUHS Infectious Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - De Yun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore.,NUHS Infectious Diseases Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
81
|
Tan Y, Zhang W, Zhu Z, Qiao N, Ling Y, Guo M, Yin T, Fang H, Xu X, Lu G, Zhang P, Yang S, Fu Z, Liang D, Xie Y, Zhang R, Jiang L, Yu S, Lu J, Jiang F, Chen J, Xiao C, Wang S, Chen S, Bian XW, Lu H, Liu F, Chen S. Integrating longitudinal clinical laboratory tests with targeted proteomic and transcriptomic analyses reveal the landscape of host responses in COVID-19. Cell Discov 2021; 7:42. [PMID: 34103487 PMCID: PMC8185699 DOI: 10.1038/s41421-021-00274-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/12/2021] [Indexed: 01/08/2023] Open
Abstract
The pathophysiology of coronavirus disease 19 (COVID-19) involves a multitude of host responses, yet how they unfold during the course of disease progression remains unclear. Here, through integrative analysis of clinical laboratory tests, targeted proteomes, and transcriptomes of 963 patients in Shanghai, we delineate the dynamics of multiple circulatory factors within the first 30 days post-illness onset and during convalescence. We show that hypercortisolemia represents one of the probable causes of acute lymphocytopenia at the onset of severe/critical conditions. Comparison of the transcriptomes of the bronchoalveolar microenvironment and peripheral blood indicates alveolar macrophages, alveolar epithelial cells, and monocytes in lungs as the potential main sources of elevated cytokines mediating systemic immune responses and organ damages. In addition, the transcriptomes of patient blood cells are characterized by distinct gene regulatory networks and alternative splicing events. Our study provides a panorama of the host responses in COVID-19, which may serve as the basis for developing further diagnostics and therapy.
Collapse
Affiliation(s)
- Yun Tan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhaoqin Zhu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Niu Qiao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yun Ling
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Mingquan Guo
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Tong Yin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hai Fang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoguang Xu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gang Lu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peipei Zhang
- Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), and Department of Pathology, The First Affritted Hospital of USTC, Hefei, Anhui, China
- Department of Pathology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuangshuang Yang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziyu Fu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongguo Liang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yinyin Xie
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruihong Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lu Jiang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuting Yu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Lu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangying Jiang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chenlu Xiao
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengyue Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuo Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Key Laboratory of the Ministry of Education, Chongqing, China.
| | - Hongzhou Lu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China.
| | - Feng Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Saijuan Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
82
|
Barron SL, Saez J, Owens RM. In Vitro Models for Studying Respiratory Host-Pathogen Interactions. Adv Biol (Weinh) 2021; 5:e2000624. [PMID: 33943040 PMCID: PMC8212094 DOI: 10.1002/adbi.202000624] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/23/2021] [Indexed: 12/22/2022]
Abstract
Respiratory diseases and lower respiratory tract infections are among the leading cause of death worldwide and, especially given the recent severe acute respiratory syndrome coronavirus-2 pandemic, are of high and prevalent socio-economic importance. In vitro models, which accurately represent the lung microenvironment, are of increasing significance given the ethical concerns around animal work and the lack of translation to human disease, as well as the lengthy time to market and the attrition rates associated with clinical trials. This review gives an overview of the biological and immunological components involved in regulating the respiratory epithelium system in health, disease, and infection. The evolution from 2D to 3D cell biology and to more advanced technological integrated models for studying respiratory host-pathogen interactions are reviewed and provide a reference point for understanding the in vitro modeling requirements. Finally, the current limitations and future perspectives for advancing this field are presented.
Collapse
Affiliation(s)
- Sarah L. Barron
- Bioassay Impurities and QualityBiopharmaceuticals DevelopmentR&DAstraZenecaCambridgeCB21 6GPUK
- Department of Chemical Engineering and BiotechnologyPhilippa Fawcett DriveCambridgeCB3 0ASUK
| | - Janire Saez
- Department of Chemical Engineering and BiotechnologyPhilippa Fawcett DriveCambridgeCB3 0ASUK
| | - Róisín M. Owens
- Department of Chemical Engineering and BiotechnologyPhilippa Fawcett DriveCambridgeCB3 0ASUK
| |
Collapse
|
83
|
Tai J, Han MS, Kwak J, Kim TH. Association Between Microbiota and Nasal Mucosal Diseases in terms of Immunity. Int J Mol Sci 2021; 22:4744. [PMID: 33947066 PMCID: PMC8124637 DOI: 10.3390/ijms22094744] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 12/23/2022] Open
Abstract
The pathogenesis of nasal inflammatory diseases is related to various factors such as anatomical structure, heredity, and environment. The nasal microbiota play a key role in coordinating immune system functions. Dysfunction of the microbiota has a significant impact on the occurrence and development of nasal inflammation. This review will introduce the positive and negative roles of microbiota involved in immunity surrounding nasal mucosal diseases such as chronic sinusitis and allergic rhinitis. In addition, we will also introduce recent developments in DNA sequencing, metabolomics, and proteomics combined with computation-based bioinformatics.
Collapse
Affiliation(s)
- Junhu Tai
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul 02841, Korea
| | - Mun Soo Han
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul 02841, Korea
| | - Jiwon Kwak
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul 02841, Korea
| | - Tae Hoon Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul 02841, Korea
| |
Collapse
|
84
|
Rodriguez-Rodriguez N, Gogoi M, McKenzie AN. Group 2 Innate Lymphoid Cells: Team Players in Regulating Asthma. Annu Rev Immunol 2021; 39:167-198. [PMID: 33534604 PMCID: PMC7614118 DOI: 10.1146/annurev-immunol-110119-091711] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Type 2 immunity helps protect the host from infection, but it also plays key roles in tissue homeostasis, metabolism, and repair. Unfortunately, inappropriate type 2 immune reactions may lead to allergy and asthma. Group 2 innate lymphoid cells (ILC2s) in the lungs respond rapidly to local environmental cues, such as the release of epithelium-derived type 2 initiator cytokines/alarmins, producing type 2 effector cytokines such as IL-4, IL-5, and IL-13 in response to tissue damage and infection. ILC2s are associated with the severity of allergic asthma, and experimental models of lung inflammation have shown how they act as playmakers, receiving signals variously from stromal and immune cells as well as the nervous system and then distributing cytokine cues to elicit type 2 immune effector functions and potentiate CD4+ T helper cell activation, both of which characterize the pathology of allergic asthma. Recent breakthroughs identifying stromal- and neuronal-derived microenvironmental cues that regulate ILC2s, along with studies recognizing the potential plasticity of ILC2s, have improved our understanding of the immunoregulation of asthma and opened new avenues for drug discovery.
Collapse
Affiliation(s)
- Noe Rodriguez-Rodriguez
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire, CB2 0QH. UK
| | - Mayuri Gogoi
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire, CB2 0QH. UK
| | - Andrew N.J. McKenzie
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire, CB2 0QH. UK,Corresponding author:
| |
Collapse
|
85
|
Rajput C, Han M, Ishikawa T, Lei J, Goldsmith AM, Jazaeri S, Stroupe CC, Bentley JK, Hershenson MB. Rhinovirus C Infection Induces Type 2 Innate Lymphoid Cell Expansion and Eosinophilic Airway Inflammation. Front Immunol 2021; 12:649520. [PMID: 33968043 PMCID: PMC8100319 DOI: 10.3389/fimmu.2021.649520] [Citation(s) in RCA: 9] [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: 01/04/2021] [Accepted: 04/07/2021] [Indexed: 12/21/2022] Open
Abstract
Rhinovirus C (RV-C) infection is associated with severe asthma exacerbations. Since type 2 inflammation is an important disease mechanism in asthma, we hypothesized that RV-C infection, in contrast to RV-A, preferentially stimulates type 2 inflammation, leading to exacerbated eosinophilic inflammation. To test this, we developed a mouse model of RV-C15 airways disease. RV-C15 was generated from the full-length cDNA clone and grown in HeLa-E8 cells expressing human CDHR3. BALB/c mice were inoculated intranasally with 5 x 106 ePFU RV-C15, RV-A1B or sham. Mice inoculated with RV-C15 showed lung viral titers of 1 x 105 TCID50 units 24 h after infection, with levels declining thereafter. IFN-α, β, γ and λ2 mRNAs peaked 24-72 hrs post-infection. Immunofluorescence verified colocalization of RV-C15, CDHR3 and acetyl-α-tubulin in mouse ciliated airway epithelial cells. Compared to RV-A1B, mice infected with RV-C15 demonstrated higher bronchoalveolar eosinophils, mRNA expression of IL-5, IL-13, IL-25, Muc5ac and Gob5/Clca, protein production of IL-5, IL-13, IL-25, IL-33 and TSLP, and expansion of type 2 innate lymphoid cells. Analogous results were found in mice treated with house dust mite before infection, including increased airway responsiveness. In contrast to Rorafl/fl littermates, RV-C-infected Rorafl/flIl7rcre mice deficient in ILC2s failed to show eosinophilic inflammation or mRNA expression of IL-13, Muc5ac and Muc5b. We conclude that, compared to RV-A1B, RV-C15 infection induces ILC2-dependent type 2 airway inflammation, providing insight into the mechanism of RV-C-induced asthma exacerbations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Marc B. Hershenson
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
86
|
Harker JA, Lloyd CM. Overlapping and distinct features of viral and allergen immunity in the human lung. Immunity 2021; 54:617-631. [PMID: 33852829 DOI: 10.1016/j.immuni.2021.03.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/28/2021] [Accepted: 03/15/2021] [Indexed: 12/30/2022]
Abstract
Immunity in the human respiratory tract is provided by a diverse range of tissue-resident cells, including specialized epithelial and macrophage populations and a network of innate and innate-like lymphocytes, such as natural killer cells, innate lymphoid cells, and invariant T cells. Lung-resident memory T and B cells contribute to this network following initial exposure to antigenic stimuli. This review explores how advances in the study of human immunology have shaped our understanding of this resident immune network and its response to two of the most commonly encountered inflammatory stimuli in the airways: viruses and allergens. It discusses the many ways in which pathogenic infection and allergic inflammation mirror each other, highlighting the key checkpoints at which they diverge and how this can result in a lifetime of allergic exacerbation versus protective anti-viral immunity.
Collapse
Affiliation(s)
- James A Harker
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Clare M Lloyd
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK.
| |
Collapse
|
87
|
Williams TC, Jackson DJ, Maltby S, Walton RP, Ching YM, Glanville N, Singanayagam A, Brewins JJ, Clarke D, Hirsman AG, Loo SL, Wei L, Beale JE, Casolari P, Caramori G, Papi A, Belvisi M, Wark PAB, Johnston SL, Edwards MR, Bartlett NW. Rhinovirus-induced CCL17 and CCL22 in Asthma Exacerbations and Differential Regulation by STAT6. Am J Respir Cell Mol Biol 2021; 64:344-356. [PMID: 33264064 PMCID: PMC7909342 DOI: 10.1165/rcmb.2020-0011oc] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 10/19/2020] [Indexed: 12/17/2022] Open
Abstract
The interplay of type-2 inflammation and antiviral immunity underpins asthma exacerbation pathogenesis. Virus infection induces type-2 inflammation-promoting chemokines CCL17 and CCL22 in asthma; however, mechanisms regulating induction are poorly understood. By using a human rhinovirus (RV) challenge model in human airway epithelial cells in vitro and mice in vivo, we assessed mechanisms regulating CCL17 and CCL22 expression. Subjects with mild to moderate asthma and healthy volunteers were experimentally infected with RV and airway CCL17 and CCL22 protein quantified. In vitro airway epithelial cell- and mouse-RV infection models were then used to define STAT6- and NF-κB-mediated regulation of CCL17 and CCL22 expression. Following RV infection, CCL17 and CCL22 expression was higher in asthma, which differentially correlated with clinical and immunological parameters. Air-liquid interface-differentiated primary epithelial cells from donors with asthma also expressed higher levels of RV-induced CCL22. RV infection boosted type-2 cytokine-induced STAT6 activation. In epithelial cells, type-2 cytokines and STAT6 activation had differential effects on chemokine expression, increasing CCL17 and suppressing CCL22, whereas NF-κB promoted expression of both chemokines. In mice, RV infection activated pulmonary STAT6, which was required for CCL17 but not CCL22 expression. STAT6-knockout mice infected with RV expressed increased levels of NF-κB-regulated chemokines, which was associated with rapid viral clearance. Therefore, RV-induced upregulation of CCL17 and CCL22 was mediated by NF-κB activation, whereas expression was differentially regulated by STAT6. Together, these findings suggest that therapeutic targeting of type-2 STAT6 activation alone will not block all inflammatory pathways during RV infection in asthma.
Collapse
Affiliation(s)
- Teresa C. Williams
- School of Biomedical Science and Pharmacy, Faculty Health and Medicine and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - David J. Jackson
- Asthma UK Centre, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
- Guy’s Severe Asthma Centre, Guy’s & St. Thomas’ National Health Service Trust, London, United Kingdom
| | - Steven Maltby
- School of Biomedical Science and Pharmacy, Faculty Health and Medicine and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - Ross P. Walton
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Yee-Mann Ching
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Nicholas Glanville
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Aran Singanayagam
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jennifer J. Brewins
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Deborah Clarke
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Respiratory, Inflammation and Autoimmunity Department, MedImmune, Cambridge, United Kingdom
| | - Aurica G. Hirsman
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Su-Ling Loo
- School of Biomedical Science and Pharmacy, Faculty Health and Medicine and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - Lan Wei
- School of Biomedical Science and Pharmacy, Faculty Health and Medicine and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
| | - Janine E. Beale
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Paolo Casolari
- Interdepartmental Study Center for Inflammatory and Smoke-Related Airway Diseases, Cardiorespiratory and Internal Medicine Section, University of Ferrara, Ferrara, Italy
| | - Gaetano Caramori
- Interdepartmental Study Center for Inflammatory and Smoke-Related Airway Diseases, Cardiorespiratory and Internal Medicine Section, University of Ferrara, Ferrara, Italy
- Dipartimento di Scienze Biomediche, Pneumologia, Odontoiatriche e delle Immagini Morfologiche e Funzionali, Università degli Studi di Messina, Messina, Italy; and
| | - Alberto Papi
- Interdepartmental Study Center for Inflammatory and Smoke-Related Airway Diseases, Cardiorespiratory and Internal Medicine Section, University of Ferrara, Ferrara, Italy
| | - Maria Belvisi
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Respiratory, Inflammation and Autoimmunity Department, MedImmune, Cambridge, United Kingdom
| | - Peter A. B. Wark
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, New Lambton Heights, New South Wales, Australia
| | | | - Michael R. Edwards
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Nathan W. Bartlett
- School of Biomedical Science and Pharmacy, Faculty Health and Medicine and Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, New South Wales, Australia
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
88
|
Pfaar O, Klimek L, Jutel M, Akdis CA, Bousquet J, Breiteneder H, Chinthrajah S, Diamant Z, Eiwegger T, Fokkens WJ, Fritsch H, Nadeau KC, O’Hehir RE, O’Mahony L, Rief W, Sampath V, Schedlowski M, Torres MJ, Traidl‐Hoffmann C, Wang DY, Zhang L, Bonini M, Brehler R, Brough HA, Chivato T, Del Giacco SR, Dramburg S, Gawlik R, Gelincik A, Hoffmann‐Sommergruber K, Hox V, Knol EF, Lauerma A, Matricardi PM, Mortz CG, Ollert M, Palomares O, Riggioni C, Schwarze J, Skypala I, Untersmayr E, Walusiak‐Skorupa J, Ansotegui IJ, Bachert C, Bedbrook A, Bosnic‐Anticevich S, Brussino L, Canonica GW, Cardona V, Carreiro‐Martins P, Cruz AA, Czarlewski W, Fonseca JA, Gotua M, Haahtela T, Ivancevich JC, Kuna P, Kvedariene V, Larenas‐Linnemann DE, Abdul Latiff AH, Mäkelä M, Morais‐Almeida M, Mullol J, Naclerio R, Ohta K, Okamoto Y, Onorato GL, Papadopoulos NG, Patella V, Regateiro FS, Samoliński B, Suppli Ulrik C, Toppila‐Salmi S, Valiulis A, Ventura M, Yorgancioglu A, Zuberbier T, Agache I. COVID-19 pandemic: Practical considerations on the organization of an allergy clinic-An EAACI/ARIA Position Paper. Allergy 2021; 76:648-676. [PMID: 32531110 PMCID: PMC7323448 DOI: 10.1111/all.14453] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) has evolved into a pandemic infectious disease transmitted by the severe acute respiratory syndrome coronavirus (SARS-CoV-2). Allergists and other healthcare providers (HCPs) in the field of allergies and associated airway diseases are on the front line, taking care of patients potentially infected with SARS-CoV-2. Hence, strategies and practices to minimize risks of infection for both HCPs and treated patients have to be developed and followed by allergy clinics. METHOD The scientific information on COVID-19 was analysed by a literature search in MEDLINE, PubMed, the National and International Guidelines from the European Academy of Allergy and Clinical Immunology (EAACI), the Cochrane Library, and the internet. RESULTS Based on the diagnostic and treatment standards developed by EAACI, on international information regarding COVID-19, on guidelines of the World Health Organization (WHO) and other international organizations, and on previous experience, a panel of experts including clinicians, psychologists, IT experts, and basic scientists along with EAACI and the "Allergic Rhinitis and its Impact on Asthma (ARIA)" initiative have developed recommendations for the optimal management of allergy clinics during the current COVID-19 pandemic. These recommendations are grouped into nine sections on different relevant aspects for the care of patients with allergies. CONCLUSIONS This international Position Paper provides recommendations on operational plans and procedures to maintain high standards in the daily clinical care of allergic patients while ensuring the necessary safety measures in the current COVID-19 pandemic.
Collapse
Affiliation(s)
- Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery Section of Rhinology and Allergy University Hospital Marburg Philipps‐Universität Marburg Marburg Germany
| | - Ludger Klimek
- Center for Rhinology and Allergology Wiesbaden Germany
| | - Marek Jutel
- Department of Clinical Immunology Wrocław Medical Universityand ALL‐MED Medical Research Institute Wrocław Poland
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Davos Switzerland
| | - Jean Bousquet
- Department of Dermatology and Allergy Comprehensive Allergy Center Charité Charité Universitaetsmedizin Berlin a Member of GA2LEN Berlin Germany
- University Hospital Montpellier Montpellier France
- MACVIA‐France Montpellier France
| | - Heimo Breiteneder
- Institute of Pathophysiology and Allergy Research Center of Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - Sharon Chinthrajah
- Sean N. Parker Center for Allergy and Asthma Research Stanford University School of Medicine Stanford CA USA
| | - Zuzana Diamant
- Department of Respiratory Medicine & Allergology Institute for Clinical Science Skane University Hospital Lund University Lund Sweden
- Department of Respiratory Medicine First Faculty of Medicine Charles University and Thomayer Hospital Prague Czech Republic
- Department of Clinical Pharmacy and Pharmacology University Medical Center Groningen University of Groningen Groningen The Netherlands
| | - Thomas Eiwegger
- Translational Medicine Program Peter Gilgan Centre for Research and Learning Hospital for Sick Children Toronto ON Canada
- Departments of Paediatrics and Immunology Division of Immunology and Allergy Food Allergy and Anaphylaxis Program The Hospital for Sick Children University of Toronto Toronto ON Canada
- Department of Immunology University of Toronto Toronto ON Canada
| | - Wytske J. Fokkens
- Department of Otorhinolaryngology Amsterdam University Medical Centres Amsterdam The Netherlands
| | - Hans‐Walter Fritsch
- Department of Information‐Technology (IT) University Hospital Marburg Marburg Germany
| | - Kari C. Nadeau
- Sean N. Parker Center for Allergy and Asthma Research Stanford University School of Medicine Stanford CA USA
| | - Robyn E. O’Hehir
- Allergy, Asthma & Clinical Immunology Alfred Health Melbourne VIC Australia
- Department of Allergy, Immunology and Respiratory Medicine Central Clinical School Monash University and Alfred Health Melbourne VIC Australia
| | - Liam O’Mahony
- Departments of Medicine and Microbiology APC Microbiome Ireland University College Cork Cork Ireland
| | - Winfried Rief
- Department of Clinical Psychology and Psychotherapy Philipps‐University of Marburg Marburg Germany
| | - Vanitha Sampath
- Sean N. Parker Center for Allergy and Asthma Research Stanford University School of Medicine Stanford CA USA
| | - Manfred Schedlowski
- Institute of Medical Psychology and Behavioral Immunobiology University Clinic Essen Essen Germany
| | - María José Torres
- Allergy Unit Malaga Regional University Hospital‐UMA‐ARADyAL Málaga Spain
| | - Claudia Traidl‐Hoffmann
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
- Outpatient Clinic for Environmental Medicine University Hospital Augsburg Germany
| | - De Yun Wang
- Department of Otolaryngology National University of Singapore Singapore Singapore
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery Beijing Tongren Hospital Beijing China
- Department of Allergy Beijing Tongren Hospital Beijing China
| | - Matteo Bonini
- Department of Cardiovascular and Thoracic Sciences Fondazione Policlinico Universitario A. Gemelli ‐ IRCCS Università Cattolica del Sacro Cuore Rome Italy
- National Heart and Lung Institute (NHLI) Imperial College London London UK
| | - Randolf Brehler
- Department of Allergology, Occupational Dermatology and Environmental Medicine University Hospital Münster Münster Germany
| | - Helen Annaruth Brough
- Children's Allergy Service Evelina Children's Hospital Guy's and St. Thomas' Hospital NHS Foundation Trust London UK
- Paediatric Allergy Group Department of Women and Children's Health School of Life Course Sciences King's College London London UK
| | - Tomás Chivato
- School of Medicine University CEU San Pablo Madrid Spain
| | - Stefano R. Del Giacco
- Department of Medical Sciences and Public Health University of Cagliari Cagliari Italy
| | - Stephanie Dramburg
- Department of Pediatric Pulmonology, Immunology and Intensive Care Medicine Charité Universitaetsmedizin Berlin Germany
| | - Radoslaw Gawlik
- Department of Internal Medicine, Allergology Clinical Immunology Medical University of Silesia Katowice Poland
| | - Aslı Gelincik
- Department of Internal Medicine Division of Immunology and Allergic Diseases Istanbul Faculty of Medicine Istanbul University Istanbul Turkey
| | - Karin Hoffmann‐Sommergruber
- Institute of Pathophysiology and Allergy Research Center of Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - Valerie Hox
- Department of Otorhinolaryngology Cliniques Universitaires Saint‐Luc Brussels Belgium
| | - Edward F. Knol
- Departments of Immunology and Dermatology/Allergology University Medical Center Utrecht Utrecht The Netherlands
| | - Antti Lauerma
- Department of Dermatology and Allergology Helsinki University Hospital Inflammation Centre University of Helsinki Helsinki Finland
| | - Paolo M. Matricardi
- Department of Pediatric Pulmonology, Immunology and Intensive Care Medicine Charité Universitaetsmedizin Berlin Germany
| | - Charlotte G. Mortz
- Department of Dermatology and Allergy Centre Odense Research Centre for Anaphylaxis (ORCA) Odense University Hospital University of Southern Denmark Odense Denmark
| | - Markus Ollert
- Department of Dermatology and Allergy Centre Odense Research Centre for Anaphylaxis (ORCA) Odense University Hospital University of Southern Denmark Odense Denmark
- Department of Infection and Immunity Luxembourg Institute of Health Esch‐sur‐Alzette Luxembourg
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology Chemistry School Complutense University of Madrid Spain
| | - Carmen Riggioni
- Pediatric Allergy and Clinical Immunology Department Hospital Sant Joan de Déu Barcelona Spain
- Institut de Recerca Sant Joan de Déu Barcelona Spain
| | - Jürgen Schwarze
- Centre for Inflammation Research and Child Life and Heath the University of Edinburgh Edinburgh UK
| | - Isabel Skypala
- National Heart and Lung Institute (NHLI) Imperial College London London UK
- Royal Brompton and Harefield NHS Foundation Trust London UK
| | - Eva Untersmayr
- Institute of Pathophysiology and Allergy Research Center of Pathophysiology, Infectiology and Immunology Medical University of Vienna Vienna Austria
| | - Jolanta Walusiak‐Skorupa
- Department of Occupational Diseases and Environmental Health Nofer Institute of Occupational Medicine Lodz Poland
| | | | - Claus Bachert
- ENT Department Upper Airways Research Laboratory Ghent University Hospital Ghent Belgium
- International Airway Research Center First Affiliated Hospital Guangzou Sun Yat‐sen University Guangzou China
- Division of ENT Diseases CLINTEC, Karolinska Institutet Stockholm Sweden
| | | | - Sinthia Bosnic‐Anticevich
- Woolcock Institute of Medical Research Woolcock Emphysema Centre University of Sydney Glebe NSW Australia
| | - Luisa Brussino
- Department of Medical Sciences Allergy and Clinical Immunology Unit University of Torino & Mauriziano Hospital Torino Italy
| | - Giorgio Walter Canonica
- Personalized Medicine Asthma & Allergy Clinic Humanitas University & Research Hospital‐IRCCS Milano Italy
| | - Victoria Cardona
- Department of Internal Medicine, Allergy Section Hospital Vall d'Hebron & ARADyAL Research Network Barcelona Spain
| | - Pedro Carreiro‐Martins
- Serviço de Imunoalergologia, Hospital de Dona Estefânia Centro Hospitalar de Lisboa Central Lisbon Portugal
- NOVA Medical School/Comprehensive Health Research Center (CHRC) Universidade Nova de Lisboa Lisbon Portugal
| | - Alvaro A. Cruz
- ProAR – Nucleo de Excelencia em Asma Federal University of Bahia Bahia Brasil
- GARD/WHO Executive Committee Bahia Brazil
| | | | - João A. Fonseca
- Faculdade de Medicina da Universidade do Porto CINTESIS Center for Research in Health Technologies and Information Systems Porto Portugal
- MEDIDA, Lda Porto Portugal
| | - Maia Gotua
- Center of Allergy and Immunology Tbilisi Georgia
- David Tvildiani Medical University in Tbilisi Tbilisi Georgia
| | - Tari Haahtela
- Skin and Allergy Hospital Helsinki University Hospital and University of Helsinki Helsinki Finland
| | | | - Piotr Kuna
- Division of Internal Medicine, Asthma and Allergy Barlicki University Hospital Medical University of Lodz Lodz Poland
| | - Violeta Kvedariene
- Department of Pathology Faculty of Medicine Institute of Biomedical Sciences Vilnius University Vilnius Lithuania
- Faculty of Medicine Institute of Clinical medicine Clinic of Chest Diseases and Allergology Vilnius University Vilnius Lithuania
| | | | | | - Mika Mäkelä
- Skin and Allergy Hospital Helsinki University Hospital and University of Helsinki Helsinki Finland
| | | | - Joaquim Mullol
- ENT Department Rhinology Unit & Smell Clinic Hospital Clinic ‐ Clinical & Experimental Respiratory Immunoallergy IDIBAPSCIBERESUniversity of Barcelona Barcelona Catalonia Spain
| | | | - Ken Ohta
- Japan Anti‐Tuberculosis Association (JATA) Fukujuji Hospital, and National Hospital Organization (NHO)Tokyo National Hospital Tokyo Japan
| | | | | | - Nikolaos G. Papadopoulos
- Division of Infection Immunity & Respiratory Medicine Royal Manchester Children's Hospital University of Manchester Manchester UK
- Allergy Department 2nd Pediatric Clinic Athens General Children's Hospital "P&A Kyriakou” University of Athens Athens Greece
| | - Vincenzo Patella
- Allergy and Clinical Immunology, Department of Internal Medicine Santa Maria della Speranza Hospital Salerno Italy
| | - Frederico S. Regateiro
- Allergy and Clinical Immunology Unit Centro Hospitalar e Universitário de Coimbra Coimbra Portugal
- Institute of Immunology Faculty of Medicine University of Coimbra Coimbra Portugal
- ICBR ‐ Coimbra Institute for Clinical and Biomedical Research CIBB Faculty of Medicine University of Coimbra Coimbra Portugal
| | - Bolesław Samoliński
- Department of Prevention of Environmental Hazards and Allergology Medical University of Warsaw Warsaw Poland
| | - Charlotte Suppli Ulrik
- Department of Respiratory Medicine Hvidovre Hospital Hvidovre Denmark
- Faculty of Health Sciences Institute of Clinical Medicine University of Copenhagen Denmark
| | - Sanna Toppila‐Salmi
- Skin and Allergy Hospital Helsinki University Hospital and University of Helsinki Helsinki Finland
| | - Arunas Valiulis
- Faculty of Medicine Institute of Clinical Medicine & Institute of Health Sciences Vilnius University Vilnius Lithuania
| | - Maria‐Teresa Ventura
- Interdisciplinary Department of Medicine Unit of Geriatric Immunoallergology University of Bari Medical School Bari Italy
| | - Arzu Yorgancioglu
- Department of Pulmonary Diseases Faculty of Medicine Celal Bayar University Manisa Turkey
| | - Torsten Zuberbier
- Department of Dermatology and Allergy Comprehensive Allergy Center Charité Charité Universitaetsmedizin Berlin a Member of GA2LEN Berlin Germany
| | - Ioana Agache
- Transylvania University Brasov Romania
- Theramed Medical Center Brasov Romania
| |
Collapse
|
89
|
Critical Roles of Balanced T Helper 9 Cells and Regulatory T Cells in Allergic Airway Inflammation and Tumor Immunity. J Immunol Res 2021; 2021:8816055. [PMID: 33748292 PMCID: PMC7943311 DOI: 10.1155/2021/8816055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/12/2021] [Accepted: 02/17/2021] [Indexed: 01/02/2023] Open
Abstract
CD4+T helper (Th) cells are important mediators of immune responses in asthma and cancer. When counteracted by different classes of pathogens, naïve CD4+T cells undergo programmed differentiation into distinct types of Th cells. Th cells orchestrate antigen-specific immune responses upon their clonal T-cell receptor (TCR) interaction with the appropriate peptide antigen presented on MHC class II molecules expressed by antigen-presenting cells (APCs). T helper 9 (Th9) cells and regulatory T (Treg) cells and their corresponding cytokines have critical roles in tumor and allergic immunity. In the context of asthma and cancer, the dynamic internal microenvironment, along with chronic inflammatory stimuli, influences development, differentiation, and function of Th9 cells and Treg cells. Furthermore, the dysregulation of the balance between Th9 cells and Treg cells might trigger aberrant immune responses, resulting in development and exacerbation of asthma and cancer. In this review, the development, differentiation, and function of Th9 cells and Treg cells, which are synergistically regulated by various factors including cytokine signals, transcriptional factors (TFs), costimulatory signals, microenvironment cues, metabolic pathways, and different signal pathways, will be discussed. In addition, we focus on the recent progress that has helped to achieve a better understanding of the roles of Th9 cells and Treg cells in allergic airway inflammation and tumor immunity. We also discuss how various factors moderate their responses in asthma and cancer. Finally, we summarize the recent findings regarding potential mechanisms for regulating the balance between Th9 and Treg cells in asthma and cancer. These advances provide opportunities for novel therapeutic strategies that are aimed at reestablishing the balance of these cells in the diseases.
Collapse
|
90
|
Cavagnero KJ, Doherty TA. Lipid-mediated innate lymphoid cell recruitment and activation in aspirin-exacerbated respiratory disease. Ann Allergy Asthma Immunol 2021; 126:135-142. [PMID: 32950684 PMCID: PMC7855910 DOI: 10.1016/j.anai.2020.09.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/02/2020] [Accepted: 09/11/2020] [Indexed: 01/17/2023]
Abstract
OBJECTIVE To synthesize investigations into the role of lipid-mediated recruitment and activation of group 2 innate lymphoid cells (ILC2s) in aspirin-exacerbated respiratory disease (AERD). DATA SOURCES A comprehensive literature review of reports pertaining to cellular mechanisms, cytokine, and lipid mediators in AERD, as well as ILC2 activation and recruitment, was performed using PubMed and Google Scholar. STUDY SELECTIONS Selections of studies were based on reports of lipid mediators in AERD, cytokine mediators in AERD, type 2 effector cells in AERD, platelets in AERD, AERD treatment, ILC2s in allergic airway disease, and ILC2 activation, inhibition, and trafficking. RESULTS The precise mechanisms of AERD pathogenesis are not well understood. Greater levels of proinflammatory lipid mediators and type 2 cytokines are found in tissues derived from patients with AERD relative to controls. After pathognomonic cyclooxygenase-1 inhibitor reactions, proinflammatory mediator concentrations (prostaglandin D2 and cysteinyl leukotrienes) are rapidly increased, as are ILC2 levels in the nasal mucosa. The ILC2s, which potently generate type 2 cytokines in response to lipid mediator stimulation, may play a key role in AERD pathogenesis. CONCLUSION Although the literature suggests that lipid-mediated ILC2 activation may occur in AERD, there is a dearth of definitive evidence. Future investigations leveraging novel next-generation single-cell sequencing approaches along with recently developed AERD murine models will better define lipid mediator-induced ILC2 trafficking in patients with AERD.
Collapse
Affiliation(s)
- Kellen J Cavagnero
- Department of Medicine, University of California, San Diego, La Jolla, California; Department of Dermatology, University of California, San Diego, La Jolla, California
| | - Taylor A Doherty
- Department of Medicine, University of California, San Diego, La Jolla, California; Veterans Affairs San Diego Health Care System, La Jolla, California.
| |
Collapse
|
91
|
Borowczyk J, Shutova M, Brembilla NC, Boehncke WH. IL-25 (IL-17E) in epithelial immunology and pathophysiology. J Allergy Clin Immunol 2021; 148:40-52. [PMID: 33485651 DOI: 10.1016/j.jaci.2020.12.628] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/08/2020] [Accepted: 12/16/2020] [Indexed: 02/07/2023]
Abstract
IL-25, also known as IL-17E, is a unique cytokine of the IL-17 family. Indeed, IL-25 exclusively was shown to strongly induce expression of the cytokines associated with type 2 immunity. Although produced by several types of immune cells, such as T cells, dendritic cells, or group 2 innate lymphoid cells, a vast amount of IL-25 derives from epithelial cells. The functions of IL-25 have been actively studied in the context of physiology and pathology of various organs including skin, airways and lungs, gastrointestinal tract, and thymus. Accumulating evidence suggests that IL-25 is a "barrier surface" cytokine whose expression depends on extrinsic environmental factors and when upregulated may lead to inflammatory disorders such as atopic dermatitis, psoriasis, or asthma. This review summarizes the progress of the recent years regarding the effects of IL-25 on the regulation of immune response and the balance between its homeostatic and pathogenic role in various epithelia. We revisit IL-25's general and tissue-specific mechanisms of action, mediated signaling pathways, and transcription factors activated in immune and resident cells. Finally, we discuss perspectives of the IL-25-based therapies for inflammatory disorders and compare them with the mainstream ones that target IL-17A.
Collapse
Affiliation(s)
- Julia Borowczyk
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Maria Shutova
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | | | - Wolf-Henning Boehncke
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland; Division of Dermatology and Venereology, University Hospitals of Geneva, Geneva, Switzerland.
| |
Collapse
|
92
|
Cristinziano L, Poto R, Criscuolo G, Ferrara AL, Galdiero MR, Modestino L, Loffredo S, de Paulis A, Marone G, Spadaro G, Varricchi G. IL-33 and Superantigenic Activation of Human Lung Mast Cells Induce the Release of Angiogenic and Lymphangiogenic Factors. Cells 2021; 10:cells10010145. [PMID: 33445787 PMCID: PMC7828291 DOI: 10.3390/cells10010145] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 02/06/2023] Open
Abstract
Human lung mast cells (HLMCs) express the high-affinity receptor FcεRI for IgE and are strategically located in different compartments of human lung, where they play a role in several inflammatory disorders and cancer. Immunoglobulin superantigens (e.g., protein A of Staphylococcus aureus and protein L of Peptostreptococcus magnus) bind to the variable regions of either the heavy (VH3) or light chain (κ) of IgE. IL-33 is a cytokine expressed by epithelial cells that exerts pleiotropic functions in the lung. The present study investigated whether immunoglobulin superantigens protein A and protein L and IL-33 caused the release of inflammatory (histamine), angiogenic (VEGF-A) and lymphangiogenic (VEGF-C) factors from HLMCs. The results show that protein A and protein L induced the rapid (30 min) release of preformed histamine from HLMCs. By contrast, IL-33 did not induce the release of histamine from lung mast cells. Prolonged incubation (12 h) of HLMCs with superantigens and IL-33 induced the release of VEGF-A and VEGF-C. Preincubation with IL-33 potentiated the superantigenic release of histamine, angiogenic and lymphangiogenic factors from HLMCs. Our results suggest that IL-33 might enhance the inflammatory, angiogenic and lymphangiogenic activities of lung mast cells in pulmonary disorders.
Collapse
Affiliation(s)
- Leonardo Cristinziano
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
| | - Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
| | - Gjada Criscuolo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Anne Lise Ferrara
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
| | - Maria Rosaria Galdiero
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
| | - Luca Modestino
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
| | - Amato de Paulis
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy; (L.C.); (R.P.); (G.C.); (A.L.F.); (M.R.G.); (L.M.); (S.L.); (A.d.P.); (G.M.); (G.S.)
- World Allergy Organization (WAO) Center of Excellence, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
- Correspondence:
| |
Collapse
|
93
|
Coronavirus disease 2019 and asthma, allergic rhinitis: molecular mechanisms and host-environmental interactions. Curr Opin Allergy Clin Immunol 2021; 21:1-7. [PMID: 33186186 DOI: 10.1097/aci.0000000000000699] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Coronavirus disease 2019 (COVID-19), a respiratory infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2 virus), is a pandemic in over 120 countries worldwide. Risk factors for severe COVID-19 include older age, ethnicity, sex, comorbidities, and living conditions. Although asthmatics and those with allergies are susceptible to more severe outcomes to viral infections, interestingly, asthma has not been reported to be a major comorbidity of COVID-19. However, there are some conflicting reports on the impact of asthma on COVID-19. The underlying immunological and molecular mechanisms may explain at least in part these observations. Furthermore, environmental factors like air pollution that have detrimental effects on asthma and respiratory illnesses also have an impact on COVID-19. RECENT FINDINGS Angiotensin-converting enzyme 2 (ACE2) is the receptor for the attachment and entry of SARS-CoV-2 into the host cells that is upregulated by Th1-mediated responses. In asthmatics, ACE2 gene expression is generally reduced and recent studies have shown a negative correlation between the levels of Th2 cytokines including IL-4, IL-5, and IL-13 in airway epithelial cells and other type 2 biomarkers with ACE2 expression. This may explain in part the potential protective role of asthma on COVID-19. Here, we review the relation of respiratory viral illnesses and asthma, the immune-molecular mechanisms of SARS-CoV-2 infection, the impact of asthma on COVID-19 and that of SARS-CoV-2 on asthma and allergic rhinitis, and the impact of environmental factors like air pollution on COVID-19. SUMMARY Expression of ACE2 in airway epithelial cells in SARS-COV-2 is influenced by inflammatory profile. Respiratory allergic diseases like asthma appear to have a protective effect against SARS-COV-2 infection. However, the clinical association between asthma and SARS-COV-2 is not fully established and the underlying immune-molecular mechanisms may explain these observations.
Collapse
|
94
|
Nawroth JC, Lucchesi C, Cheng D, Shukla A, Ngyuen J, Shroff T, Varone A, Karalis K, Lee HH, Alves S, Hamilton GA, Salmon M, Villenave R. A Microengineered Airway Lung Chip Models Key Features of Viral-induced Exacerbation of Asthma. Am J Respir Cell Mol Biol 2020; 63:591-600. [PMID: 32706623 DOI: 10.1165/rcmb.2020-0010ma] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Viral-induced exacerbation of asthma remains a major cause of hospitalization and mortality. New human-relevant models of the airways are urgently needed to understand how respiratory infections may trigger asthma attacks and to advance treatment development. Here, we describe a new human-relevant model of rhinovirus-induced asthma exacerbation that recapitulates viral infection of asthmatic airway epithelium and neutrophil transepithelial migration, and enables evaluation of immunomodulatory therapy. Specifically, a microengineered model of fully differentiated human mucociliary airway epithelium was stimulated with IL-13 to induce a T-helper cell type 2 asthmatic phenotype and infected with live human rhinovirus 16 (HRV16) to reproduce key features of viral-induced asthma exacerbation. We observed that the infection with HRV16 replicated key hallmarks of the cytopathology and inflammatory responses observed in human airways. Generation of a T-helper cell type 2 microenvironment through exogenous IL-13 stimulation induced features of asthmatic airways, including goblet cell hyperplasia, reduction of cilia beating frequency, and endothelial activation, but did not alter rhinovirus infectivity or replication. High-resolution kinetic analysis of secreted inflammatory markers revealed that IL-13 treatment altered IL-6, IFN-λ1, and CXCL10 secretion in response to HRV16. Neutrophil transepithelial migration was greatest when viral infection was combined with IL-13 treatment, whereas treatment with MK-7123, a CXCR2 antagonist, reduced neutrophil diapedesis in all conditions. In conclusion, our microengineered Airway Lung-Chip provides a novel human-relevant platform for exploring the complex mechanisms underlying viral-induced asthma exacerbation. Our data suggest that IL-13 may impair the hosts' ability to mount an appropriate and coordinated immune response to rhinovirus infection. We also show that the Airway Lung-Chip can be used to assess the efficacy of modulators of the immune response.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hyun-Hee Lee
- Merck Research Laboratories, Boston, Massachusetts
| | | | | | | | | |
Collapse
|
95
|
Andersson CK, Iwasaki J, Cook J, Robinson P, Nagakumar P, Mogren S, Fleming L, Bush A, Saglani S, Lloyd CM. Impaired airway epithelial cell wound-healing capacity is associated with airway remodelling following RSV infection in severe preschool wheeze. Allergy 2020; 75:3195-3207. [PMID: 32578219 DOI: 10.1111/all.14466] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/06/2020] [Accepted: 04/20/2020] [Indexed: 01/02/2023]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) causes exacerbations of asthma and preschool wheeze (PSW). However, the anti-viral and repair responses of the bronchial epithelium in children with severe therapy-resistant asthma (STRA) and PSW are poorly understood. METHODS Children with STRA (age 12 [6-16] years), PSW (age 2 [1-5] years) and non-asthmatic controls (age 7 [2-14] years) underwent bronchoscopy with endobronchial brushings and biopsies. Anti-viral, wound injury responses were quantified in biopsies and primary bronchial epithelial cells (PBECs) in response to RSV, poly(I:C), house dust mite (HDM) or IL-33 using RT-qPCR, Luminex and live cell imaging. Collagen deposition and tissue expression of epithelial growth factor receptor (EGFR), IL-33 and receptor ST2 were investigated in bronchial biopsies. RESULTS PBECs from STRA and PSW had increased TLR3 gene expression and increased secretion of anti-viral and pro-inflammatory cytokines (IFN-γ, IL-6 and IL-13) in response to RSV compared to controls. Exposure of PBECs to concomitant TLR3 agonist poly(I:C) and HDM resulted in a significant reduction in epithelial cell proliferation in PSW compared to controls. Wound-healing was also impaired in PSW compared to controls at baseline and following IL-33 stimulation. In addition, tissue EGFR expression was significantly reduced in PSW and correlated with collagen deposition in endobronchial biopsies. CONCLUSIONS Despite increased anti-viral responses, preschool children with severe wheeze had impaired airway epithelial proliferative responses following damage. This might be connected to the low expression of EGFR in PSW which may affect epithelial function and contribute to asthma pathogenesis.
Collapse
Affiliation(s)
- Cecilia K. Andersson
- Inflammation, Repair and Development Section National Heart and Lung InstituteImperial College London
- Respiratory Cell Biology Lund University Lund Sweden
| | - Jua Iwasaki
- Inflammation, Repair and Development Section National Heart and Lung InstituteImperial College London
| | - James Cook
- Inflammation, Repair and Development Section National Heart and Lung InstituteImperial College London
- Respiratory Paediatricsthe Royal Brompton and Harefield NHS Trust London UK
| | - Polly Robinson
- Inflammation, Repair and Development Section National Heart and Lung InstituteImperial College London
- Respiratory Paediatricsthe Royal Brompton and Harefield NHS Trust London UK
| | - Prasad Nagakumar
- Inflammation, Repair and Development Section National Heart and Lung InstituteImperial College London
- Respiratory Paediatricsthe Royal Brompton and Harefield NHS Trust London UK
| | - Sofia Mogren
- Respiratory Cell Biology Lund University Lund Sweden
| | - Louise Fleming
- Respiratory Paediatricsthe Royal Brompton and Harefield NHS Trust London UK
| | - Andrew Bush
- Respiratory Paediatricsthe Royal Brompton and Harefield NHS Trust London UK
| | - Sejal Saglani
- Inflammation, Repair and Development Section National Heart and Lung InstituteImperial College London
- Respiratory Paediatricsthe Royal Brompton and Harefield NHS Trust London UK
| | - Clare M. Lloyd
- Inflammation, Repair and Development Section National Heart and Lung InstituteImperial College London
| |
Collapse
|
96
|
Breiteneder H, Peng Y, Agache I, Diamant Z, Eiwegger T, Fokkens WJ, Traidl‐Hoffmann C, Nadeau K, O'Hehir RE, O'Mahony L, Pfaar O, Torres MJ, Wang D, Zhang L, Akdis CA. Biomarkers for diagnosis and prediction of therapy responses in allergic diseases and asthma. Allergy 2020; 75:3039-3068. [PMID: 32893900 PMCID: PMC7756301 DOI: 10.1111/all.14582] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/25/2020] [Accepted: 08/27/2020] [Indexed: 02/06/2023]
Abstract
Modern health care requires a proactive and individualized response to diseases, combining precision diagnosis and personalized treatment. Accordingly, the approach to patients with allergic diseases encompasses novel developments in the area of personalized medicine, disease phenotyping and endotyping, and the development and application of reliable biomarkers. A detailed clinical history and physical examination followed by the detection of IgE immunoreactivity against specific allergens still represents the state of the art. However, nowadays, further emphasis focuses on the optimization of diagnostic and therapeutic standards and a large number of studies have been investigating the biomarkers of allergic diseases, including asthma, atopic dermatitis, allergic rhinitis, food allergy, urticaria and anaphylaxis. Various biomarkers have been developed by omics technologies, some of which lead to a better classification of distinct phenotypes or endotypes. The introduction of biologicals to clinical practice increases the need for biomarkers for patient selection, prediction of outcomes and monitoring, to allow for an adequate choice of the duration of these costly and long‐lasting therapies. Escalating healthcare costs together with questions about the efficacy of the current management of allergic diseases require further development of a biomarker‐driven approach. Here, we review biomarkers in diagnosis and treatment of asthma, atopic dermatitis, allergic rhinitis, viral infections, chronic rhinosinusitis, food allergy, drug hypersensitivity and allergen immunotherapy with a special emphasis on specific IgE, the microbiome and the epithelial barrier. In addition, EAACI guidelines on biologicals are discussed within the perspective of biomarkers.
Collapse
Affiliation(s)
- Heimo Breiteneder
- Institute of Pathophysiology and Allergy Research Medical University of Vienna Vienna Austria
| | - Ya‐Qi Peng
- Swiss Institute of Allergy and Asthma Research (SIAF) University Zurich Davos Switzerland
- CK CARE Christine Kühne Center for Allergy Research and Education Davos Switzerland
- Otorhinolaryngology Hospital The First Affiliated Hospital Sun Yat‐Sen University Guangzhou China
| | - Ioana Agache
- Department of Allergy and Clinical Immunology Faculty of Medicine Transylvania University of Brasov Brasov Romania
| | - Zuzana Diamant
- Department of Respiratory Medicine & Allergology Institute for Clinical Science Skane University Hospital Lund University Lund Sweden
- Department of Respiratory Medicine First Faculty of Medicine Charles University and Thomayer Hospital Prague Czech Republic
- Department of Clinical Pharmacy & Pharmacology University of GroningenUniversity Medical Center Groningen Groningen Netherlands
| | - Thomas Eiwegger
- Translational Medicine Program, Research Institute Hospital for Sick Children Toronto ON Canada
- Department of Immunology University of Toronto Toronto ON Canada
- Division of Immunology and Allergy Food Allergy and Anaphylaxis Program The Hospital for Sick Children Departments of Paediatrics and Immunology University of Toronto Toronto ON Canada
| | - Wytske J. Fokkens
- Department of Otorhinolaryngology Amsterdam University Medical Centres Amsterdam The Netherlands
| | - Claudia Traidl‐Hoffmann
- CK CARE Christine Kühne Center for Allergy Research and Education Davos Switzerland
- Chair and Institute of Environmental Medicine UNIKA‐T Technical University of Munich and Helmholtz Zentrum München Augsburg Germany
- ZIEL ‐ Institute for Food & Health Technical University of Munich Freising‐Weihenstephan Germany
| | - Kari Nadeau
- Sean N. Parker Center for Allergy & Asthma Research Stanford University Stanford CA USA
| | - Robyn E. O'Hehir
- Department of Allergy, immunology and Respiratory Medicine Central Clinical School Monash University Melbourne Vic. Australia
- Allergy, Asthma and Clinical Immunology Service Alfred Health Melbourne Vic. Australia
| | - Liam O'Mahony
- Departments of Medicine and Microbiology APC Microbiome Ireland National University of Ireland Cork Ireland
| | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery Section of Rhinology and Allergy University Hospital MarburgPhilipps‐Universität Marburg Marburg Germany
| | - Maria J. Torres
- Allergy Unit Regional University Hospital of Malaga‐IBIMA‐UMA‐ARADyAL Malaga Spain
| | - De‐Yun Wang
- Department of Otolaryngology Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery and Department of Allergy Beijing TongRen Hospital Beijing China
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University Zurich Davos Switzerland
- CK CARE Christine Kühne Center for Allergy Research and Education Davos Switzerland
| |
Collapse
|
97
|
The Airway Epithelium-A Central Player in Asthma Pathogenesis. Int J Mol Sci 2020; 21:ijms21238907. [PMID: 33255348 PMCID: PMC7727704 DOI: 10.3390/ijms21238907] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/20/2020] [Accepted: 11/21/2020] [Indexed: 12/11/2022] Open
Abstract
Asthma is a chronic inflammatory airway disease characterized by variable airflow obstruction in response to a wide range of exogenous stimuli. The airway epithelium is the first line of defense and plays an important role in initiating host defense and controlling immune responses. Indeed, increasing evidence indicates a range of abnormalities in various aspects of epithelial barrier function in asthma. A central part of this impairment is a disruption of the airway epithelial layer, allowing inhaled substances to pass more easily into the submucosa where they may interact with immune cells. Furthermore, many of the identified susceptibility genes for asthma are expressed in the airway epithelium. This review focuses on the biology of the airway epithelium in health and its pathobiology in asthma. We will specifically discuss external triggers such as allergens, viruses and alarmins and the effect of type 2 inflammatory responses on airway epithelial function in asthma. We will also discuss epigenetic mechanisms responding to external stimuli on the level of transcriptional and posttranscriptional regulation of gene expression, as well the airway epithelium as a potential treatment target in asthma.
Collapse
|
98
|
Michi AN, Love ME, Proud D. Rhinovirus-Induced Modulation of Epithelial Phenotype: Role in Asthma. Viruses 2020; 12:v12111328. [PMID: 33227953 PMCID: PMC7699223 DOI: 10.3390/v12111328] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/17/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022] Open
Abstract
Human rhinoviruses have been linked both to the susceptibility of asthma development and to the triggering of acute exacerbations. Given that the human airway epithelial cell is the primary site of human rhinovirus (HRV) infection and replication, the current review focuses on how HRV-induced modulation of several aspects of epithelial cell phenotype could contribute to the development of asthma or to the induction of exacerbations. Modification of epithelial proinflammatory and antiviral responses are considered, as are alterations in an epithelial barrier function and cell phenotype. The contributions of the epithelium to airway remodeling and to the potential modulation of immune responses are also considered. The potential interactions of each type of HRV-induced epithelial phenotypic changes with allergic sensitization and allergic phenotype are also considered in the context of asthma development and of acute exacerbations.
Collapse
|
99
|
Brevi A, Cogrossi LL, Grazia G, Masciovecchio D, Impellizzieri D, Lacanfora L, Grioni M, Bellone M. Much More Than IL-17A: Cytokines of the IL-17 Family Between Microbiota and Cancer. Front Immunol 2020; 11:565470. [PMID: 33244315 PMCID: PMC7683804 DOI: 10.3389/fimmu.2020.565470] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/15/2020] [Indexed: 12/30/2022] Open
Abstract
The interleukin-(IL-)17 family of cytokines is composed of six members named IL-17A, IL-17B, IL-17C, IL-17D, IL-17E, and IL-17F. IL-17A is the prototype of this family, and it was the first to be discovered and targeted in the clinic. IL-17A is essential for modulating the interplay between commensal microbes and epithelial cells at our borders (i.e., skin and mucosae), and yet, for protecting us from microbial invaders, thus preserving mucosal and skin integrity. Interactions between the microbiota and cells producing IL-17A have also been implicated in the pathogenesis of immune mediated inflammatory diseases and cancer. While interactions between microbiota and IL-17B-to-F have only partially been investigated, they are by no means less relevant. The cellular source of IL-17B-to-F, their main targets, and their function in homeostasis and disease distinguish IL-17B-to-F from IL-17A. Here, we intentionally overlook IL-17A, and we focus instead on the role of the other cytokines of the IL-17 family in the interplay between microbiota and epithelial cells that may contribute to cancer pathogenesis and immune surveillance. We also underscore differences and similarities between IL-17A and IL-17B-to-F in the microbiota-immunity-cancer axis, and we highlight therapeutic strategies that directly or indirectly target IL-17 cytokines in diseases.
Collapse
Affiliation(s)
- Arianna Brevi
- Cellular Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, I.R.C.C.S. Ospedale San Raffaele, Milan, Italy
| | - Laura Lucia Cogrossi
- Cellular Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, I.R.C.C.S. Ospedale San Raffaele, Milan, Italy.,Department of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Giulia Grazia
- Cellular Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, I.R.C.C.S. Ospedale San Raffaele, Milan, Italy
| | - Desirée Masciovecchio
- Cellular Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, I.R.C.C.S. Ospedale San Raffaele, Milan, Italy
| | - Daniela Impellizzieri
- Cellular Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, I.R.C.C.S. Ospedale San Raffaele, Milan, Italy
| | - Lucrezia Lacanfora
- Cellular Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, I.R.C.C.S. Ospedale San Raffaele, Milan, Italy
| | - Matteo Grioni
- Cellular Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, I.R.C.C.S. Ospedale San Raffaele, Milan, Italy
| | - Matteo Bellone
- Cellular Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, I.R.C.C.S. Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
100
|
Porsbjerg CM, Sverrild A, Lloyd CM, Menzies-Gow AN, Bel EH. Anti-alarmins in asthma: targeting the airway epithelium with next-generation biologics. Eur Respir J 2020; 56:2000260. [PMID: 32586879 PMCID: PMC7676874 DOI: 10.1183/13993003.00260-2020] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 06/06/2020] [Indexed: 12/12/2022]
Abstract
Monoclonal antibody therapies have significantly improved treatment outcomes for patients with severe asthma; however, a significant disease burden remains. Available biologic treatments, including anti-immunoglobulin (Ig)E, anti-interleukin (IL)-5, anti-IL-5Rα and anti-IL-4Rα, reduce exacerbation rates in study populations by approximately 50% only. Furthermore, there are currently no effective treatments for patients with severe, type 2-low asthma. Existing biologics target immunological pathways that are downstream in the type 2 inflammatory cascade, which may explain why exacerbations are only partly abrogated. For example, type 2 airway inflammation results from several inflammatory signals in addition to IL-5. Clinically, this can be observed in how fractional exhaled nitric oxide (F eNO), which is driven by IL-13, may remain unchanged during anti-IL-5 treatment despite reduction in eosinophils, and how eosinophils may remain unchanged during anti-IL-4Rα treatment despite reduction in F eNO The broad inflammatory response involving cytokines including IL-4, IL-5 and IL-13 that ultimately results in the classic features of exacerbations (eosinophilic inflammation, mucus production and bronchospasm) is initiated by release of "alarmins" thymic stromal lymphopoietin (TSLP), IL-33 and IL-25 from the airway epithelium in response to triggers. The central, upstream role of these epithelial cytokines has identified them as strong potential therapeutic targets to prevent exacerbations and improve lung function in patients with type 2-high and type 2-low asthma. This article describes the effects of alarmins and discusses the potential role of anti-alarmins in the context of existing biologics. Clinical phenotypes of patients who may benefit from these treatments are also discussed, including how biomarkers may help identify potential responders.
Collapse
Affiliation(s)
| | - Asger Sverrild
- Dept of Respiratory Medicine, Bispebjerg Hospital, Copenhagen, Denmark
| | - Clare M Lloyd
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Elisabeth H Bel
- Dept of Respiratory Medicine, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|